51
|
Utembe W, Kamng'ona AW. Gut microbiota-mediated pesticide toxicity in humans: Methodological issues and challenges in the risk assessment of pesticides. CHEMOSPHERE 2021; 271:129817. [PMID: 33736210 DOI: 10.1016/j.chemosphere.2021.129817] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/20/2021] [Accepted: 01/25/2021] [Indexed: 06/12/2023]
Abstract
Many in vivo and in vitro studies have shown that pesticides can disrupt the functioning of gut microbiota (GM), which can lead to many diseases in humans. While the tests developed by the Organization of Economic Cooperation and Development (OECD) are expected to capture most apical effects resulting from GM disruptions, exclusion of GM in the risk assessment might mischaracterize hazards or overestimate/underestimate risks, especially when extrapolating results from one species to another species or population with a substantially different GM. On the other hand, direct assessment of GM-mediated effects may face challenges in identifying hazards, since not all GM perturbations will lead to human adverse effects. In this regard, reliable and validated biomarkers for common GM-mediated adverse effects may be very useful in the identification of GM-mediated pesticide toxicity. Nevertheless, proving causality of GM-mediated effects will need modifications of Bradford Hill criteria as well as Koch's postulates, which are more suitable for the "one-pathogen" paradigm. Furthermore, risk assessment of GM-mediated effects may require pesticide toxicokinetics along the gut, possibly through modeling, and the establishment of the involvement of GM in the mechanism of action (MOA) of the pesticide. Risk assessment of GM mediated effects also requires the standardization of experimental approaches as well as the establishment of microbial reference communities, since variations exist among GM in human populations.
Collapse
Affiliation(s)
- Wells Utembe
- Toxicology Department, National Institute for Occupational Health (a division of the National Health Laboratory Service), Johannesburg, 2000, South Africa; Department of Environmental Heath, Faculty of Health Sciences, University of Johannesburg, Johannesburg, 2000, South Africa.
| | - Arox Wadson Kamng'ona
- Department of Biomedical Sciences, College of Medicine, University Of Malawi, Blantyre, Malawi; Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| |
Collapse
|
52
|
Chiu CJ, Huang MT. Asthma in the Precision Medicine Era: Biologics and Probiotics. Int J Mol Sci 2021; 22:4528. [PMID: 33926084 PMCID: PMC8123613 DOI: 10.3390/ijms22094528] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 02/07/2023] Open
Abstract
Asthma is a major global health issue. Over 300 million people worldwide suffer from this chronic inflammatory airway disease. Typical clinical symptoms of asthma are characterized by a recurrent wheezy cough, chest tightness, and shortness of breath. The main goals of asthma management are to alleviate asthma symptoms, reduce the risk of asthma exacerbations, and minimize long-term medicinal adverse effects. However, currently available type 2 T helper cells (Th2)-directed treatments are often ineffective due to the heterogeneity of the asthma subgroups, which manifests clinically with variable and poor treatment responses. Personalized precision therapy of asthma according to individualized clinical characteristics (phenotype) and laboratory biomarkers (endotype) is the future prospect. This mini review discusses the molecular mechanisms underlying asthma pathogenesis, including the hot sought-after topic of microbiota, add-on therapies and the potential application of probiotics in the management of asthma.
Collapse
Affiliation(s)
- Chiao-Juno Chiu
- Graduate Institute of Clinical Medicine, School of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - Miao-Tzu Huang
- Graduate Institute of Clinical Medicine, School of Medicine, National Taiwan University, Taipei 100, Taiwan;
- Department of Medical Research, National Taiwan University Hospital, No. 7 Chung-Shan South Road, Taipei 100, Taiwan
- Department of Pediatrics, National Taiwan University Hospital, No. 7 Chung-Shan South Road, Taipei 100, Taiwan
| |
Collapse
|
53
|
Palkova L, Tomova A, Repiska G, Babinska K, Bokor B, Mikula I, Minarik G, Ostatnikova D, Soltys K. Evaluation of 16S rRNA primer sets for characterisation of microbiota in paediatric patients with autism spectrum disorder. Sci Rep 2021; 11:6781. [PMID: 33762692 PMCID: PMC7991656 DOI: 10.1038/s41598-021-86378-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/08/2021] [Indexed: 12/15/2022] Open
Abstract
intestinal microbiota is becoming a significant marker that reflects differences between health and disease status also in terms of gut-brain axis communication. Studies show that children with autism spectrum disorder (ASD) often have a mix of gut microbes that is distinct from the neurotypical children. Various assays are being used for microbiota investigation and were considered to be universal. However, newer studies showed that protocol for preparing DNA sequencing libraries is a key factor influencing results of microbiota investigation. The choice of DNA amplification primers seems to be the crucial for the outcome of analysis. In our study, we have tested 3 primer sets to investigate differences in outcome of sequencing analysis of microbiota in children with ASD. We found out that primers detected different portion of bacteria in samples especially at phylum level; significantly higher abundance of Bacteroides and lower Firmicutes were detected using 515f/806r compared to 27f/1492r and 27f*/1495f primers. So, the question is whether a gold standard of Firmicutes/Bacteroidetes ratio is a valuable and reliable universal marker, since two primer sets towards 16S rRNA can provide opposite information. Moreover, significantly higher relative abundance of Proteobacteria was detected using 27f/1492r. The beta diversity of sample groups differed remarkably and so the number of observed bacterial genera.
Collapse
Affiliation(s)
- L Palkova
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
- Medirex Inc., Bratislava, Slovakia
| | - A Tomova
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - G Repiska
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - K Babinska
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - B Bokor
- Comenius University Science Park, Comenius University in Bratislava, Bratislava, Slovakia
| | - I Mikula
- Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - D Ostatnikova
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - K Soltys
- Comenius University Science Park, Comenius University in Bratislava, Bratislava, Slovakia.
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia.
| |
Collapse
|
54
|
Huang YC, Wu MC, Wang YH, Wei JCC. Influence of constipation on atopic dermatitis: A nationwide population-based cohort study in Taiwan. Int J Clin Pract 2021; 75:e13691. [PMID: 32852877 DOI: 10.1111/ijcp.13691] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/21/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Atopic dermatitis (AD) is the chronic inflammatory disorder that affects both in childhood and adulthood. Mounting evidence indicates that gut dysbiosis contributes to AD via the gut-skin axis. Constipation can result in alteration of the gut microflora. The clinical impact of constipation on AD has not been researched. Therefore, we aim to assess the risk of AD in constipated patients by the population-based cohort study. METHODS We collected 85 554 constipated people and 85 554 people without constipation between 1999 and 2013 from the Taiwanese National Health Insurance Research Database. Propensity score analysis was administrated to match age, gender, comorbidities and medications at a ratio of 1:1. Multiple Cox regression analysis was utilised to evaluate the adjusted hazard ratio of AD. In addition, sensitivity tests and a stratified analysis were conducted. RESULTS The incidence of AD was 4.9 per 1000 person-years in the constipation group, which was higher than the rate of 2.1 per 1000 person-years observed in the non-constipation group. After adjustment for age, gender, comorbidities, corticosteroids, antihistamine and antibiotics, constipated people had a 2.31-fold greater risk of AD compared with those without constipation (adjusted hazard ratio [aHR]: 2.31 (95% CI 2.17-2.46). Moreover, constipated people had a higher likelihood of AD, regardless of gender, comorbidities, as well as the usage of corticosteroids, antihistamines and antibiotics. CONCLUSION Constipation is associated with a significantly risk factor of AD. Clinicians should be careful of the possibility of AD in constipated people. Further study is warranted to investigate the possible pathological mechanisms of this relationship.
Collapse
Affiliation(s)
- Yen-Chu Huang
- Division of Pediatric Gastroenterology, Children's Medical Center, Taichung Veterans General Hospital, Taichung City, Taiwan
| | - Meng-Che Wu
- Division of Pediatric Gastroenterology, Children's Medical Center, Taichung Veterans General Hospital, Taichung City, Taiwan
| | - Yu-Hsun Wang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung City, Taiwan
| | - James Cheng-Chung Wei
- Department of Rheumatology, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Division of Allergy, Immunology and Rheumatology, Chung Shan Medical University Hospital, Taichung City, Taiwan
- Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung City, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung City, Taiwan
| |
Collapse
|
55
|
Atyeo C, Alter G. The multifaceted roles of breast milk antibodies. Cell 2021; 184:1486-1499. [PMID: 33740451 DOI: 10.1016/j.cell.2021.02.031] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/07/2021] [Accepted: 02/12/2021] [Indexed: 12/20/2022]
Abstract
Neonates are born with an immature immune system and rely on the transfer of immunity from their mothers. Maternal antibodies are transferred via the placenta and breast milk. Although the role of placentally transferred immunoglobulin G (IgG) is established, less is known about the selection of antibodies transferred via breast milk and the mechanisms by which they provide protection against neonatal disease. Evidence suggests that breast milk antibodies play multifaceted roles, preventing infection and supporting the selection of commensals and tolerizing immunity during infancy. Here, we discuss emerging data related to the importance of breast milk antibodies in neonatal immunity and development.
Collapse
Affiliation(s)
- Caroline Atyeo
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; PhD Program in Virology, Division of Medical Sciences, Harvard University, Boston, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
56
|
Ronan V, Yeasin R, Claud EC. Childhood Development and the Microbiome-The Intestinal Microbiota in Maintenance of Health and Development of Disease During Childhood Development. Gastroenterology 2021; 160:495-506. [PMID: 33307032 PMCID: PMC8714606 DOI: 10.1053/j.gastro.2020.08.065] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/25/2020] [Accepted: 08/29/2020] [Indexed: 12/11/2022]
Abstract
The composition of the intestinal microbiome affects health from the prenatal period throughout childhood, and many diseases have been associated with dysbiosis. The gut microbiome is constantly changing, from birth throughout adulthood, and several variables affect its development and content. Features of the intestinal microbiota can affect development of the brain, immune system, and lungs, as well as body growth. We review the development of the gut microbiome, proponents of dysbiosis, and interactions of the microbiota with other organs. The gut microbiome should be thought of as an organ system that has important effects on childhood development. Dysbiosis has been associated with diseases in children and adults, including autism, attention deficit hyperactivity disorder, asthma, and allergies.
Collapse
Affiliation(s)
- Victoria Ronan
- Department of Pediatrics, The University of Chicago, Chicago, Illinois
| | - Rummanu Yeasin
- Department of Pediatrics, The University of Chicago, Chicago, Illinois; Windsor University School of Medicine, Cayon, St Kitts, West Indies
| | - Erika C Claud
- Department of Pediatrics, The University of Chicago, Chicago, Illinois.
| |
Collapse
|
57
|
Wark G, Samocha-Bonet D, Ghaly S, Danta M. The Role of Diet in the Pathogenesis and Management of Inflammatory Bowel Disease: A Review. Nutrients 2020; 13:nu13010135. [PMID: 33396537 PMCID: PMC7823614 DOI: 10.3390/nu13010135] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/21/2020] [Accepted: 12/29/2020] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel diseases, which include ulcerative colitis and Crohn’s disease, are chronic relapsing and remitting inflammatory diseases of the gastrointestinal tract that are increasing in prevalence and incidence globally. They are associated with significant morbidity, reduced quality of life to individual sufferers and are an increasing burden on society through direct and indirect costs. Current treatment strategies rely on immunosuppression, which, while effective, is associated with adverse events. Epidemiological evidence suggests that diet impacts the risk of developing IBD and modulates disease activity. Using diet as a therapeutic option is attractive to patients and clinicians alike due to its availability, low cost and few side effects. Diet may influence IBD risk and disease behaviour through several mechanisms. Firstly, some components of the diet influence microbiota structure and function with downstream effects on immune activity. Secondly, dietary components act to alter the structure and permeability of the mucosal barrier, and lastly dietary elements may have direct interactions with components of the immune response. This review will summarise the mechanisms of diet–microbial–immune system interaction, outline key studies examining associations between diet and IBD and evidence demonstrating the impact of diet on disease control. Finally, this review will outline current prescribed dietary therapies for active CD.
Collapse
Affiliation(s)
- Gabrielle Wark
- St Vincent’s Clinical School, UNSW, Sydney, NSW 2052, Australia; (G.W.); (D.S.-B.); (S.G.)
- Department of Gastroenterology and Hepatology, St Vincent’s Hospital, Sydney, SW 2010, Australia
| | - Dorit Samocha-Bonet
- St Vincent’s Clinical School, UNSW, Sydney, NSW 2052, Australia; (G.W.); (D.S.-B.); (S.G.)
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Simon Ghaly
- St Vincent’s Clinical School, UNSW, Sydney, NSW 2052, Australia; (G.W.); (D.S.-B.); (S.G.)
- Department of Gastroenterology and Hepatology, St Vincent’s Hospital, Sydney, SW 2010, Australia
| | - Mark Danta
- St Vincent’s Clinical School, UNSW, Sydney, NSW 2052, Australia; (G.W.); (D.S.-B.); (S.G.)
- Department of Gastroenterology and Hepatology, St Vincent’s Hospital, Sydney, SW 2010, Australia
- Correspondence:
| |
Collapse
|
58
|
Sitarik AR, Arora M, Austin C, Bielak LF, Eggers S, Johnson CC, Lynch SV, Kyun Park S, Hank Wu KH, Yong GJM, Cassidy-Bushrow AE. Fetal and early postnatal lead exposure measured in teeth associates with infant gut microbiota. ENVIRONMENT INTERNATIONAL 2020; 144:106062. [PMID: 32871381 PMCID: PMC7572588 DOI: 10.1016/j.envint.2020.106062] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/11/2020] [Accepted: 08/17/2020] [Indexed: 05/18/2023]
Abstract
BACKGROUND Lead (Pb) is an environmentally ubiquitous heavy metal associated with a wide range of adverse health effects in children. Both lead exposure and the early life microbiome- which plays a critical role in human development-have been linked to similar health outcomes, but it is unclear if the adverse effects of lead are partially driven by early life gut microbiota dysbiosis. The objective of this study was to examine the association between in utero and postnatal lead levels (measured in deciduous baby teeth) and early life bacterial and fungal gut microbiota in the first year of life. METHODS Data from the Wayne County Health, Environment, Allergy and Asthma Longitudinal Study (WHEALS) birth cohort were analyzed. Tooth lead levels during the 2nd and 3rd trimesters and postnatally (<1 year of age) were quantified using high-resolution microspatial mapping of dentin growth rings. Early life microbiota were measured in stool samples collected at approximately 1 and 6 months of age, using both 16S rRNA (bacterial) and ITS2 (fungal) sequencing. Of the 1,258 maternal-child pairs in WHEALS, 146 had data on both tooth metals and early life microbiome. RESULTS In utero tooth lead levels were significantly associated with gut fungal community composition at 1-month of age, where higher levels of 2nd trimester tooth lead was associated with lower abundances of Candida and Aspergillus and higher abundances of Malassezia and Saccharomyces; 3rd trimester lead was also associated with lower abundances of Candida. Though lead did not significantly associate with the overall structure of the infant gut bacterial community, it associated with the abundance of some specific bacterial taxa, including the increased abundance of Collinsella and Bilophila and a decreased abundance of Bacteroides taxa. CONCLUSIONS The observed associations between lead exposure and infant gut microbiota could play a role in the impact of lead on childhood development. Given the paucity of research examining these associations in humans-particularly for fungal microbiota-further investigation is needed.
Collapse
Affiliation(s)
- Alexandra R Sitarik
- Department of Public Health Sciences, Henry Ford Health System, Detroit, USA.
| | - Manish Arora
- Senator Frank R Lautenberg Environmental Health Sciences Laboratory, Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York City, USA.
| | - Christine Austin
- Senator Frank R Lautenberg Environmental Health Sciences Laboratory, Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York City, USA.
| | | | - Shoshannah Eggers
- Senator Frank R Lautenberg Environmental Health Sciences Laboratory, Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York City, USA.
| | - Christine C Johnson
- Department of Public Health Sciences, Henry Ford Health System, Detroit, USA.
| | - Susan V Lynch
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, USA.
| | - Sung Kyun Park
- Department of Epidemiology, University of Michigan, Ann Arbor, USA; Department of Environmental Health Sciences, University of Michigan, Ann Arbor, USA.
| | - Kuan-Han Hank Wu
- Department of Public Health Sciences, Henry Ford Health System, Detroit, USA.
| | - Germaine J M Yong
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, USA.
| | | |
Collapse
|
59
|
Kim JA, Kim SH, Kim IS, Yu DY, Kim GI, Moon YS, Kim SC, Lee SH, Lee SS, Yun CH, Choi IS, Cho KK. Galectin-9 Induced by Dietary Prebiotics Regulates Immunomodulation to Reduce Atopic Dermatitis Symptoms in 1-Chloro-2,4-Dinitrobenzene (DNCB)-Treated NC/Nga Mice. J Microbiol Biotechnol 2020; 30:1343-1354. [PMID: 32699202 PMCID: PMC9745654 DOI: 10.4014/jmb.2005.05017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/13/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022]
Abstract
Atopic dermatitis (AD) is a skin disorder that causes chronic itch. We investigated the inhibitory effects of a mixture of prebiotic short-chain galacto-oligosaccharides and long-chain fructooligosaccharides (scGOS/lcFOS), inulin, or β-glucan on AD development in 1-chloro-2,4- dinitrobenzene (DNCB)-treated NC/Nga mice. Mice were randomly assigned to six groups: untreated mice, AD control, positive control (DNCB-treated NC/Nga mice fed a dietary supplement of Zyrtec), and DNCB-treated NC/Nga mice fed a dietary supplement of prebiotics such as scGOS/lcFOS (T1), inulin (T2), or β-glucan (T3). The prebiotic treatment groups (T1, T2, and T3) showed suppression of AD symptoms, Th2 cell differentiation, and AD-like skin lesions induced by DNCB. In addition, prebiotic treatment also reduced the number of microorganisms such as Firmicutes, which is associated with AD symptoms, and increased the levels of Bacteroidetes and Ruminococcaceae, which are associated with alleviation of AD symptoms. Our findings demonstrate the inhibitory effects of prebiotics on AD development by improving the Th1/Th2 cytokine balance and beneficial symbiotic microorganisms in in vitro and in vivo models.
Collapse
Affiliation(s)
- Jeong A Kim
- Department of Animal Resources Technology, Gyeongnam National University of Science and Technology, Jinju 52725, Republic of Korea
| | - Sung Hak Kim
- Department of Animal Science, Chonnam National University, Gwangju 61186, Republic of Korea
| | - In Sung Kim
- Department of Animal Resources Technology, Gyeongnam National University of Science and Technology, Jinju 52725, Republic of Korea
| | - Da Yoon Yu
- Department of Animal Resources Technology, Gyeongnam National University of Science and Technology, Jinju 52725, Republic of Korea
| | - Gwang Il Kim
- Department of Animal Resources Technology, Gyeongnam National University of Science and Technology, Jinju 52725, Republic of Korea
| | - Yang Soo Moon
- Department of Animal Science & Biotechnology, Gyeongnam National University of Science and Technology, Jinju 52725, Republic of Korea
| | - Sung Chan Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Seung Ho Lee
- Department of Nano-Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Sang Suk Lee
- Department of Animal Science and Technology, Sunchon National University, Sunchon 57922, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - In Soon Choi
- Department of Life Science, Silla University, Busan 46958, Republic of Korea
| | - Kwang Keun Cho
- Department of Animal Resources Technology, Gyeongnam National University of Science and Technology, Jinju 52725, Republic of Korea
| |
Collapse
|
60
|
Chen Y, Lyu J, Xia Y, Zhu J, Tong S, Ying Y, Qu J, Li S. Effect of maternal sleep, physical activity and screen time during pregnancy on the risk of childhood respiratory allergies: a sex-specific study. Respir Res 2020; 21:230. [PMID: 32883301 PMCID: PMC7650521 DOI: 10.1186/s12931-020-01497-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/27/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Early life exposure in the uterus had a long-term effect on children's health. As the prevalence of allergies is increasing with a remarkable sex difference, very few studies have traced back to their early origins. We sought to investigate if maternal behavioral exposure, herein sleep, physical activity, and screen time during pregnancy is associated with childhood respiratory allergies. The sex difference would be examined. METHODS Six thousand two hundred thirty-six mother-child pairs from Shanghai Children Allergy Study (SCAS) were enrolled, The International Study of Asthma and Allergies in Childhood questionnaire was adopted to evaluate respiratory allergic diseases. RESULTS 14.6, 16.2, and 21.0% of children had asthma, wheeze, and allergic rhinitis, respectively. Maternal short sleep duration, lack of physical activity, and too much screen exposure during pregnancy could increase the risk of childhood respiratory allergies, however, the significance was found only in males. Moreover, a dose-response trend was clearly shown, any two of the three combined could increase the risk (OR,1.921; 95% CI,1.217-3.033), and the coexistence of all three further amplified the risk (OR,2.412; 95% CI,1.489-3.906). The findings can be verified in allergen test subgroup and each single type of respiratory allergies in most cases. CONCLUSIONS Maternal unhealthy behaviors during pregnancy could increase the risk of childhood respiratory allergies with a dose-response pattern. Males were more susceptible to the association. The identification of modifiable maternal risk behaviors lies in the emphasis of intervention in early life to face up increasing childhood allergies.
Collapse
Affiliation(s)
- Yiting Chen
- School of Public Health, Shanghai Jiao Tong University School of Medicine, 227 South Chongqing Road, Huangpu District, Shanghai, 200025, China
| | - Jiajun Lyu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, 227 South Chongqing Road, Huangpu District, Shanghai, 200025, China
| | - Yuanqing Xia
- School of Public Health, Shanghai Jiao Tong University School of Medicine, 227 South Chongqing Road, Huangpu District, Shanghai, 200025, China
| | - Jianzhen Zhu
- School of International and Public Affairs, Shanghai Jiao Tong University, Shanghai, China
| | - Shilu Tong
- School of Public Health, Shanghai Jiao Tong University School of Medicine, 227 South Chongqing Road, Huangpu District, Shanghai, 200025, China.,Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,School of Public Health and Social Work, Queensland University of Technology, Brisbane, Australia
| | - Yong Ying
- Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiajie Qu
- Shanghai Municipal Education Commission, 100 Dagu Road, Huangpu District, Shanghai, 200003, China.
| | - Shenghui Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, 227 South Chongqing Road, Huangpu District, Shanghai, 200025, China. .,MOE-Shanghai Key Laboratory of Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
61
|
Björkander S, Carvalho-Queiroz C, Hallberg J, Persson JO, Johansson MA, Nussbaum B, Jenmalm MC, Nilsson C, Sverremark-Ekström E. Childhood allergy is preceded by an absence of gut lactobacilli species and higher levels of atopy-related plasma chemokines. Clin Exp Immunol 2020; 202:288-299. [PMID: 32652542 PMCID: PMC7670166 DOI: 10.1111/cei.13494] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/21/2020] [Accepted: 06/30/2020] [Indexed: 11/14/2022] Open
Abstract
Alterations in the composition and reduced diversity of the infant microbiome are associated with allergic disease in children. Further, an altered microbiota is linked to immune dysregulation, including skewing of different T helper (Th) subsets, which is also seen in atopic individuals. The aim of this study was, therefore, to investigate the associations between gut lactobacilli and Th‐related plasma factors in allergy development during childhood. A total of 194 children with known allergy status at 1 year of age were followed to 10 years of age. We used real‐time polymerase chain reaction (PCR) to investigate the presence of three lactobacilli species (Lactobacillus casei, L. paracasei, L. rhamnosus) in infant fecal samples (collected between 1 week and 2 months of age) from a subgroup of children. Plasma chemokines and cytokines were quantified at 6 months and at 1, 2, 5 and 10 years of age with Luminex or enzyme‐linked immunosorbent assay (ELISA). Fractional exhaled nitrogen oxide (FeNO) was measured and spirometry performed at 10 years of age. The data were analysed by non‐parametric testing and a logistic regression model adjusted for parental allergy. An absence of these lactobacilli and higher levels of the chemokines BCA‐1/CXCL13, CCL17/TARC, MIP‐3α/CCL20 and MDC/CCL22 in plasma at 6 months of age preceded allergy development. The presence of lactobacilli associated with lower levels of atopy‐related chemokines during infancy, together with higher levels of interferon (IFN)‐γ and lower FeNO during later childhood. The results indicate that the presence of certain lactobacilli species in the infant gut may influence allergy‐related parameters in the peripheral immune system, and thereby contribute to allergy protection.
Collapse
Affiliation(s)
- S Björkander
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.,Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - C Carvalho-Queiroz
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - J Hallberg
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden.,Sachs' Children and Youth Hospital, Stockholm, Sweden.,Institute for Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - J-O Persson
- Department of Mathematics, Stockholm University, Stockholm, Sweden
| | - M A Johansson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - B Nussbaum
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - M C Jenmalm
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - C Nilsson
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden.,Sachs' Children and Youth Hospital, Stockholm, Sweden
| | - E Sverremark-Ekström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
62
|
The Infant Gut Microbiota and Risk of Asthma: The Effect of Maternal Nutrition during Pregnancy and Lactation. Microorganisms 2020; 8:microorganisms8081119. [PMID: 32722458 PMCID: PMC7466123 DOI: 10.3390/microorganisms8081119] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
Research has amply demonstrated that early life dysbiosis of the gut microbiota influences the propensity to develop asthma. The influence of maternal nutrition on infant gut microbiota is therefore of growing interest. However, a handful of prospective studies have examined the role of maternal dietary patterns during pregnancy in influencing the infant gut microbiota but did not assess whether this resulted in an increased risk of asthma later in life. The mechanisms involved in the process are also, thus far, poorly documented. There have also been few studies examining the effect of maternal dietary nutrient intake during lactation on the milk microbiota, the effect on the infant gut microbiota and, furthermore, the consequences for asthma development remain largely unknown. Therefore, the specific aim of this mini review is summarizing the current knowledge regarding the effect of maternal nutrition during pregnancy and lactation on the infant gut microbiota composition, and whether it has implications for asthma development.
Collapse
|
63
|
Kim MJ, Kim JY, Kang M, Won MH, Hong SH, Her Y. Reduced Fecal Calprotectin and Inflammation in a Murine Model of Atopic Dermatitis Following Probiotic Treatment. Int J Mol Sci 2020; 21:ijms21113968. [PMID: 32486523 PMCID: PMC7312066 DOI: 10.3390/ijms21113968] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022] Open
Abstract
Atopic dermatitis (AD) is one of the most common skin diseases with inflammation, chronic relapses, and intense pruritus. Its pathogenesis includes genetic susceptibility, an abnormal epidermal lipid barrier, and an increased production of IgE due to immune dysregulation. Recently, AD has been reported to be associated with intestinal inflammation and dysbiosis in human and murine models. Various probiotics are being used to control intestinal dysbiosis and inflammatory reactions. However, it is difficult to predict or determine the therapeutic effects of the probiotics, since it is rare for clinicians to use the probiotics alone to treat AD. It is also difficult to check whether the intestinal inflammation in patients with AD has improved since probiotic treatment. The aim of the present study was to determine whether mice with induced atopic dermatitis had any changes in fecal calprotectin, an indicator of intestinal inflammation, after probiotic administration. Our results showed that the fecal calprotectin levels in mice with induced dermatitis decreased significantly after the administration of probiotics. In addition, epidermal skin lesions were attenuated and inflammatory-related cytokines were downregulated after the administration of probiotics in mice with induced dermatitis. These results suggest that changes in fecal calprotectin levels could be used to assess the effectiveness of a probiotic strain as an adjuvant treatment for AD.
Collapse
Affiliation(s)
- Myoung-Ju Kim
- Department of Internal Medicine, School of Medicine, Kangon National University, Chuncheon 24341, Korea; (M.-J.K.); (J.-Y.K.)
| | - Ji-Young Kim
- Department of Internal Medicine, School of Medicine, Kangon National University, Chuncheon 24341, Korea; (M.-J.K.); (J.-Y.K.)
| | - Minje Kang
- Division of Biomedical Convergence, Department of Biomedical Sciences, Kangwon National University, Chuncheon 24341, Korea;
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Korea;
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangon National University, Chuncheon 24341, Korea; (M.-J.K.); (J.-Y.K.)
- Correspondence: (S.-H.H.); (Y.H.); Tel.: +82-33-250-7819 (S.-H.H. & Y.H.); Fax: +82-33-244-2367 (S.-H.H. & Y.H.)
| | - Young Her
- Department of Dermatology, School of Medicine, Kangwon National University, Kangwon National University Hospital, Chuncheon 24341, Korea
- Correspondence: (S.-H.H.); (Y.H.); Tel.: +82-33-250-7819 (S.-H.H. & Y.H.); Fax: +82-33-244-2367 (S.-H.H. & Y.H.)
| |
Collapse
|
64
|
Hooker BS, Miller NZ. Analysis of health outcomes in vaccinated and unvaccinated children: Developmental delays, asthma, ear infections and gastrointestinal disorders. SAGE Open Med 2020; 8:2050312120925344. [PMID: 32537156 PMCID: PMC7268563 DOI: 10.1177/2050312120925344] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 04/20/2020] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE The aim of this study was to compare the health of vaccinated versus unvaccinated pediatric populations. METHODS Using data from three medical practices in the United States with children born between November 2005 and June 2015, vaccinated children were compared to unvaccinated children during the first year of life for later incidence of developmental delays, asthma, ear infections and gastrointestinal disorders. All diagnoses utilized International Classification of Diseases-9 and International Classification of Diseases-10 codes through medical chart review. Subjects were a minimum of 3 years of age, stratified based on medical practice, year of birth and gender and compared using a logistic regression model. RESULTS Vaccination before 1 year of age was associated with increased odds of developmental delays (OR = 2.18, 95% CI 1.47-3.24), asthma (OR = 4.49, 95% CI 2.04-9.88) and ear infections (OR = 2.13, 95% CI 1.63-2.78). In a quartile analysis, subjects were grouped by number of vaccine doses received in the first year of life. Higher odds ratios were observed in Quartiles 3 and 4 (where more vaccine doses were received) for all four health conditions considered, as compared to Quartile 1. In a temporal analysis, developmental delays showed a linear increase as the age cut-offs increased from 6 to 12 to 18 to 24 months of age (ORs = 1.95, 2.18, 2.92 and 3.51, respectively). Slightly higher ORs were also observed for all four health conditions when time permitted for a diagnosis was extended from ⩾ 3 years of age to ⩾ 5 years of age. CONCLUSION In this study, which only allowed for the calculation of unadjusted observational associations, higher ORs were observed within the vaccinated versus unvaccinated group for developmental delays, asthma and ear infections. Further study is necessary to understand the full spectrum of health effects associated with childhood vaccination.
Collapse
Affiliation(s)
- Brian S Hooker
- Department of Sciences and Mathematics,
Simpson University, Redding, CA, USA
| | - Neil Z Miller
- Institute of Medical and Scientific
Inquiry, Santa Fe, NM, USA
| |
Collapse
|
65
|
Zhang D, Li S, Wang N, Tan HY, Zhang Z, Feng Y. The Cross-Talk Between Gut Microbiota and Lungs in Common Lung Diseases. Front Microbiol 2020; 11:301. [PMID: 32158441 PMCID: PMC7052046 DOI: 10.3389/fmicb.2020.00301] [Citation(s) in RCA: 231] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
Emerging findings indicate there is a vital cross-talk between gut microbiota and the lungs, which is known as gut-lung axis. The gut disturbances in lung diseases including allergy, asthma, chronic obstructive pulmonary disease, cystic fibrosis and lung cancer were observed by extensive studies. Investigating how gut microbiota impact other distant organs is of great interest in recent years. Although it has not been fully understood whether the disturbance is the cause or effect of lung diseases, alterations in the gut microbial species and metabolites have been linked to changes in immune responses and inflammation as well as the disease development in the lungs. In this article, we systemically review the role and mechanisms underlying the changes in the constituent of gut microbiota and metabolites in lung diseases. In particular, the roles of gut-lung axis in mediating immune responses and reshaping inflammation are highlighted. Furthermore, we discuss the potential of strategies to manipulate the gut microbiota and metabolites as the therapeutic approach for lung diseases.
Collapse
Affiliation(s)
- Dapeng Zhang
- First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Sha Li
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ning Wang
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hor-Yue Tan
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhimin Zhang
- First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yibin Feng
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
66
|
Apostol AC, Jensen KDC, Beaudin AE. Training the Fetal Immune System Through Maternal Inflammation-A Layered Hygiene Hypothesis. Front Immunol 2020; 11:123. [PMID: 32117273 PMCID: PMC7026678 DOI: 10.3389/fimmu.2020.00123] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/17/2020] [Indexed: 12/14/2022] Open
Abstract
Over the last century, the alarming surge in allergy and autoimmune disease has led to the hypothesis that decreasing exposure to microbes, which has accompanied industrialization and modern life in the Western world, has fundamentally altered the immune response. In its current iteration, the “hygiene hypothesis” suggests that reduced microbial exposures during early life restricts the production and differentiation of immune cells suited for immune regulation. Although it is now well-appreciated that the increase in hypersensitivity disorders represents a “perfect storm” of many contributing factors, we argue here that two important considerations have rarely been explored. First, the window of microbial exposure that impacts immune development is not limited to early childhood, but likely extends into the womb. Second, restricted microbial interactions by an expectant mother will bias the fetal immune system toward hypersensitivity. Here, we extend this discussion to hypothesize that the cell types sensing microbial exposures include fetal hematopoietic stem cells, which drive long-lasting changes to immunity.
Collapse
Affiliation(s)
- April C Apostol
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States
| | - Kirk D C Jensen
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States
| | - Anna E Beaudin
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States
| |
Collapse
|
67
|
Turroni F, Milani C, Duranti S, Lugli GA, Bernasconi S, Margolles A, Di Pierro F, van Sinderen D, Ventura M. The infant gut microbiome as a microbial organ influencing host well-being. Ital J Pediatr 2020; 46:16. [PMID: 32024556 PMCID: PMC7003403 DOI: 10.1186/s13052-020-0781-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/29/2020] [Indexed: 12/16/2022] Open
Abstract
Initial establishment of the human gut microbiota is generally believed to occur immediately following birth, involving key gut commensals such as bifidobacteria that are acquired from the mother. The subsequent development of this early gut microbiota is driven and modulated by specific dietary compounds present in human milk that support selective colonization. This represents a very intriguing example of host-microbe co-evolution, where both partners are believed to benefit. In recent years, various publications have focused on dissecting microbial infant gut communities and their interaction with their human host, being a determining factor in host physiology and metabolic activities. Such studies have highlighted a reduction of microbial diversity and/or an aberrant microbiota composition, sometimes referred to as dysbiosis, which may manifest itself during the early stage of life, i.e., in infants, or later stages of life. There are growing experimental data that may explain how the early human gut microbiota affects risk factors related to adult health conditions. This concept has fueled the development of various nutritional strategies, many of which are based on probiotics and/or prebiotics, to shape the infant microbiota. In this review, we will present the current state of the art regarding the infant gut microbiota and the role of key commensal microorganisms like bifidobacteria in the establishment of the first microbial communities in the human gut.
Collapse
Affiliation(s)
- Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, 43124, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, 43124, Parma, Italy
| | - Sabrina Duranti
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, 43124, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, 43124, Parma, Italy
| | | | - Abelardo Margolles
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA - CSIC, Villaviciosa, Asturias, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias-ISPA, Oviedo, Spain
| | | | - Douwe van Sinderen
- School of Microbiology & APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, 43124, Parma, Italy.
- Microbiome Research Hub, University of Parma, Parma, Italy.
| |
Collapse
|
68
|
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: 107] [Impact Index Per Article: 21.4] [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
|
69
|
The vaginal microbial communities of healthy expectant Brazilian mothers and its correlation with the newborn's gut colonization. World J Microbiol Biotechnol 2019; 35:159. [PMID: 31602538 PMCID: PMC6787113 DOI: 10.1007/s11274-019-2737-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 09/30/2019] [Indexed: 01/22/2023]
Abstract
The female lower genital tract harbors a complex microbial community essential for homeostasis and health. During pregnancy, the female body undergoes unique hormonal changes that contribute to weight gain as well as modulations in immune function that can affect microbiota composition. Several studies have described the vaginal microbiota of pregnant women from the USA, Europe and Mexico. Here we expand our knowledge about the vaginal microbial communities during the third trimester to healthy expectant Brazilian mothers. Vaginal samples were collected from patients delivering at the Hospital de Clínicas de Porto Alegre, Brazil. Microbial DNA was isolated from samples and the V4 region of the 16S rRNA gene was amplified and sequenced using the PGM Ion Torrent. Brazilian pregnant women presented three distinct types of microbial community at the time of labor. Two microbial communities, Cluster 1 and Cluster 3, presented an overall dominance of Lactobacillus while Cluster 2 tended to present higher diversity and richness, with the presence of Pseudomonas, Prevotella and other vaginosis related bacteria. About half of the Brazilian mothers sampled here had dominance of L. iners. The proportion of mothers without dominance of any Lactobacillus was higher in Brazil (22%) compared to UK (2.4%) and USA, where this community type was not detected. The vaginal microbiota showed significant correlation with the composition of the babies' gut microbiota (p-value = 0.002 with a R2 of 15.8%). Mothers presenting different vaginal microbiota shared different microorganisms with their newborns, which would reflect on initial colonizers of the developing newborns' gut.
Collapse
|
70
|
Rhoades N, Barr T, Hendrickson S, Prongay K, Haertel A, Gill L, Garzel L, Whiteson K, Slifka M, Messaoudi I. Maturation of the infant rhesus macaque gut microbiome and its role in the development of diarrheal disease. Genome Biol 2019; 20:173. [PMID: 31451108 PMCID: PMC6709555 DOI: 10.1186/s13059-019-1789-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 08/09/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Diarrhea is the second leading cause of death in children under 5 years of age. Enhanced understanding of causal pathways, pathogenesis, and sequelae of diarrhea is urgently needed. Although the gut microbiota is believed to play a role in susceptibility to diarrheal diseases, our understanding of this association remains incomplete. Infant rhesus macaques (Macaca mulatta) are susceptible to diarrhea making them an ideal model to address this question. RESULTS The maturation of the infant rhesus macaque gut microbiome throughout the first 8 months of life occurs in a similar pattern as that described for human infants. Moreover, the microbiome of the captive reared infant rhesus macaque more closely resembles that of human infants in the developing world than in the western world. Importantly, prior to disease onset, the gut microbiome of infants that later develop diarrhea is enriched in pathways of immunomodulatory metabolite synthesis, while those of infants that remain asymptomatic are enriched in pathways for short-chain fatty acid production. We identify Prevotella strains that are more abundant at 1 month in infants that later develop diarrhea. At 8 months, the microbiomes of animals that experience diarrhea show increased abundance of Campylobacter and a reduction in Helicobacter macacae. CONCLUSION The composition of the microbial community could provide a phenotypic marker of an infant's susceptibility to diarrheal disease. Given the significant physiological and immunological similarities between human and nonhuman primates, these findings provide potential markers of susceptibility to diarrhea that could be modulated to improve infant health, especially in the developing world.
Collapse
Affiliation(s)
- Nicholas Rhoades
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Tasha Barr
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Sara Hendrickson
- Division of Neuroscience, Oregon National Primate Research Center, Portland, OR, USA
| | - Kamm Prongay
- Division of Comparative Medicine, Oregon National Primate Research Center, Oregon Health and Science University West Campus, Portland, OR, USA
| | - Andrew Haertel
- Division of Comparative Medicine, Oregon National Primate Research Center, Oregon Health and Science University West Campus, Portland, OR, USA
| | - Leanne Gill
- California National Primate Research Center, Davis, CA, USA
| | - Laura Garzel
- California National Primate Research Center, Davis, CA, USA
| | - Katrine Whiteson
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Mark Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Portland, OR, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
71
|
Rusconi F, Gagliardi L. Pregnancy Complications and Wheezing and Asthma in Childhood. Am J Respir Crit Care Med 2019; 197:580-588. [PMID: 29064265 DOI: 10.1164/rccm.201704-0744pp] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Franca Rusconi
- 1 Epidemiology Unit, Anna Meyer Children's University Hospital, Florence, Italy; and
| | - Luigi Gagliardi
- 2 Pediatrics and Neonatology Division, Versilia Hospital, Azienda Toscana Nord Ovest, Pisa, Italy
| |
Collapse
|
72
|
Keij FM, Kornelisse RF, Hartwig NG, Mauff K, Poley MJ, Allegaert K, Reiss IKM, Tramper-Stranders GA. RAIN study: a protocol for a randomised controlled trial evaluating efficacy, safety and cost-effectiveness of intravenous-to-oral antibiotic switch therapy in neonates with a probable bacterial infection. BMJ Open 2019; 9:e026688. [PMID: 31289068 PMCID: PMC6615779 DOI: 10.1136/bmjopen-2018-026688] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
INTRODUCTION High morbidity and mortality rates of proven bacterial infection are the main reason for substantial use of intravenous antibiotics in neonates during the first week of life. In older children, intravenous-to-oral switch after 48 hours of intravenous therapy has been shown to have many advantages and is nowadays commonly practised. We, therefore, aim to evaluate the effectiveness, safety and cost-effectiveness of an early intravenous-to-oral switch in neonates with a probable bacterial infection. METHODS AND ANALYSIS We present a protocol for a multicentre randomised controlled trial assessing the non-inferiority of an early intravenous-to-oral antibiotic switch compared with a full course of intravenous antibiotics in neonates (0-28 days of age) with a probable bacterial infection. Five hundred and fifty patients will be recruited in 17 hospitals in the Netherlands. After 48 hours of intravenous treatment, they will be assigned to either continue with intravenous therapy for another 5 days (control) or switch to amoxicillin/clavulanic acid suspension (intervention). Both groups will be treated for a total of 7 days. The primary outcome will be bacterial (re)infection within 28 days after treatment completion. Secondary outcomes are the pharmacokinetic profile of oral amoxicillin/clavulanic acid, the impact on quality of life, cost-effectiveness, impact on microbiome development and additional yield of molecular techniques in diagnosis of probable bacterial infection. ETHICS AND DISSEMINATION This study has been approved by the Medical Ethics Committee of the Erasmus Medical Centre. Results will be presented in peer-reviewed journals and at international conferences. TRIAL REGISTRATION NUMBER NCT03247920.
Collapse
Affiliation(s)
- Fleur M Keij
- Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
- Pediatrics, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - René F Kornelisse
- Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Nico G Hartwig
- Pediatrics, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Katya Mauff
- Biostatistics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marten J Poley
- Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
- Medical Technology Assessment (iMTA), Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Karel Allegaert
- Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Irwin K M Reiss
- Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Gerdien A Tramper-Stranders
- Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
- Pediatrics, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| |
Collapse
|
73
|
Shen X, Wang M, Zhang X, He M, Li M, Cheng G, Wan C, He F. Dynamic construction of gut microbiota may influence allergic diseases of infants in Southwest China. BMC Microbiol 2019; 19:123. [PMID: 31182034 PMCID: PMC6558729 DOI: 10.1186/s12866-019-1489-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/10/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Gut microbes have been suggested as the possible targets in the management of allergic diseases. However, the way in which these microbes influence allergic diseases remain unclear. Forty-seven full-term newborns were selected from a 1000-infant birth cohort. Among them were 23 allergic infants, whereas 24 infants were healthy without allergic symptoms at 1 year of age. Two hundred and sixty-four fecal samples were collected at 7 time points following their birth. These fecal samples were microbiologically analyzed using 16S rRNA gene sequencing. The dynamic processes involved in gut microbiota diversity and composition in the tested infants were constructed. RESULTS Healthy infants demonstrated more statistical differences in longitudinal changes in the alpha diversity of their microbiota at the time points compared with day 0 (meconium) than did allergic infants. Analysis of beta diversity showed that the fecal microbiota of days 0 and 2 comprised different communities in healthy infants, and that there were three separate communities in the fecal microbiota of day 0 of the healthy infants, those of day 2 of the healthy infants, and those of days 0-2 of the allergic infants. The relative abundance of dominant gut microbiota at phylum level varied at different time points in the healthy and diseased groups. Bifidobacterium, Escherichia-Shigella, Streptococcus, Clostridium_sensu_stricto_1, Akkermansia and Erysipelatoclostridium were significantly different between the healthy and diseased groups at a different time points. CONCLUSION The dynamic construction processes of gut microbiota during early life might be associated with the occurrence of long-term allergic diseases, with the first month following birth potentially being the most critical.
Collapse
Affiliation(s)
- Xi Shen
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, No.17 People's South Road, Chengdu, Sichuan, 610041, People's Republic of China
| | - Maolin Wang
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, No.17 People's South Road, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xiao Zhang
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, No.17 People's South Road, Chengdu, Sichuan, 610041, People's Republic of China
| | - Miao He
- West China Second University Hospital, Sichuan University, 610041, Chengdu, Sichuan, People's Republic of China
| | - Ming Li
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, No.17 People's South Road, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Guo Cheng
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, No.17 People's South Road, Chengdu, Sichuan, 610041, People's Republic of China
| | - Chaomin Wan
- Department of Pediatrics of Western China Second Hospital of Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children, 610041, Chengdu, Sichuan, People's Republic of China
| | - Fang He
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and Healthy Food Evaluation Research Center, Sichuan University, No.17 People's South Road, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
74
|
Brewster R, Tamburini FB, Asiimwe E, Oduaran O, Hazelhurst S, Bhatt AS. Surveying Gut Microbiome Research in Africans: Toward Improved Diversity and Representation. Trends Microbiol 2019; 27:824-835. [PMID: 31178123 DOI: 10.1016/j.tim.2019.05.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/09/2019] [Accepted: 05/15/2019] [Indexed: 12/16/2022]
Abstract
Descriptive and translational investigations into the human gut microbiome (GM) are rapidly expanding; however, studies are largely restricted to industrialized populations in the USA and Europe. Little is known about microbial variability and its implications for health and disease in other parts of the world. Populations in Africa are particularly underrepresented. What limited research has been performed has focused on a few subject domains, including the impact of long-term lifestyle and dietary factors on GM ecology, its maturation during infancy, and the interrelationships between the microbiome, infectious disease, and undernutrition. Recently, international consortia have laid the groundwork for large-scale genomics and microbiome studies on the continent, with a particular interest in the epidemiologic transition to noncommunicable disease. Here, we survey the current landscape of GM scholarship in Africa and propose actionable recommendations to improve research capacity and output.
Collapse
Affiliation(s)
- Ryan Brewster
- School of Medicine, Stanford University, Stanford, CA, USA
| | | | - Edgar Asiimwe
- School of Medicine, Stanford University, Stanford, CA, USA
| | - Ovokeraye Oduaran
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa
| | - Scott Hazelhurst
- Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa; School of Electrical and Information Engineering, University of the Witwatersrand, Johannesburg, South Africa.
| | - Ami S Bhatt
- School of Medicine, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University, Stanford, CA, USA; Department of Medicine (Hematology), Stanford University, Stanford, CA, USA.
| |
Collapse
|
75
|
Microbiome of the Skin and Gut in Atopic Dermatitis (AD): Understanding the Pathophysiology and Finding Novel Management Strategies. J Clin Med 2019; 8:jcm8040444. [PMID: 30987008 PMCID: PMC6518061 DOI: 10.3390/jcm8040444] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/25/2019] [Accepted: 03/28/2019] [Indexed: 12/14/2022] Open
Abstract
Atopic dermatitis (AD) is a long-standing inflammatory skin disease that is highly prevalent worldwide. Multiple factors contribute to AD, with genetics as well as the environment affecting disease development. Although AD shows signs of skin barrier defect and immunological deviation, the mechanism underlying AD is not well understood, and AD treatment is often very difficult. There is substantial data that AD patients have a disturbed microbial composition and lack microbial diversity in their skin and gut compared to controls, which contributes to disease onset and atopic march. It is not clear whether microbial change in AD is an outcome of barrier defect or the cause of barrier dysfunction and inflammation. However, a cross-talk between commensals and the immune system is now noticed, and their alteration is believed to affect the maturation of innate and adaptive immunity during early life. The novel concept of modifying skin and gut microbiome by applying moisturizers that contain nonpathogenic biomass or probiotic supplementation during early years may be a preventive and therapeutic option in high risk groups, but currently lacks evidence. This review discusses the nature of the skin and gut flora in AD, possible mechanisms of skin-gut interaction, and the therapeutic implications of microbiome correction in AD.
Collapse
|
76
|
Nicklaus S, Divaret‐Chauveau A, Chardon M, Roduit C, Kaulek V, Ksiazek E, Dalphin M, Karvonen AM, Kirjavainen P, Pekkanen J, Lauener R, Schmausser‐Hechfellner E, Renz H, Braun‐Fahrländer C, Riedler J, Vuitton DA, Mutius EV, Dalphin J. The protective effect of cheese consumption at 18 months on allergic diseases in the first 6 years. Allergy 2019; 74:788-798. [PMID: 30368847 DOI: 10.1111/all.13650] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 09/19/2018] [Accepted: 10/15/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND The effect of exposure to microorganisms on allergic diseases has been well studied. The protective effect of early food diversity against allergic diseases was previously shown in the PASTURE cohort study. The consumption of cheese, a food potentially rich in microbial diversity, deserves further examination. We aimed to evaluate whether cheese consumption is associated with allergic diseases. METHODS In the PASTURE study (birth cohort in 5 European countries), data on feeding practices, environmental factors, and allergic diseases were collected by questionnaires from birth to 6 years (N = 931). Cheese consumption at 18 months of age was quantified in terms of frequency and diversity (ie, number of consumed types among 6 types: hard pressed, semipressed, soft, blue, fresh cheese, and cheese from the farm). Multiple logistic regressions were performed to evaluate the effect of cheese consumption on atopic dermatitis (AD), food allergy (FA), allergic rhinitis, asthma, and atopic sensitization at 6 years after adjustment for confounders of atopy. RESULTS Cheese consumption (vs. nonconsumption) had a significant protective effect on AD (OR = 0.51 [0.29-0.90], P = 0.02) and FA (OR = 0.32, [0.15-0.71], P = 0.004), but no effect on atopic sensitization, allergic rhinitis, and asthma at 6 years. This effect on AD and FA may be related to the diversity of consumed cheeses (OR = 0.64 [0.48-0.85] per cheese type, P = 0.002; OR = 0.55 [0.33-0.92], P = 0.02, respectively). CONCLUSION Although reverse causality cannot totally be ruled out, cheese diversity at 18 months had a protective effect against AD and FA at 6 years in addition to the protective effect of diversity of other foods.
Collapse
Affiliation(s)
- Sophie Nicklaus
- Centre des Sciences du Goût et de l'Alimentation AgroSup Dijon, CNRS INRA Université Bourgogne Franche‐Comté Dijon France
| | - Amandine Divaret‐Chauveau
- Pediatrics Department University Hospital of Besançon Besançon France
- Pediatric Allergy Department University Hospital of Nancy Nancy France
| | - Marie‐Laure Chardon
- University Hospital of Besançon, Respiratory Disease Besançon France
- Hospital of Haute‐Saône, Respiratory disease Vesoul France
| | - Caroline Roduit
- Christine Kühne Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
- Children's Hospital University of Zürich Zürich Switzerland
| | - Vincent Kaulek
- University Hospital of Besançon, Respiratory Disease UMR6249 ChronoEnvironnement CNRS and Université Bourgogne Franche‐Comté Besançon France
| | - Eléa Ksiazek
- Centre des Sciences du Goût et de l'Alimentation AgroSup Dijon, CNRS INRA Université Bourgogne Franche‐Comté Dijon France
| | - Marie‐Laure Dalphin
- University Hospital of Besançon, Respiratory Disease UMR6249 ChronoEnvironnement CNRS and Université Bourgogne Franche‐Comté Besançon France
| | - Anne M. Karvonen
- Department of Health Security National Institute for Health and Welfare Kuopio Finland
| | - Pirkka Kirjavainen
- Department of Health Security National Institute for Health and Welfare Kuopio Finland
| | - Juha Pekkanen
- Department of Health Security National Institute for Health and Welfare Kuopio Finland
- Department of Public Health University of Helsinki Helsinki Finland
| | - Roger Lauener
- Christine Kühne Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
- Children's Hospital of Eastern Switzerland St. Gallen Switzerland
| | | | - Harald Renz
- Department of Clinical Chemistry and Molecular Diagnostics Philipps University of Marburg Marburg Germany
| | | | - Josef Riedler
- Children's Hospital Schwarzach Schwarzach Austria
- Teaching Hospital of Paracelsus Medical Private University Salzburg Salzburg Austria
| | | | - Erika Von Mutius
- Dr. Von Hauner Children's Hospital Ludwig Maximilians University Munich Munich Germany
- Institute for Asthma and Allergy Prevention Helmholtz Centre Munich Neuherberg Germany
- German Center for Lung Research Munich Germany
| | - Jean‐Charles Dalphin
- University Hospital of Besançon, Respiratory Disease UMR6249 ChronoEnvironnement CNRS and Université Bourgogne Franche‐Comté Besançon France
| | | |
Collapse
|
77
|
Gut microbial dysbiosis is associated with allergen-specific IgE responses in young children with airway allergies. World Allergy Organ J 2019; 12:100021. [PMID: 30937143 PMCID: PMC6439417 DOI: 10.1016/j.waojou.2019.100021] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 01/02/2019] [Accepted: 01/29/2019] [Indexed: 12/21/2022] Open
Abstract
Background There is increasing evidence linking alterations of the gut microbial composition during early infancy to the development of atopic diseases and asthma. However, few studies have addressed the association of dysbiotic gut microbiota with allergic reactions through evaluation of feces in young children with allergic airway diseases. Methods We sought to evaluate relationships among gut microbiota, total fecal immunoglobulin E (IgE) levels, serum allergic sensitization, and their relevance to childhood allergic rhinitis and asthma. Microbial composition and diversity were analyzed with Illumina-based 16S rRNA gene sequencing of 89 stool samples collected from children with asthma (n = 35) and allergic rhinitis (n = 28), and from healthy controls (n = 26). Data analysis was performed using Quantitative Insights into Microbial Ecology (QIIME) software. Results A significantly lower abundance of organisms of the phylum Firmicutes were found in children with asthma and allergic rhinitis than in the healthy controls. Relatively lower Chao1 and Shannon indices were also found in children with allergic airway diseases but without any significant difference. Total fecal IgE levels in early childhood were strongly correlated with serum D. pteronyssinus- and D. farinae-specific IgE but not with food-specific IgE levels. In comparison with healthy controls, the genus Dorea was less abundant and negatively correlated with total fecal IgE levels in children with rhinitis, whereas the genus Clostridium was abundant and positively correlated with fecal IgE levels in children with asthma. Conclusions An interaction between particular subsets of gut microbial dysbiosis and IgE-mediated responses to allergens may contribute to the susceptibility to allergic rhinitis and asthma in early childhood.
Collapse
|
78
|
Kim H, Sitarik AR, Woodcroft K, Johnson CC, Zoratti E. Birth Mode, Breastfeeding, Pet Exposure, and Antibiotic Use: Associations With the Gut Microbiome and Sensitization in Children. Curr Allergy Asthma Rep 2019; 19:22. [PMID: 30859338 PMCID: PMC7376540 DOI: 10.1007/s11882-019-0851-9] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW The infant gut microbiota has become a focus of multiple epidemiologic and cohort studies. This microbiome is derived from the mother (via the vaginal canal, maternal skin contact, breastfeeding, and possibly in utero microbial transfer) and is likely influenced by multiple external factors. It is now believed by some experts that colonization and formation of the newborn and alterations of gut microbiota in children are dependent on earlier alterations of the microbiota of mothers during or perhaps even before pregnancy. This review will focus on specific factors (pet keeping, breastfeeding, antibiotic use, and mode of delivery) that influence the infant gut microbiome and atopy. RECENT FINDINGS This is a review of recent literature describing how pet keeping, breastfeeding, antibiotic use, and mode of delivery influences and changes the infant gut microbiome and atopy. General trends in gut microbiota differences have emerged in different birth cohorts when each external factor is analyzed, but consistency between studies is difficult to replicate. The aforementioned factors do not seem to confer an overwhelming risk for development of atopy alone. This review provides a comprehensive review of early life environmental factors and their influence on the infant gut microbiome and atopy.
Collapse
Affiliation(s)
- Haejin Kim
- Division of Allergy and Clinical Immunology, Henry Ford Health System, WSU School of Medicine, One Ford Place 4B, Detroit, MI, 48202, USA.
| | - Alexandra R Sitarik
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | - Kimberley Woodcroft
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | | | - Edward Zoratti
- Division of Allergy and Clinical Immunology, Henry Ford Health System, WSU School of Medicine, One Ford Place 4B, Detroit, MI, 48202, USA
| |
Collapse
|
79
|
Kang X, Tu H, Tian T, Huang Z, Luo L, Shen L, Ye J. Home environment and diseases in early life are associated with allergic rhinitis. Int J Pediatr Otorhinolaryngol 2019; 118:47-52. [PMID: 30578996 DOI: 10.1016/j.ijporl.2018.12.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Risk factors in relation to allergic rhinitis (AR) remain unclear despite considerable interest. This study aimed to analysis the relationship between home environment and diseases in early life and AR. METHODS In a case-control survey, 242 AR patients and 258 healthy persons responded to questionnaires designed to capture pre-pregnancy/pregnancy diseases, maternal medication usage, diseases in early life of participants, family allergic history and home environmental factors. Forty risk factors potentially connected with AR were investigated and analyzed with chi-square test and logistic regression. RESULTS There was no correlation between mother's disorders such as periodontitis, chronic rhinitis, diabetes etc. and AR (p > 0.05). A logistic regression analysis showed that neonatal jaundice (p < 0.001), respiratory system infection (p < 0.001), diarrhea (p < 0.01), eczema (p < 0.01) in the early life and home environmental factors such as house decoration (p < 0.01), mold environment (p < 0.001), keeping flowers and plants (p < 0.001), passive smoking (p < 0.01) were associated with AR. CONCLUSION Diseases in early life and home environment are closely associated with AR.
Collapse
Affiliation(s)
- Xue Kang
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hailuo Tu
- Department of Otorhinolaryngology Head and Neck Surgery, Xinjian District People's Hospital of Nanchang, Nanchang, China
| | - Tengfei Tian
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhiqun Huang
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Liping Luo
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Li Shen
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Ye
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
80
|
Terciolo C, Dapoigny M, Andre F. Beneficial effects of Saccharomyces boulardii CNCM I-745 on clinical disorders associated with intestinal barrier disruption. Clin Exp Gastroenterol 2019; 12:67-82. [PMID: 30804678 PMCID: PMC6375115 DOI: 10.2147/ceg.s181590] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Intestinal barrier defects lead to "leaky gut syndrome", defined as an increase in intestinal permeability that allows the passage of luminal content into intestinal tissue and the bloodstream. Such a compromised intestinal barrier is the main factor underlying the pathogenesis of inflammatory bowel disease, but also commonly occurs in various systemic diseases such as viral infections and metabolic syndrome. The non-pathogenic yeast Saccharomyces boulardii CNCM I-745 has demonstrated its effectiveness as a probiotic in the prevention and treatment of antibiotic-associated, infectious and functional diarrhea. Via multiple mechanisms of action implicated in intestinal barrier function, S. boulardii has beneficial effects on altered intestinal microbiota and epithelial barrier defects in different pathologies. The well-studied probiotic yeast S. boulardii plays a crucial role in the preservation and/or restoration of intestinal barrier function in multiple disorders. This could be of major interest in diseases characterized by alterations in intestinal barrier function.
Collapse
Affiliation(s)
- Chloe Terciolo
- INRA, UMR 1331 Toxalim, Research Center in Food Toxicology, F-31027 Toulouse, France,
- Aix-Marseille Université, INSERM, UMR 911, CRO2, Marseille, France,
| | - Michel Dapoigny
- Médecine Digestive, CHU Estaing, CHU Clermont-Ferrand, Université Clermont Auvergne, INSERM UMR 1107, Neuro-Dol, Clermont-Ferrand, France
| | - Frederic Andre
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc scientifique et technologique de Luminy, Marseille, France
| |
Collapse
|
81
|
Wei X, Jiang P, Liu J, Sun R, Zhu L. Association between probiotic supplementation and asthma incidence in infants: a meta-analysis of randomized controlled trials. J Asthma 2019; 57:167-178. [PMID: 30656984 DOI: 10.1080/02770903.2018.1561893] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective: The increased social and economic burdens for asthma in infants make the prevention of asthma a major public health goal. Probiotics may reduce the risk of asthma in infants. However, randomized controlled trials (RCTs) have shown mixed efficacy outcomes. We performed a meta-analysis of RCTs to investigate whether probiotics are associated with a lower asthma incidence in infants. Methods: The PubMed, Cochrane library, and EMBASE databases were systematically searched from the inception dates to August 2018. RCTs comparing the effects of probiotic supplements with a placebo for asthma or wheeze incidence in infants were included. A meta-analysis was performed to calculate risk ratio (RR) and 95% confidence interval (CI) using the Mantel-Haenszel statistical method. Results: A total of 19 randomized trials involving 5157 children fulfilled the inclusion criteria. There was no significant association of probiotics with risk of asthma (RR, 0.94 [95% CI, 0.82-1.09]) or wheeze (RR, 0.97 [95% CI, 0.88-1.06]) compared with placebo. Subgroup analysis by asthma risk showed that probiotics significantly reduced wheeze incidence among infants with atopy disease (RR, 0.61 [95% CI, 0.42-0.90]), but no significant associations were found in the other subgroup analyses by participants receiving the intervention, timing of intervention, prevention regimen, probiotic organism, duration of intervention, and duration of follow-up. Conclusions: The use of probiotic supplementation compared with placebo was not associated with a lower risk of asthma in infants. These findings do not support recommendation to use probiotics in the prevention of asthma in infants.
Collapse
Affiliation(s)
- Xiaochen Wei
- Department of Pharmacy, Tianjin First Central Hospital, Tianjin, People's Republic of China
| | - Ping Jiang
- Department of Respiration, Tianjin First Central Hospital, Tianjin, People's Republic of China
| | - Jiangbo Liu
- Department of Respiration, Tianjin First Central Hospital, Tianjin, People's Republic of China
| | - Rongfei Sun
- Department of Respiration, Tianjin First Central Hospital, Tianjin, People's Republic of China
| | - Liqin Zhu
- Department of Pharmacy, Tianjin First Central Hospital, Tianjin, People's Republic of China
| |
Collapse
|
82
|
Abstract
Asthma is among the most common chronic diseases worldwide and is a significant contributor to the global health burden, highlighting the urgent need for primary prevention. This article outlines several practical and conceptual challenges that accompany primary prevention efforts. It advocates for improved predictive modeling to identify those at high-risk of developing asthma using automated algorithms within electronic medical records systems and explanatory modeling to refine understanding of causal pathways. Understanding the many issues that are likely to affect the success of primary prevention efforts helps the community of individuals invested in asthma prevention organize efforts and maximize their impact.
Collapse
|
83
|
Boutin RCT, Dwyer Z, Farmer K, Rudyk C, Forbes MR, Hayley S. Perinatal antibiotic exposure alters composition of murine gut microbiota and may influence later responses to peanut antigen. Allergy Asthma Clin Immunol 2018; 14:42. [PMID: 30410548 PMCID: PMC6211427 DOI: 10.1186/s13223-018-0263-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 04/30/2018] [Indexed: 01/06/2023] Open
Abstract
Background Accumulating evidence suggests that the gut microbiota shapes developmental processes within the immune system. Early life antibiotic use is one factor which may contribute to immune dysfunction and the recent surge in allergies by virtue of its effects on gut microbiota. Objective and methods As a first step towards determining whether a relationship exists between perinatal antibiotic induced changes in the gut microbiota and the later development of a peanut allergy, we exposed newborn mice to either the broad-spectrum antibiotic vancomycin or to a vehicle for 6 weeks and then used a novel murine model of peanut allergy. Results Early-life treatment with vancomycin resulted in a significant shift in the gut microbiota community characterized by a reduction in the abundance of firmicutes and preponderance of inflammatory proteobacteria. Mice with an antibiotic-altered microbiota, showed a localized allergic-like response characterized by ear swelling and scratching following intra-dermal peanut antigen challenge. Likewise, circulating IgE levels were increased in antibiotic-treated mice, but no evidence of a systemic allergic or anaphylactic-like response was observed. Importantly, we utilized the naturally occurring pro-inflammatory cytokine, tumor necrosis factor-α (TNF-α), rather than the more commonly used cholera toxin, as an adjuvant together with the peanut antigen. Conclusion Our data suggest that early antibiotic exposure promotes a shift in the gut microbiota community that may in turn, influence how mice later respond to a TNF-α + antigen challenge. However, further studies verifying the capacity of microbiota restoration to protect against allergic responses will be needed to confirm a causal role of antibiotic-induced microbiota variations in promoting allergic disease phenotypes.
Collapse
Affiliation(s)
- Rozlyn C T Boutin
- 1Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6 ON Canada.,2Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6 ON Canada
| | - Zach Dwyer
- 1Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6 ON Canada.,2Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6 ON Canada
| | - Kyle Farmer
- 1Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6 ON Canada.,2Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6 ON Canada
| | - Chris Rudyk
- 1Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6 ON Canada.,2Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6 ON Canada
| | - Mark R Forbes
- 1Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6 ON Canada.,2Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6 ON Canada
| | - Shawn Hayley
- 1Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6 ON Canada.,2Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, K1S 5B6 ON Canada
| |
Collapse
|
84
|
Mitre E, Susi A, Nylund CM. Antibiotics and Acid-Suppressing Medications in Early Life and Allergic Disorders-Reply. JAMA Pediatr 2018; 172:990-991. [PMID: 30128543 DOI: 10.1001/jamapediatrics.2018.2513] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Edward Mitre
- Department of Microbiology and Immunology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Apryl Susi
- Department of Pediatrics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Cade M Nylund
- Department of Pediatrics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
85
|
Salem I, Ramser A, Isham N, Ghannoum MA. The Gut Microbiome as a Major Regulator of the Gut-Skin Axis. Front Microbiol 2018; 9:1459. [PMID: 30042740 PMCID: PMC6048199 DOI: 10.3389/fmicb.2018.01459] [Citation(s) in RCA: 314] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022] Open
Abstract
The adult intestine hosts a myriad of diverse bacterial species that reside mostly in the lower gut maintaining a symbiosis with the human habitat. In the current review, we describe the neoteric advancement in our comprehension of how the gut microbiota communicates with the skin as one of the main regulators in the gut-skin axis. We attempted to explore how this potential link affects skin differentiation and keratinization, its influence on modulating the cutaneous immune response in various diseases, and finally how to take advantage of this communication in the control of different skin conditions.
Collapse
Affiliation(s)
- Iman Salem
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
| | - Amy Ramser
- Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Nancy Isham
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
| | - Mahmoud A. Ghannoum
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
- Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
86
|
Kelly RS, Sordillo JE, Lasky-Su J, Dahlin A, Perng W, Rifas-Shiman SL, Weiss ST, Gold DR, Litonjua AA, Hivert MF, Oken E, Wu AC. Plasma metabolite profiles in children with current asthma. Clin Exp Allergy 2018; 48:1297-1304. [PMID: 29808611 DOI: 10.1111/cea.13183] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 04/13/2018] [Accepted: 04/19/2018] [Indexed: 01/09/2023]
Abstract
BACKGROUND Identifying metabolomic profiles of children with asthma has the potential to increase understanding of asthma pathophysiology. OBJECTIVE To identify differences in plasma metabolites between children with and without current asthma at mid-childhood. METHODS We used untargeted mass spectrometry to measure plasma metabolites in 237 children (46 current asthma cases and 191 controls) in Project Viva, a birth cohort from eastern Massachusetts, USA. Current asthma was assessed at mid-childhood (mean age 8.0 years). The ability of a broad spectrum metabolic profile to distinguish between cases and controls was assessed using partial least squares discriminant analysis. We used logistic regression models to identify individual metabolites that were differentially abundant by case-control status. We tested significant metabolites for replication in 411 children from the VDAART clinical trial. RESULTS There was no evidence of a systematic difference in the metabolome of children reporting current asthma vs. healthy controls according to partial least squares discriminant analysis. However, several metabolites were associated with odds of current asthma at a nominally significant threshold (P < .05), including a metabolite of nicotinamide (N1-Methyl-2-pyridone-5-carboxamide (Odds Ratio (OR) = 2.8 (95% CI 1.1-8.0)), a pyrimidine metabolite (5,6-dihydrothymine (OR = 0.4 (95% CI 0.2-0.9)), bile constituents (biliverdin (OR = 0.4 (95%CI 0.1-0.9), taurocholate (OR = 2.0 (95% CI 1.2-3.4)), two peptides likely derived from fibrinopeptide A (ORs from 1.6 to 1.7), and a gut microbiome metabolite (p-cresol sulphate OR = 0.5 (95% CI 0.2-0.9)). The associations for N1-Methyl-2-pyridone-5-carboxamide and p-cresol sulphate replicated in the independent VDAART population (one-sided P values = .03-.04). CONCLUSIONS AND CLINICAL RELEVANCE Current asthma is nominally associated with altered levels of several metabolites, including metabolites in the nicotinamide pathway, and a bacterial metabolite derived from the gut microbiome.
Collapse
Affiliation(s)
- R S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - J E Sordillo
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA, USA
| | - J Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - A Dahlin
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - W Perng
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - S L Rifas-Shiman
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA, USA
| | - S T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - D R Gold
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - A A Litonjua
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - M-F Hivert
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA, USA.,Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
| | - E Oken
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA, USA.,Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - A C Wu
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA, USA.,Division of General Pediatrics, Department of Pediatrics, Children's Hospital, Boston, MA, USA
| |
Collapse
|
87
|
Sitarik A, Havstad S, Levin A, Lynch SV, Fujimura K, Ownby D, Johnson C, Wegienka G. Dog introduction alters the home dust microbiota. INDOOR AIR 2018; 28:539-547. [PMID: 29468742 PMCID: PMC6003855 DOI: 10.1111/ina.12456] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 02/13/2018] [Indexed: 05/13/2023]
Abstract
Research has largely reported that dog exposure is associated with reduced allergic disease risk. Responsible mechanism(s) are not understood. The goal was to investigate whether introducing a dog into the home changes the home dust microbiota. Families without dogs or cats planning to adopt a dog and those who were not were recruited. Dust samples were collected from the homes at recruitment and 12 months later. Microbiota composition and taxa (V4 region of the 16S rRNA gene) were compared between homes that did and did not adopt a dog. A total of 91 dust samples from 54 families (27 each, dog and no dog; 17 dog and 20 no dog homes with paired samples) were analyzed. A significant dog effect was seen across time in both unweighted UniFrac and Canberra metrics (both P = .008), indicating dog introduction may result in rapid establishment of rarer and phylogenetically related taxa. A significant dog-time interaction was seen in both weighted UniFrac (P < .001) and Bray-Curtis (P = .002) metrics, suggesting that while there may not initially be large relative abundance shifts following dog introduction, differences can be seen within a year. Therefore, dog introduction into the home has both immediate effects and effects that emerge over time.
Collapse
Affiliation(s)
- Alexandra Sitarik
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI
- The in-FLAME Global Network, an affiliate of the World Universities Network (WUN), West New York, NJ 07093 USA
| | - Suzanne Havstad
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI
- The in-FLAME Global Network, an affiliate of the World Universities Network (WUN), West New York, NJ 07093 USA
| | - Albert Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI
| | - Susan V. Lynch
- Division of Gastroenterology, University of California, San Francisco, California
| | - Kei Fujimura
- Division of Gastroenterology, University of California, San Francisco, California
| | - Dennis Ownby
- Department of Pediatrics, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Christine Johnson
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI
- The in-FLAME Global Network, an affiliate of the World Universities Network (WUN), West New York, NJ 07093 USA
| | - Ganesa Wegienka
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI
- The in-FLAME Global Network, an affiliate of the World Universities Network (WUN), West New York, NJ 07093 USA
| |
Collapse
|
88
|
Mogren I, Lindqvist M, Petersson K, Nilses C, Small R, Granåsen G, Edvardsson K. Maternal height and risk of caesarean section in singleton births in Sweden-A population-based study using data from the Swedish Pregnancy Register 2011 to 2016. PLoS One 2018; 13:e0198124. [PMID: 29813118 PMCID: PMC5973605 DOI: 10.1371/journal.pone.0198124] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 05/14/2018] [Indexed: 11/18/2022] Open
Abstract
Caesarean section (CS) has short and long term adverse health consequences, and should therefore only be undertaken when necessary. Risk factors such as maternal age, maternal body mass index (BMI) and fetal weight have been extensively investigated in relation to CS, but the significance of maternal height has been less explored in Sweden. The aim was to investigate the significance of maternal height on risk of CS in a representative, population-based sample from Sweden, also taking into account confounders. Data on singleton births in the Swedish Pregnancy Register 2011 to 2016 were collected, including women with heights of 140 cm and above, constituting a sample of 581,844 women. Data were analysed with epidemiological and biostatistical methods. Mean height was 166.1 cm. Women born outside Sweden were significantly shorter than women born in Sweden (162.8 cm vs. 167.1 cm, p<0.001). There was a decreasing risk of CS with increasing maternal height. This effect remained after adjustment for other risk factors for CS such as maternal age, BMI, gestational age, parity, high birth weight and country of birth. Frequency of CS was higher among women born outside Sweden compared with Swedish-born women (17.3% vs. 16.0%), however, in a multiple regression model country of birth outside Sweden diminished as a risk factor for CS. Maternal height of 178-179 cm was associated with the lowest risk of CS (OR = 0.76, CI95% 0.71-0.81), whereas height below 160 cm explained 7% of CS cases. BMI and maternal age are established factors involved in clinical assessments related to birth, and maternal height should increasingly enjoy a similar status in these considerations. Moreover, when healthcare professionals are counselling pregnant women, taller stature should be more emphasized as a positive indicator for successful vaginal birth to increase pregnant women's confidence in giving birth vaginally, with possible positive impacts for lowering CS rates.
Collapse
Affiliation(s)
- Ingrid Mogren
- Obstetrics and Gynecology, Department of Clinical Sciences, Umeå University, Umeå, Sweden
- Judith Lumley Centre, La Trobe University, Melbourne, Australia
| | - Maria Lindqvist
- Obstetrics and Gynecology, Department of Clinical Sciences, Umeå University, Umeå, Sweden
| | - Kerstin Petersson
- Obstetrics and Gynecology, Department of Clinical Sciences, Umeå University, Umeå, Sweden
| | - Carin Nilses
- Department of Obstetrics and Gynecology, Västernorrland County Hospital, Sundsvall, Sweden
| | - Rhonda Small
- Judith Lumley Centre, La Trobe University, Melbourne, Australia
- Department of Women’s and Children’s Health, Division of Reproductive Health, Karolinska Institute, Stockholm, Sweden
| | - Gabriel Granåsen
- Department of Public Health and Clinical Medicine, Epidemiology and Global Health Unit, Umeå University, Umeå, Sweden
| | - Kristina Edvardsson
- Obstetrics and Gynecology, Department of Clinical Sciences, Umeå University, Umeå, Sweden
- Judith Lumley Centre, La Trobe University, Melbourne, Australia
| |
Collapse
|
89
|
Kang Y, Cai Y. Future prospect of faecal microbiota transplantation as a potential therapy in asthma. Allergol Immunopathol (Madr) 2018; 46:307-309. [PMID: 28803667 DOI: 10.1016/j.aller.2017.04.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 04/17/2017] [Accepted: 04/20/2017] [Indexed: 02/08/2023]
Abstract
There is convincing evidence from both human and animal studies suggesting that the gut microbiota plays an important role in regulating immune responses associated with the development of asthma. Certain intestinal microbial strains have been demonstrated to suppress or impair immune responsiveness in asthma experimental models, suggesting that specific species among gut commensal microbiota may play either a morbific or phylactic role in the progression of asthma. Evidence to date suggests that the intestinal microbiota represent fertile targets for prevention or management of asthma. The faecal microbiota transplantation (FMT) is a rather straightforward therapy that manipulates the human gastrointestinal (GI) microbiota, by which a healthy donor microbiota is transferred into an existing but disturbed microbial ecosystem. The FMT may therefore represent a therapeutic approach for asthma treatment in the foreseeable future. At present, FMT therapy for asthma is very limited and should be actively studied. Considerable efforts are needed to increase our knowledge in the field of FMT therapy for asthma. In this review, we aimed to provide several insights into the development of FMT therapy for asthma.
Collapse
Affiliation(s)
- Y Kang
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China; Genetics and Pharmacogenomics Laboratory, Kunming University of Science and Technology, Kunming, Yunnan, China.
| | - Y Cai
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China; Pathogen Biology Laboratory, Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
90
|
Abstract
Early nutrition may have long-lasting metabolic impacts in adulthood. Even though breast milk is the gold standard, most infants are at least partly formula-fed. Despite obvious improvements, infant formulas remain perfectible to reduce the gap between breastfed and formula-fed infants. Improvements such as reducing the protein content, modulating the lipid matrix and adding prebiotics, probiotics and synbiotics, are discussed regarding metabolic health. Numerous questions remain to be answered on how impacting the infant formula composition may modulate the host metabolism and exert long-term benefits. Interactions between early nutrition (composition of human milk and infant formula) and the gut microbiota profile, as well as mechanisms connecting gut microbiota to metabolic health, are highlighted. Gut microbiota stands as a key actor in the nutritional programming but additional well-designed longitudinal human studies are needed.
Collapse
|
91
|
Milani C, Duranti S, Bottacini F, Casey E, Turroni F, Mahony J, Belzer C, Delgado Palacio S, Arboleya Montes S, Mancabelli L, Lugli GA, Rodriguez JM, Bode L, de Vos W, Gueimonde M, Margolles A, van Sinderen D, Ventura M. The First Microbial Colonizers of the Human Gut: Composition, Activities, and Health Implications of the Infant Gut Microbiota. Microbiol Mol Biol Rev 2017; 81:e00036-17. [PMID: 29118049 PMCID: PMC5706746 DOI: 10.1128/mmbr.00036-17] [Citation(s) in RCA: 1017] [Impact Index Per Article: 145.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The human gut microbiota is engaged in multiple interactions affecting host health during the host's entire life span. Microbes colonize the neonatal gut immediately following birth. The establishment and interactive development of this early gut microbiota are believed to be (at least partially) driven and modulated by specific compounds present in human milk. It has been shown that certain genomes of infant gut commensals, in particular those of bifidobacterial species, are genetically adapted to utilize specific glycans of this human secretory fluid, thus representing a very intriguing example of host-microbe coevolution, where both partners are believed to benefit. In recent years, various metagenomic studies have tried to dissect the composition and functionality of the infant gut microbiome and to explore the distribution across the different ecological niches of the infant gut biogeography of the corresponding microbial consortia, including those corresponding to bacteria and viruses, in healthy and ill subjects. Such analyses have linked certain features of the microbiota/microbiome, such as reduced diversity or aberrant composition, to intestinal illnesses in infants or disease states that are manifested at later stages of life, including asthma, inflammatory bowel disease, and metabolic disorders. Thus, a growing number of studies have reported on how the early human gut microbiota composition/development may affect risk factors related to adult health conditions. This concept has fueled the development of strategies to shape the infant microbiota composition based on various functional food products. In this review, we describe the infant microbiota, the mechanisms that drive its establishment and composition, and how microbial consortia may be molded by natural or artificial interventions. Finally, we discuss the relevance of key microbial players of the infant gut microbiota, in particular bifidobacteria, with respect to their role in health and disease.
Collapse
Affiliation(s)
- Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Sabrina Duranti
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Francesca Bottacini
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Eoghan Casey
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Jennifer Mahony
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Susana Delgado Palacio
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Silvia Arboleya Montes
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Leonardo Mancabelli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Juan Miguel Rodriguez
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Lars Bode
- Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California-San Diego, La Jolla, California, USA
| | - Willem de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Department of Bacteriology & Immunology, RPU Immunobiology, University of Helsinki, Helsinki, Finland
| | - Miguel Gueimonde
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Abelardo Margolles
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
92
|
Vieira Borba V, Sharif K, Shoenfeld Y. Breastfeeding and autoimmunity: Programing health from the beginning. Am J Reprod Immunol 2017; 79. [PMID: 29083070 DOI: 10.1111/aji.12778] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 10/10/2017] [Indexed: 12/19/2022] Open
Abstract
Breast milk is not only a completely adapted nutrition source for the newborn but also an impressive array of immune-active molecules that afford protection against infections and shape mucosal immune responses. Decisive imprinting events might be modulated during the first months of life with potential health long-term effects, enhancing the importance of breastfeeding as a major influence on the immune system correct development and modifying disease susceptibility. The aim of this review was to clarify the link between breastfeeding and autoimmune diseases, inquiring the related mechanisms, based on data available in the literature. Being breastfed was associated with a lower incidence of diabetes, celiac disease, multiple sclerosis and asthma, explained by the protection against early infections, anti-inflammatory properties, antigen-specific tolerance induction, and regulation of infant's microbiome. The protective role of human milk in idiopathic juvenile arthritis, rheumatoid arthritis, and inflammatory bowel diseases remains controversial. On the other hand, the breastfeeding mother faces a health-challenging period in life. High levels of prolactin may lead either to the development of autoimmune diseases in susceptible mothers or exacerbations of current immune-mediated disorders. These features raise the question if mothers with autoimmune diseases, mainly systemic lupus erythematosus, should avoid breastfeeding.
Collapse
Affiliation(s)
- Vânia Vieira Borba
- Department 'A' of Internal Medicine, Coimbra University Hospital Centre, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| | - Kassem Sharif
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel.,Department 'B' of Internal Medicine, Sheba Medical Center, Tel-Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
93
|
Gold DR, Adamkiewicz G, Arshad SH, Celedón JC, Chapman MD, Chew GL, Cook DN, Custovic A, Gehring U, Gern JE, Johnson CC, Kennedy S, Koutrakis P, Leaderer B, Mitchell H, Litonjua AA, Mueller GA, O'Connor GT, Ownby D, Phipatanakul W, Persky V, Perzanowski MS, Ramsey CD, Salo PM, Schwaninger JM, Sordillo JE, Spira A, Suglia SF, Togias A, Zeldin DC, Matsui EC. NIAID, NIEHS, NHLBI, and MCAN Workshop Report: The indoor environment and childhood asthma-implications for home environmental intervention in asthma prevention and management. J Allergy Clin Immunol 2017; 140:933-949. [PMID: 28502823 PMCID: PMC5632590 DOI: 10.1016/j.jaci.2017.04.024] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/14/2017] [Indexed: 01/19/2023]
Abstract
Environmental exposures have been recognized as critical in the initiation and exacerbation of asthma, one of the most common chronic childhood diseases. The National Institute of Allergy and Infectious Diseases; National Institute of Environmental Health Sciences; National Heart, Lung, and Blood Institute; and Merck Childhood Asthma Network sponsored a joint workshop to discuss the current state of science with respect to the indoor environment and its effects on the development and morbidity of childhood asthma. The workshop included US and international experts with backgrounds in allergy/allergens, immunology, asthma, environmental health, environmental exposures and pollutants, epidemiology, public health, and bioinformatics. Workshop participants provided new insights into the biologic properties of indoor exposures, indoor exposure assessment, and exposure reduction techniques. This informed a primary focus of the workshop: to critically review trials and research relevant to the prevention or control of asthma through environmental intervention. The participants identified important limitations and gaps in scientific methodologies and knowledge and proposed and prioritized areas for future research. The group reviewed socioeconomic and structural challenges to changing environmental exposure and offered recommendations for creative study design to overcome these challenges in trials to improve asthma management. The recommendations of this workshop can serve as guidance for future research in the study of the indoor environment and on environmental interventions as they pertain to the prevention and management of asthma and airway allergies.
Collapse
Affiliation(s)
- Diane R Gold
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass.
| | - Gary Adamkiewicz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Syed Hasan Arshad
- David Hide Asthma and Allergy Research Centre, Isle of Wight, and Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Juan C Celedón
- Division of Pulmonary Medicine, Allergy and Immunology, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pa
| | | | - Ginger L Chew
- Centers for Disease Control and Prevention (CDC), National Center for Environmental Health, Division of Environmental Hazards and Health Effects | Air Pollution and Respiratory Health Branch, Atlanta, Ga
| | - Donald N Cook
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Adnan Custovic
- Section of Paediatrics and MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Ulrike Gehring
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - James E Gern
- Departments of Pediatrics and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Christine C Johnson
- Department of Public Health Sciences, Henry Ford Hospital & Health System, Detroit, Mich
| | - Suzanne Kennedy
- Department of Pediatrics, NC Children's Hospital, University of North Carolina, Chapel Hill, NC
| | - Petros Koutrakis
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Brian Leaderer
- Yale School of Public Health, Yale School of Medicine, Yale School of Forestry and Environmental Studies, Center for Perinatal, Pediatric and Environmental Epidemiology (CPPEE), New Haven, Conn
| | | | - Augusto A Litonjua
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass
| | - Geoffrey A Mueller
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - George T O'Connor
- Pulmonary Center, Boston University School of Medicine, Boston, Mass
| | - Dennis Ownby
- Division of Allergy-Immunology and Rheumatology, Department of Pediatrics, Augusta University, Augusta, Ga
| | - Wanda Phipatanakul
- Asthma, Allergy and Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Victoria Persky
- Division of Epidemiology and Biostatistics, School of Public Health, University of Illinois at Chicago, Chicago, Ill
| | - Matthew S Perzanowski
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY
| | - Clare D Ramsey
- Departments of Medicine and Community Health Sciences, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Päivi M Salo
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Julie M Schwaninger
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Md
| | - Joanne E Sordillo
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass
| | - Avrum Spira
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, Mass
| | - Shakira F Suglia
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Ga
| | - Alkis Togias
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Md
| | - Darryl C Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Elizabeth C Matsui
- Division of Pediatric Allergy/Immunology, Johns Hopkins University, Baltimore, Md
| |
Collapse
|
94
|
Amenyogbe N, Kollmann TR, Ben-Othman R. Early-Life Host-Microbiome Interphase: The Key Frontier for Immune Development. Front Pediatr 2017; 5:111. [PMID: 28596951 PMCID: PMC5442244 DOI: 10.3389/fped.2017.00111] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 04/28/2017] [Indexed: 12/14/2022] Open
Abstract
Human existence can be viewed as an "animal in a microbial world." A healthy interaction of the human host with the microbes in and around us heavily relies on a well-functioning immune system. As development of both the microbiota and the host immune system undergo rapid changes in early life, it is not surprising that even minor alterations during this co-development can have profound consequences. Scrutiny of existing data regarding pre-, peri-, as well as early postnatal modulators of newborn microbiota indeed suggest strong associations with several immune-mediated diseases with onset far beyond the newborn period. We here summarize these data and extract overarching themes. This same effort in turn sets the stage to guide effective countermeasures, such as probiotic administration. The objective of our review is to highlight the interaction of host immune ontogeny with the developing microbiome in early life as a critical window of susceptibility for lifelong disease, as well as to identify the enormous potential to protect and promote lifelong health by specifically targeting this window of opportunity.
Collapse
Affiliation(s)
- Nelly Amenyogbe
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tobias R. Kollmann
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Rym Ben-Othman
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
95
|
Lee-Sarwar KA, Bacharier LB, Litonjua AA. Strategies to alter the natural history of childhood asthma. Curr Opin Allergy Clin Immunol 2017; 17:139-145. [PMID: 28079559 PMCID: PMC5664210 DOI: 10.1097/aci.0000000000000340] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Asthma exhibits significant heterogeneity in occurrence and severity over the lifespan. Our goal is to discuss recent evidence regarding determinants of the natural history of asthma during childhood, and review the rationale behind and status of major efforts to alter its course. RECENT FINDINGS Variations in microbial exposures are associated with risk of allergic disease, and the use of bacterial lysates may be a promising preventive strategy. Exposure to air pollution appears to be particularly damaging in prenatal and early life, and interventions to reduce pollution are feasible and result in clinical benefit. E-cigarette use may have a role in harm reduction for conventional cigarette smokers with asthma, but has undefined short-term and long-term effects that must be clarified. Vitamin D insufficiency over the first several years of life is associated with risk of asthma, and vitamin D supplementation reduces the risk of severe exacerbations. SUMMARY The identification of risk factors for asthma occurrence, persistence and severity will continue to guide efforts to alter the natural history of the disease. We have reviewed several promising strategies that are currently under investigation. Vitamin D supplementation and air pollution reduction have been shown to be effective strategies and warrant increased investigation and implementation.
Collapse
Affiliation(s)
- K A Lee-Sarwar
- aDivision of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, Massachusetts bDivision of Pediatric Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine cSt Louis Children's Hospital, St Louis, Missouri dChanning Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
96
|
Remote Sensing between Liver and Intestine: Importance of Microbial Metabolites. ACTA ACUST UNITED AC 2017; 3:101-113. [PMID: 28983453 DOI: 10.1007/s40495-017-0087-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent technological advancements including metagenomics sequencing and metabolomics have allowed the discovery of critical functions of gut microbiota in obesity, malnutrition, neurological disorders, asthma, and xenobiotic metabolism. Classification of the human gut microbiome into distinct "enterotypes" has been proposed to serve as a new paradigm for understanding the interplay between microbial variation and human disease phenotypes, as many organs are affected by gut microbiota modifications during the pathogenesis of diseases. Gut microbiota remotely interacts with liver and other metabolic organs of the host through various microbial metabolites that are absorbed into the systemic circulation. PURPOSE OF REVIEW The present review summarizes recent literature regarding the importance of gut microbiota in modulating the physiological and pathological responses of various host organs, and describes the functions of the known microbial metabolites that are involved in this remote sensing process, with a primary focus on the gut microbiota-liver axis. RECENT FINDINGS Under physiological conditions, gut microbiota modulates the hepatic transcriptome, proteome, and metabolome, most notably down-regulating cytochrome P450 3a mediated xenobiotic metabolism. Gut microbiome also modulates the rhythmicity in liver gene expression, likely through microbial metabolites, such as butyrate and propionate that serve as epigenetic modifiers. Additionally, the production of host hormones such as primary bile acids and glucagon like peptide 1 is altered by gut microbiota to modify intermediary metabolism of the host. SUMMARY Dysregulation of gut microbiota is implicated in various liver diseases such as alcoholic liver disease, non-alcoholic steatohepatitis, liver cirrhosis, cholangitis, and liver cancer. Gut microbiota modifiers such as probiotics and prebiotics are increasingly recognized as novel therapeutic modalities for liver and other types of human diseases.
Collapse
|
97
|
Janoff EN. WITHDRAWN: The microbiome and human disease pathogenesis: how do you do what you do to me …? Transl Res 2016:S1931-5244(16)30273-0. [PMID: 27832937 DOI: 10.1016/j.trsl.2016.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 10/10/2016] [Indexed: 11/30/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Edward N Janoff
- Mucosal and Vaccine Research Program Colorado (MAVRC), Department of Medicine, Division of Infectious Disease, University of Colorado School of Medicine, Aurora, Colo; Denver, Veterans Affairs Medical Center, Denver, Colo
| |
Collapse
|