1
|
Behl A, Sharma KK. Xenobiotics mediated modulation of gut microbiota and its role in lifestyle diseases: a critical appraisal on exposomics. Lett Appl Microbiol 2025; 78:ovaf067. [PMID: 40312786 DOI: 10.1093/lambio/ovaf067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/23/2025] [Accepted: 04/30/2025] [Indexed: 05/03/2025]
Abstract
Gastrointestinal tract of humans provides a niche to thousands of microbes, referred as gut microbiota (GM). GM establishes an intricate relationship with other organs via gut-organ axis, and modulates host health. The structure and functioning of these gut microbes can be influenced by the type of external exposome an individual experiences. Depending upon GM perturbations and host genotype, this can result in variable health implications. On the other hand, the huge arsenal of enzymes possessed by GM can chemically alter the xenobiotic structure. Its consequences can be numerous, including formation of harmful metabolites that cause organ damage, reversal of host detoxification pathways, or favourable health effects. Additionally, GM-mediated bio-transformation of pharmaceuticals can alter their pharmacokinetics and pharmacodynamics, potentially yielding variable drug responses, resulting into prolonged or ineffective treatments. To address this bi-facial relationship and the pivotal role of GM, this review incorporates recent in vitro, in vivo, and multiomics studies. It also suggests the need of machine learning approaches to decode the complex host-microbiota-xenobiotics interactions. These knowledge will aid in comprehending recent rise in chronic lifestyle-diseases which poses a huge burden on the health sector, and can also be a learning curve in making formulations and therapies for personalized treatment.
Collapse
Affiliation(s)
- Arush Behl
- Laboratory of Enzymology and Gut Microbiology, Maharshi Dayanand University, Rohtak 124001, India
| | - Krishna Kant Sharma
- Laboratory of Enzymology and Gut Microbiology, Maharshi Dayanand University, Rohtak 124001, India
| |
Collapse
|
2
|
Konkol JE, Becker B, Paulmann D, Steinmann E, Todt D, Meister TL, Evers S, Dotzauer A, Weide M, Brill FHH. Virucidal activity of household laundry detergents and additives in simulated wash cycles. J Appl Microbiol 2025; 136:lxaf057. [PMID: 40097304 DOI: 10.1093/jambio/lxaf057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 02/11/2025] [Accepted: 03/16/2025] [Indexed: 03/19/2025]
Abstract
AIMS In this study, we evaluated the antiviral performance of domestic laundry detergents and additives to provide information for selecting a suitable product for reducing viruses on textiles during viral infections at home. METHODS AND RESULTS Three laundry heavy-duty detergents (HDDs)-universal solid, regular liquid, and compact single-dose liquid-and a laundry additive (hygiene rinser) were tested for their efficacy using a suspension test and lab-scale laundering against two viruses: enveloped modified vaccinia virus Ankara (MVA) and nonenveloped murine norovirus (MNV). All HDD detergents effectively inactivated MVA at 30°C (titers reduced below quantification limit), whereas the hygiene rinser at 20°C exhibited lower efficacy, with 2.19 log10 reduction after 30 min. Notably, at low temperatures, HDD solid effectively reduced the MNV titer (5 log10 reduction after 30 min). CONCLUSIONS Laundry detergents, including HDD liquid detergents, can inactivate MVA at 30°C. However, for complete inactivation of nonenveloped viruses like norovirus, detergents containing bleach, like the universal solid, are required.
Collapse
Affiliation(s)
- Justyna E Konkol
- Dr. Brill + Partner GmbH, Institute for Hygiene and Microbiology, 28259 Bremen, Germany
| | - Britta Becker
- Dr. Brill + Partner GmbH, Institute for Hygiene and Microbiology, 28259 Bremen, Germany
| | - Dajana Paulmann
- Dr. Brill + Partner GmbH, Institute for Hygiene and Microbiology, 28259 Bremen, Germany
| | - Eike Steinmann
- Department of Molecular and Medical Virology, Ruhr University Bochum, 44801 Bochum, Germany
- German Center for Infection Research (DZIF), External Partner Site, Bochum, Germany
| | - Daniel Todt
- Department of Molecular and Medical Virology, Ruhr University Bochum, 44801 Bochum, Germany
- Department of Translational and Computational Infection Research, Ruhr University Bochum, Bochum, Germany
- European Virus Bioinformatics Center, Friedrich-Schiller University Jena, 07743 Jena, Germany
| | - Toni L Meister
- Department of Molecular and Medical Virology, Ruhr University Bochum, 44801 Bochum, Germany
- Institute for Infection Research and Vaccine Development, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine (BNITM), 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Stefan Evers
- Henkel AG & Co. KGaA, Corporate Microbiology, Henkelstraße 67, 40191 Düsseldorf, Germany
| | - Andreas Dotzauer
- Laboratory for Virus Research, University Bremen, 28359 Bremen, Germany
| | - Mirko Weide
- Henkel AG & Co KGaA, Microbiology and Hygiene, International R&D Henkel Consumer Brands, Henkelstraße 67, 40191 Düsseldorf, Germany
| | - Florian H H Brill
- Dr. Brill + Partner GmbH, Institute for Hygiene and Microbiology, 28259 Bremen, Germany
| |
Collapse
|
3
|
Donkers A, Seel W, Klümpen L, Simon MC. The Multiple Challenges of Nutritional Microbiome Research During COVID-19-A Perspective and Results of a Single-Case Study. Nutrients 2024; 16:3693. [PMID: 39519526 PMCID: PMC11547757 DOI: 10.3390/nu16213693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
The global coronavirus disease 2019 (COVID-19) pandemic has affected multiple aspects of people's lives, which may also influence the results of studies conducted during this period across diverse research domains. This particularly includes the field of nutritional science, investigating the gut microbiota as a potential mediator in the association between dietary intake and health-related outcomes. This article identifies the challenges currently facing this area of research, points out potential solutions, and highlights the necessity to consider a range of issues when interpreting trials conducted during this period. Some of these issues have arisen specifically because of the measures implemented to interrupt the spread of small acute respiratory syndrome coronavirus 2 (SARS-CoV-2), while others remain relevant beyond the pandemic.
Collapse
Affiliation(s)
| | | | | | - Marie-Christine Simon
- Nutrition and Microbiota, Institute of Nutrition and Food Science, University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
4
|
Bhatt S, Patel A, Kesselman MM, Demory ML. Hand Sanitizer: Stopping the Spread of Infection at a Cost. Cureus 2024; 16:e61846. [PMID: 38975405 PMCID: PMC11227450 DOI: 10.7759/cureus.61846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/06/2024] [Indexed: 07/09/2024] Open
Abstract
The recent rise in hand sanitizer use due to the COVID-19 pandemic has had a beneficial impact on stopping the spread of disease, but the potential negative implications of its overuse on the body and the microbiome have yet to be thoroughly reviewed. Epidermal layers absorb hand sanitizer from direct application to the skin, making them some of the most susceptible cells to the adverse effects of overuse. The increased usage of hand sanitizer can affect the variation, quantity, and diversity of the skin microflora, leading to conditions such as eczema, atopic dermatitis, and even systemic toxicity due to colonization of the skin with pathogenic bacteria. Due to the close-knit relationship between the skin and gut, the gastrointestinal system can also incur disruptions due to the negative effects on the skin as a result of excessive hand sanitizer use, leading to gut dysbiosis. Additionally, the accidental ingestion of hand sanitizer, and its abuse or misuse, can be toxic and lead to alcohol poisoning, which is an issue most commonly seen not only in the pediatric population but also in adolescents and adults due to aberrant recreational exposure. As a vulnerable body system, the eyes can also be negatively impacted by hand sanitizer misuse leading to chemical injury, visual impairment, and even blindness. In this review, we aim to highlight the variations in hand sanitizer formulation, the benefits, and how misuse or overuse may lead to adverse effects on the skin, gut, and eyes. In particular, we review the advantages and disadvantages of alcohol-based hand sanitizers (ABHSs) and non-alcohol-based hand sanitizers (NABHSs) and how the components and chemicals used in each can contribute to organ dysbiosis and systemic damage.
Collapse
Affiliation(s)
- Shreya Bhatt
- Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, USA
| | - Aasha Patel
- Dentistry, Roseman University College of Dental Medicine, South Jordan, USA
| | - Marc M Kesselman
- Rheumatology, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, USA
| | - Michelle L Demory
- Microbiology and Immunology, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, USA
| |
Collapse
|
5
|
Lahiani M, Gokulan K, Sutherland V, Cunny HC, Cerniglia CE, Khare S. Early Developmental Exposure to Triclosan Impacts Fecal Microbial Populations, IgA and Functional Activities of the Rat Microbiome. J Xenobiot 2024; 14:193-213. [PMID: 38390992 PMCID: PMC10885032 DOI: 10.3390/jox14010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/30/2023] [Accepted: 01/06/2024] [Indexed: 02/24/2024] Open
Abstract
Triclosan (TCS), a broad-spectrum antibacterial chemical, is detected in human urine, breast milk, amniotic fluid, and feces; however, little is known about its impact on the intestinal microbiome and host mucosal immunity during pregnancy and early development. Pregnant female rats were orally gavaged with TCS from gestation day (GD) 6 to postpartum (PP) day 28. Offspring were administered TCS from postnatal day (PND) 12 to 28. Studies were conducted to assess changes in the intestinal microbial population (16S-rRNA sequencing) and functional analysis of microbial genes in animals exposed to TCS during pregnancy (GD18), and at PP7, PP28 and PND28. Microbial abundance was compared with the amounts of TCS excreted in feces and IgA levels in feces. The results reveal that TCS decreases the abundance of Bacteroidetes and Firmicutes with a significant increase in Proteobacteria. At PND28, total Operational Taxonomic Units (OTUs) were higher in females and showed correlation with the levels of TCS and unbound IgA in feces. The significant increase in Proteobacteria in all TCS-treated rats along with the increased abundance in OTUs that belong to pathogenic bacterial communities could serve as a signature of TCS-induced dysbiosis. In conclusion, TCS can perturb the microbiome, the functional activities of the microbiome, and activate mucosal immunity during pregnancy and early development.
Collapse
Affiliation(s)
- Mohamed Lahiani
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
| | - Kuppan Gokulan
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
| | - Vicki Sutherland
- National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA
| | - Helen C Cunny
- National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA
| | - Carl E Cerniglia
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
| | - Sangeeta Khare
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
| |
Collapse
|
6
|
Peng Y, Tun HM, Ng SC, Wai HKF, Zhang X, Parks J, Field CJ, Mandhane P, Moraes TJ, Simons E, Turvey SE, Subbarao P, Brook JR, Takaro TK, Scott JA, Chan FKL, Kozyrskyj AL. Maternal smoking during pregnancy increases the risk of gut microbiome-associated childhood overweight and obesity. Gut Microbes 2024; 16:2323234. [PMID: 38436093 PMCID: PMC10913716 DOI: 10.1080/19490976.2024.2323234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 02/21/2024] [Indexed: 03/05/2024] Open
Abstract
Childhood obesity is linked to maternal smoking during pregnancy. Gut microbiota may partially mediate this association and could be potential targets for intervention; however, its role is understudied. We included 1,592 infants from the Canadian Healthy Infants Longitudinal Development Cohort. Data on environmental exposure and lifestyle factors were collected prenatally and throughout the first three years. Weight outcomes were measured at one and three years of age. Stool samples collected at 3 and 12 months were analyzed by sequencing the V4 region of 16S rRNA to profile microbial compositions and magnetic resonance spectroscopy to quantify the metabolites. We showed that quitting smoking during pregnancy did not lower the risk of offspring being overweight. However, exclusive breastfeeding until the third month of age may alleviate these risks. We also reported that maternal smoking during pregnancy significantly increased Firmicutes abundance and diversity. We further revealed that Firmicutes diversity mediates the elevated risk of childhood overweight and obesity linked to maternal prenatal smoking. This effect possibly occurs through excessive microbial butyrate production. These findings add to the evidence that women should quit smoking before their pregnancies to prevent microbiome-mediated childhood overweight and obesity risk, and indicate the potential obesogenic role of excessive butyrate production in early life.
Collapse
Affiliation(s)
- Ye Peng
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Microbiota I-Center (MagIC), Hong Kong, SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Hein M Tun
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Microbiota I-Center (MagIC), Hong Kong, SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Siew C Ng
- Microbiota I-Center (MagIC), Hong Kong, SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Department of Medicine and Therapeutics, Institute of Digestive Disease, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Hogan Kok-Fung Wai
- HKU-Pasteur Research Pole, School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Xi Zhang
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Jaclyn Parks
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
- Cancer Control Research, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Catherine J Field
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Piush Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Theo J Moraes
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Elinor Simons
- Department of Pediatrics and Child Health, Children’s Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| | - Stuart E Turvey
- Department of Pediatrics, Child and Family Research Institute, BC Children’s Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Padmaja Subbarao
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Jeffrey R Brook
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Tim K Takaro
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - James A Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Francis KL Chan
- Microbiota I-Center (MagIC), Hong Kong, SAR, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Anita L Kozyrskyj
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
7
|
Bradley E, Haran J. The human gut microbiome and aging. Gut Microbes 2024; 16:2359677. [PMID: 38831607 PMCID: PMC11152108 DOI: 10.1080/19490976.2024.2359677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
The composition of the human gut microbiome has been observed to change over the course of an individual's life. From birth, it is shaped by mode of delivery, diet, environmental exposures, geographic location, exposures to medications, and by aging itself. Here, we present a narrative review of the gut microbiome across the lifespan with a focus on its impacts on aging and age-related diseases in humans. We will describe how it is shaped, and features of the gut microbiome that have been associated with diseases at different phases of life and how this can adversely affect healthy aging. Across the lifespan, and especially in old age, a diverse microbiome that includes organisms suspected to produce anti-inflammatory metabolites such as short-chain fatty acids, has been reported to be associated with healthy aging. These findings have been remarkably consistent across geographic regions of the world suggesting that they could be universal features of healthy aging across all cultures and genetic backgrounds. Exactly how these features of the microbiome affect biologic processes associated with aging thus promoting healthy aging will be crucial to targeting the gut microbiome for interventions that will support health and longevity.
Collapse
Affiliation(s)
- Evan Bradley
- UMass Chan Medical School, Department of Emergency Medicine and Department of Microbiology and Physiologic Systems, Program in Microbiome Dynamics, Worcester, MA, USA
| | - John Haran
- UMass Chan Medical School, Department of Emergency Medicine and Department of Microbiology and Physiologic Systems, Program in Microbiome Dynamics, Worcester, MA, USA
| |
Collapse
|
8
|
Yong GJM, Porsche CE, Sitarik AR, Fujimura KE, McCauley K, Nguyen DT, Levin AM, Woodcroft KJ, Ownby DR, Rundle AG, Johnson CC, Cassidy-Bushrow A, Lynch SV. Precocious infant fecal microbiome promotes enterocyte barrier dysfuction, altered neuroendocrine signaling and associates with increased childhood obesity risk. Gut Microbes 2024; 16:2290661. [PMID: 38117587 PMCID: PMC10761186 DOI: 10.1080/19490976.2023.2290661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/29/2023] [Indexed: 12/22/2023] Open
Abstract
Early life gut microbiome composition has been correlated with childhood obesity, though microbial functional contributions to disease origins remain unclear. Here, using an infant birth cohort (n = 349) we identify a distinct fecal microbiota composition in 1-month-old infants with the lowest rate of exclusive breastfeeding, that relates with higher relative risk for obesity and overweight phenotypes at two years. Higher-risk infant fecal microbiomes exhibited accelerated taxonomic and functional maturation and broad-ranging metabolic reprogramming, including reduced concentrations of neuro-endocrine signals. In vitro, exposure of enterocytes to fecal extracts from higher-risk infants led to upregulation of genes associated with obesity and with expansion of nutrient sensing enteroendocrine progenitor cells. Fecal extracts from higher-risk infants also promoted enterocyte barrier dysfunction. These data implicate dysregulation of infant microbiome functional development, and more specifically promotion of enteroendocrine signaling and epithelial barrier impairment in the early-life developmental origins of childhood obesity.
Collapse
Affiliation(s)
- Germaine J. M. Yong
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA, USA
- Asian Microbiome Library Pte Ltd, Singapore and Singapore Institute of Food and Biotechnology Innovation, Singapore, Singapore
| | - Cara E. Porsche
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Alexandra R. Sitarik
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | - Kei E. Fujimura
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA, USA
- Genetic Disease Laboratory, California Department of Public Health, San Francisco, CA, USA
| | - Kathryn McCauley
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Dat T. Nguyen
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Albert M. Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | | | - Dennis R. Ownby
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Augusta University, Augusta, GA, USA
| | - Andrew G. Rundle
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Christine C. Johnson
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | | | - Susan V. Lynch
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
9
|
Yan M, Xu C, Li C, Feng Y, Duan J, Zhao K, Wu D, Li G, Yang S, Han X, Xie Y, Huang Y, Yu X, Wu J, Zou L. Effects of environmental disinfection on microbial population and resistance genes: A case study of the microecology within a panda enclosure. ENVIRONMENTAL RESEARCH 2023; 235:116662. [PMID: 37453509 DOI: 10.1016/j.envres.2023.116662] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/02/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Widespread use of disinfectants raises concerns over their involvement in altering microbial communities and promoting antimicrobial resistance. This study explores the influence of disinfection protocols on microbial populations and resistance genes within an isolated enclosure environment and in the gut of giant pandas (GPs) held within. Samples of panda feces, air conditioning ducts, soil and bamboo were collected before and after disinfection. High-throughput sequencing characterized the microbial flora of GP gut and environmental microbes inside the artificial habitat. Microbial cultures showed that Escherichia coli (34.6%), Enterococcus (15.4%) and other pathogenic bacteria deposited in feces and the enclosure. Isolates exhibit a consistent resistance to disinfectant, with the greatest resistance shown to cyanuric acid, and the lowest to glutaraldehyde-dodecyl dimethyl ammonium bromide (GD-DDAB) and dodecyl dimethyl ammonium bromide (DDAB). The total number of the culturable bacteria in soil and bamboo were significantly diminished after disinfection but increased in the gut. After disinfection, the richness (Chao1 index) of environment samples increased significantly (P < 0.05), while the richness in gut decreased significantly (P < 0.05). Ten genera showed significant change in feces after disinfection. Metagenome sequencing showed that 126 types of virulence genes were present in feces before disinfection and 37 in soil. After disinfection, 110 virulence genes localized in feces and 53 in soil. Eleven virulence genes including ECP and T2SS increased in feces. A total of 182 antibiotic resistance genes (ARGs) subtypes, potentially conferring resistance to 20 classes of drugs, were detected in the soils and feces, with most belonging to efflux pump protein pathways. After disinfection, the number of resistance genes increased both in gut and soil, which suggests disinfection protocols increase the number of resistance pathways. Our study shows that the use of disinfectants helps to shape the microbial community of GPs and their habitat, and increases populations of resistant strain bacteria.
Collapse
Affiliation(s)
- Min Yan
- College of Resources, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Chunzhong Xu
- Shanghai Wild Animal Park, Shanghai, 201399, China
| | - Caiwu Li
- Key Laboratory of State Forestry and Grassland Administration (SFGA) on Conservation Biology of Rare Animals in the Giant Panda National Park, The China Conservation and Research Center for the Giant Panda (CCRCGP), Dujiangyan, 611830, China
| | - Yongqi Feng
- Shanghai Wild Animal Park, Shanghai, 201399, China
| | - Juntang Duan
- Shanghai Wild Animal Park, Shanghai, 201399, China
| | - Ke Zhao
- College of Resources, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Daifu Wu
- Key Laboratory of State Forestry and Grassland Administration (SFGA) on Conservation Biology of Rare Animals in the Giant Panda National Park, The China Conservation and Research Center for the Giant Panda (CCRCGP), Dujiangyan, 611830, China
| | - Guo Li
- Key Laboratory of State Forestry and Grassland Administration (SFGA) on Conservation Biology of Rare Animals in the Giant Panda National Park, The China Conservation and Research Center for the Giant Panda (CCRCGP), Dujiangyan, 611830, China
| | - Shengzhi Yang
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, Sichuan, China
| | - Xinfeng Han
- College of Veterinary Science, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yue Xie
- College of Veterinary Science, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yan Huang
- Key Laboratory of State Forestry and Grassland Administration (SFGA) on Conservation Biology of Rare Animals in the Giant Panda National Park, The China Conservation and Research Center for the Giant Panda (CCRCGP), Dujiangyan, 611830, China
| | - Xiumei Yu
- College of Resources, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Jiawei Wu
- College of Resources, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Likou Zou
- College of Resources, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
10
|
Querdasi FR, Vogel SC, Thomason ME, Callaghan BL, Brito NH. A comparison of the infant gut microbiome before versus after the start of the covid-19 pandemic. Sci Rep 2023; 13:13289. [PMID: 37587195 PMCID: PMC10432475 DOI: 10.1038/s41598-023-40102-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/04/2023] [Indexed: 08/18/2023] Open
Abstract
The COVID-19 pandemic and resulting public health directives led to many changes in families' social and material environments. Prior research suggests that these changes are likely to impact composition of the gut microbiome, particularly during early childhood when the gut microbiome is developing most rapidly. Importantly, disruption to the gut microbiome during this sensitive period can have potentially long-lasting impacts on health and development. In the current study, we compare gut microbiome composition among a socioeconomically and racially diverse group of 12-month old infants living in New York City who provided stool samples before the pandemic (N = 34) to a group who provided samples during the first 9-months of the pandemic (March-December 2020; N = 20). We found that infants sampled during the pandemic had lower alpha diversity of the microbiome, lower abundance of Pasteurellaceae and Haemophilus, and significantly different beta diversity based on unweighted Unifrac distance than infants sampled before the pandemic. Exploratory analyses suggest that gut microbiome changes due to the pandemic occurred relatively quickly after the start of the pandemic and were sustained. Our results provide evidence that pandemic-related environmental disruptions had an impact on community-level taxonomic diversity of the developing gut microbiome, as well as abundance of specific members of the gut bacterial community.
Collapse
|
11
|
Ren J, Li H, Zeng G, Pang B, Wang Q, Wei J. Gut microbiome-mediated mechanisms in aging-related diseases: are probiotics ready for prime time? Front Pharmacol 2023; 14:1178596. [PMID: 37324466 PMCID: PMC10267478 DOI: 10.3389/fphar.2023.1178596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/24/2023] [Indexed: 06/17/2023] Open
Abstract
Chronic low-grade inflammation affects health and is associated with aging and age-related diseases. Dysregulation of the gut flora is an important trigger for chronic low-grade inflammation. Changes in the composition of the gut flora and exposure to related metabolites have an effect on the inflammatory system of the host. This results in the development of crosstalk between the gut barrier and immune system, contributing to chronic low-grade inflammation and impairment of health. Probiotics can increase the diversity of gut microbiota, protect the gut barrier, and regulate gut immunity, thereby reducing inflammation. Therefore, the use of probiotics is a promising strategy for the beneficial immunomodulation and protection of the gut barrier through gut microbiota. These processes might positively influence inflammatory diseases, which are common in the elderly.
Collapse
Affiliation(s)
- Jing Ren
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Huimin Li
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guixing Zeng
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Boxian Pang
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Qiuhong Wang
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junping Wei
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
12
|
Merenstein D, Pot B, Leyer G, Ouwehand AC, Preidis GA, Elkins CA, Hill C, Lewis ZT, Shane AL, Zmora N, Petrova MI, Collado MC, Morelli L, Montoya GA, Szajewska H, Tancredi DJ, Sanders ME. Emerging issues in probiotic safety: 2023 perspectives. Gut Microbes 2023; 15:2185034. [PMID: 36919522 PMCID: PMC10026873 DOI: 10.1080/19490976.2023.2185034] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/22/2023] [Indexed: 03/16/2023] Open
Abstract
Probiotics are used for both generally healthy consumers and in clinical settings. However, theoretical and proven adverse events from probiotic consumption exist. New probiotic strains and products, as well as expanding use of probiotics into vulnerable populations, warrants concise, and actionable recommendations on how to work toward their safe and effective use. The International Scientific Association for Probiotics and Prebiotics convened a meeting to discuss and produce evidence-based recommendations on potential acute and long-term risks, risks to vulnerable populations, the importance for probiotic product quality to match the needs of vulnerable populations, and the need for adverse event reporting related to probiotic use. The importance of whole genome sequencing, which enables determination of virulence, toxin, and antibiotic resistance genes, as well as clear assignment of species and strain identity, is emphasized. We present recommendations to guide the scientific and medical community on judging probiotic safety.
Collapse
Affiliation(s)
- Daniel Merenstein
- Department of Family Medicine, Georgetown University Medical Center, Washington, DCUSA
| | - Bruno Pot
- Yakult Europe BV, Almere, Netherlands
| | | | - Arthur C. Ouwehand
- Global Health & Nutrition Sciences, International Flavors & Fragrances, Kantvik, Finland
| | - Geoffrey A. Preidis
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Christopher A. Elkins
- Clinical and Environmental Microbiology Branch, Division of Healthcare Quality Promotion, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Colin Hill
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Andi L. Shane
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Emory Children’s Center, Atlanta, Georgia
| | - Niv Zmora
- Scientific consultant, Elinav Lab, Immunology Department, Weizmann Institute of Science, Department of Gastroenterology and Liver Diseases, Tel Aviv, Israel
| | | | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain
| | - Lorenzo Morelli
- Department of Food Science and Technology, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Gina A. Montoya
- Department of Chemical Risk Assessment, Nestlé S.A., Lausanne, Switzerland
| | - Hania Szajewska
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Daniel J. Tancredi
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, USA
| | - Mary Ellen Sanders
- International Scientific Association for Probiotics and Prebiotics, Centennial, CO, USA
| |
Collapse
|
13
|
Obiakor CV, Parks J, Takaro TK, Tun HM, Morales-Lizcano N, Azad MB, Mandhane PJ, Moraes TJ, Simons E, Turvey SE, Subbarao P, Scott JA, Kozyrskyj AL. Early Life Antimicrobial Exposure: Impact on Clostridioides difficile Colonization in Infants. Antibiotics (Basel) 2022; 11:antibiotics11070981. [PMID: 35884235 PMCID: PMC9311587 DOI: 10.3390/antibiotics11070981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022] Open
Abstract
The relationship between antibiotic use and Clostridioides difficile (C. difficile) has been well established in adults and older children but remains unclear and is yet to be fully examined in infant populations. This study aimed to determine the separate and cumulative impact from antibiotics and household cleaning products on C. difficile colonization in infants. This study included 1429 infants at 3–4 months of age and 1728 infants at 12 months of age from the Canadian Healthy Infant Longitudinal Development (CHILD) birth cohort. The levels of infant antimicrobial exposure were obtained from hospital birth charts and standardized questionnaires. Infant gut microbiota was characterized by Illumina 16S ribosomal ribonucleic acid (rRNA) gene sequencing. Analysis of C. difficile was performed using a quantitative polymerase chain reaction (qPCR). Overall, C. difficile colonized 31% and 46% of infants at 3–4 months and 12 months, respectively. At 3–4 months, C. difficile colonization was significantly higher in infants exposed to both antibiotics and higher (above average) usage of household cleaning products (adjusted odds ratio (aOR) 1.50, 95% CI 1.03–2.17; p = 0.032) than in infants who had the least antimicrobial exposure. This higher colonization persisted up to 12 months of age. Our study suggests that cumulative exposure to systemic antibiotics and higher usage of household cleaning products facilitates C. difficile colonization in infants. Further research is needed to understand the future health impacts.
Collapse
Affiliation(s)
| | - Jaclyn Parks
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC V5A 1S6, Canada; (J.P.); (T.K.T.)
- Cancer Control Research, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Tim K. Takaro
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC V5A 1S6, Canada; (J.P.); (T.K.T.)
| | - Hein M. Tun
- School of Public Health, University of Hong Kong, Hong Kong;
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong
- Microbiota I-Center (MagIC), The Chinese University of Hong Kong, Hong Kong
| | - Nadia Morales-Lizcano
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada; (N.M.-L.); (J.A.S.)
| | - Meghan B. Azad
- Department of Pediatrics & Child Health, University of Manitoba, Winnipeg, MB R3A 1S1, Canada; (M.B.A.); (E.S.)
| | | | - Theo J. Moraes
- Department of Pediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada; (T.J.M.); (P.S.)
| | - Elinor Simons
- Department of Pediatrics & Child Health, University of Manitoba, Winnipeg, MB R3A 1S1, Canada; (M.B.A.); (E.S.)
| | - Stuart E. Turvey
- Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 0B3, Canada;
| | - Padmaja Subbarao
- Department of Pediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada; (T.J.M.); (P.S.)
| | - James A. Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada; (N.M.-L.); (J.A.S.)
| | - Anita L. Kozyrskyj
- School of Public Health, University of Alberta, Edmonton, AB T6G 1C9, Canada;
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 1C9, Canada;
- Correspondence:
| |
Collapse
|
14
|
Reyna ME, Petersen C, Dai DLY, Dai R, Becker AB, Azad MB, Miliku K, Lefebvre DL, Moraes TJ, Mandhane PJ, Boutin RCT, Finlay BB, Simons E, Kozyrskyj AL, Lou W, Turvey SE, Subbarao P. Longitudinal body mass index trajectories at preschool age: children with rapid growth have differential composition of the gut microbiota in the first year of life. Int J Obes (Lond) 2022; 46:1351-1358. [PMID: 35428865 PMCID: PMC9239911 DOI: 10.1038/s41366-022-01117-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 12/20/2022]
Abstract
BACKGROUND/OBJECTIVE The steep rise in childhood obesity has emerged as a worldwide public health problem. The first 4 years of life are a critical window where long-term developmental patterns of body mass index (BMI) are established and a critical period for microbiota maturation. Understanding how the early-life microbiota relate to preschool growth may be useful for identifying preventive interventions for childhood obesity. We aim to investigate whether longitudinal shifts within the bacterial community between 3 months and 1 year of life are associated with preschool BMI z-score trajectories. METHODS BMI trajectories from birth to 5 years of age were identified using group-based trajectory modeling in 3059 children. Their association with familial and environmental factors were analyzed. Infant gut microbiota at 3 months and 1 year was defined by 16S RNA sequencing and changes in diversity and composition within each BMIz trajectory were analyzed. RESULTS Four BMIz trajectories were identified: low stable, normative, high stable, and rapid growth. Infants in the rapid growth trajectory were less likely to have been breastfed, and gained less microbiota diversity in the first year of life. Relative abundance of Akkermansia increased with age in children with stable growth, but decreased in those with rapid growth, abundance of Ruminococcus and Clostridium at 1 year were elevated in children with rapid growth. Children who were breastfed at 6 months had increased levels of Sutterella, and decreased levels of Ruminococcus and Clostridium. CONCLUSION This study provides new insights into the relationship between the gut microbiota in infancy and patterns of growth in a cohort of preschool Canadian children. We highlight that rapid growth since birth is associated with bacteria shown in animal models to have a causative role in weight gain. Our findings support a novel avenue of research targeted on tangible interventions to reduce childhood obesity.
Collapse
Affiliation(s)
- Myrtha E Reyna
- Translational Medicine Program, Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Charisse Petersen
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Darlene L Y Dai
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Ruixue Dai
- Translational Medicine Program, Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Allan B Becker
- Section of Allergy and Immunology, Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
- Developmental Origins of Chronic Diseases in Children Network (DEVOTION), Children's Hospital, Winnipeg, MB, Canada
| | - Meghan B Azad
- Section of Allergy and Immunology, Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
- Developmental Origins of Chronic Diseases in Children Network (DEVOTION), Children's Hospital, Winnipeg, MB, Canada
- Department of Food and Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Kozeta Miliku
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Diana L Lefebvre
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Theo J Moraes
- Translational Medicine Program, Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| | | | - Rozlyn C T Boutin
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - B Brett Finlay
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Michael Smith Laboratories, UBC, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Elinor Simons
- Section of Allergy and Immunology, Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Anita L Kozyrskyj
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Wendy Lou
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Stuart E Turvey
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Padmaja Subbarao
- Translational Medicine Program, Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
15
|
Moving beyond descriptive studies: harnessing metabolomics to elucidate the molecular mechanisms underpinning host-microbiome phenotypes. Mucosal Immunol 2022; 15:1071-1084. [PMID: 35970917 DOI: 10.1038/s41385-022-00553-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/04/2022] [Accepted: 07/20/2022] [Indexed: 02/04/2023]
Abstract
Advances in technology and software have radically expanded the scope of metabolomics studies and allow us to monitor a broad transect of central carbon metabolism in routine studies. These increasingly sophisticated tools have shown that many human diseases are modulated by microbial metabolism. Despite this, it remains surprisingly difficult to move beyond these statistical associations and identify the specific molecular mechanisms that link dysbiosis to the progression of human disease. This difficulty stems from both the biological intricacies of host-microbiome dynamics as well as the analytical complexities inherent to microbiome metabolism research. The primary objective of this review is to examine the experimental and computational tools that can provide insights into the molecular mechanisms at work in host-microbiome interactions and to highlight the undeveloped frontiers that are currently holding back microbiome research from fully leveraging the benefits of modern metabolomics.
Collapse
|
16
|
Abdelhamid L, Luo XM. Diet and Hygiene in Modulating Autoimmunity During the Pandemic Era. Front Immunol 2022; 12:749774. [PMID: 35069526 PMCID: PMC8766844 DOI: 10.3389/fimmu.2021.749774] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022] Open
Abstract
The immune system is an efficiently toned machinery that discriminates between friends and foes for achieving both host defense and homeostasis. Deviation of immune recognition from foreign to self and/or long-lasting inflammatory responses results in the breakdown of tolerance. Meanwhile, educating the immune system and developing immunological memory are crucial for mounting defensive immune responses while protecting against autoimmunity. Still to elucidate is how diverse environmental factors could shape autoimmunity. The emergence of a world pandemic such as SARS-CoV-2 (COVID-19) not only threatens the more vulnerable individuals including those with autoimmune conditions but also promotes an unprecedented shift in people's dietary approaches while urging for extraordinary hygiene measures that likely contribute to the development or exacerbation of autoimmunity. Thus, there is an urgent need to understand how environmental factors modulate systemic autoimmunity to better mitigate the incidence and or severity of COVID-19 among the more vulnerable populations. Here, we discuss the effects of diet (macronutrients and micronutrients) and hygiene (the use of disinfectants) on autoimmunity with a focus on systemic lupus erythematosus.
Collapse
Affiliation(s)
- Leila Abdelhamid
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
- Department of Microbiology, College of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
17
|
Seo M, Lim H, Park M, Ha K, Kwon S, Shin J, Lee J, Hwang Y, Oh Y, Shin Y. Field study of the indoor environments for preventing the spread of the SARS-CoV-2 in Seoul. INDOOR AIR 2022; 32:e12959. [PMID: 34806218 PMCID: PMC9011577 DOI: 10.1111/ina.12959] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/14/2021] [Accepted: 11/07/2021] [Indexed: 05/30/2023]
Abstract
Despite the prolonged global spread of COVID-19, few studies have investigated the environmental influence on the spread of SARS-CoV-2 RNA with a metropolitan scale, particularly the detection of SARS-CoV-2 after disinfection at multi-use facilities. Between February 2020 and January 2021, 1,769 indoor air samples and object surfaces were tested at 231 multi-use facilities where confirmed cases were known to have occurred in Seoul, to determine whether SARS-CoV-2 RNA could be detected even after disinfection. Samples were collected by air scanner and swab pipette and detected by real-time RT-PCR. As a result, 10 (0.56%) positive samples were detected despite disinfection. The common environmental features of all 10 were surfaces that contained moisture and windowless buildings. With the aim of preventing the spread of COVID-19, from January to February 2021, we next conducted 643 preemptive tests before the outbreak of infections at 22 multi-use facilities where cluster infections were frequent. From these preemptive inspections, we obtained five (0.78%) positive results from two facilities, which enabled us to disinfect the buildings and give all the users a COVID-19 test. Based on the study purpose of finding and investigating cases of positive detection even after disinfection in the field through long-term environmental detection in a large city, our preemptive investigation results helped to prevent the spread of infectious diseases by confirming the potential existence of an asymptomatic patient.
Collapse
Affiliation(s)
- Minjeong Seo
- Seoul Metropolitan Government Research Institute of Public Health and EnvironmentSeoulSouth Korea
| | - Hakmyeong Lim
- Seoul Metropolitan Government Research Institute of Public Health and EnvironmentSeoulSouth Korea
| | - Myungkyu Park
- Seoul Metropolitan Government Research Institute of Public Health and EnvironmentSeoulSouth Korea
| | - Kwangtae Ha
- Seoul Metropolitan Government Research Institute of Public Health and EnvironmentSeoulSouth Korea
| | - Seungmi Kwon
- Seoul Metropolitan Government Research Institute of Public Health and EnvironmentSeoulSouth Korea
| | - Jinho Shin
- Seoul Metropolitan Government Research Institute of Public Health and EnvironmentSeoulSouth Korea
| | - Jaein Lee
- Seoul Metropolitan Government Research Institute of Public Health and EnvironmentSeoulSouth Korea
| | - Youngok Hwang
- Seoul Metropolitan Government Research Institute of Public Health and EnvironmentSeoulSouth Korea
| | - Younghee Oh
- Seoul Metropolitan Government Research Institute of Public Health and EnvironmentSeoulSouth Korea
| | - Yongseung Shin
- Seoul Metropolitan Government Research Institute of Public Health and EnvironmentSeoulSouth Korea
| |
Collapse
|
18
|
de Cuevillas B, Milagro FI, Tur JA, Gil-Campos M, de Miguel-Etayo P, Martínez JA, Navas-Carretero S. Fecal microbiota relationships with childhood obesity: A scoping comprehensive review. Obes Rev 2022; 23 Suppl 1:e13394. [PMID: 34913242 DOI: 10.1111/obr.13394] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 12/18/2022]
Abstract
Childhood obesity is a costly burden in most regions with relevant and adverse long-term health consequences in adult life. Several studies have associated excessive body weight with a specific profile of gut microbiota. Different factors related to fecal microorganism abundance seem to contribute to childhood obesity, such as gestational weight gain, perinatal diet, antibiotic administration to the mother and/or child, birth delivery, and feeding patterns, among others. This review reports and discusses diverse factors that affect the infant intestinal microbiota with putative or possible implications on the increase of the obesity childhood rates as well as microbiota shifts associated with excessive body weight in children.
Collapse
Affiliation(s)
- Begoña de Cuevillas
- Center for Nutrition Research, Department of Nutrition, Food Sciences and Physiology. School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Fermín I Milagro
- Center for Nutrition Research, Department of Nutrition, Food Sciences and Physiology. School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- IdiSNA, Health Research Institute of Navarra, Pamplona, Spain
| | - Josep A Tur
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands-IUNICS & IDISBA, Palma de Mallorca, Spain
| | - Mercedes Gil-Campos
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Metabolism and Investigation Unit, Reina Sofia University Hospital, Maimónides Institute of Biomedicine Research of Córdoba (IMIBIC), University of Córdoba, Córdoba, Spain
| | - Pilar de Miguel-Etayo
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Growth, Exercise, Nutrition and Development (GENUD) Research Group, Instituto Agroalimentario de Aragón (IA2), Universidad de Zaragoza. Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain
| | - J Alfredo Martínez
- Center for Nutrition Research, Department of Nutrition, Food Sciences and Physiology. School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- IdiSNA, Health Research Institute of Navarra, Pamplona, Spain
- Precision Nutrition Program, Research Institute on Food and Health Sciences IMDEA Food. CSIC-UAM, Madrid, Spain
| | - Santiago Navas-Carretero
- Center for Nutrition Research, Department of Nutrition, Food Sciences and Physiology. School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- IdiSNA, Health Research Institute of Navarra, Pamplona, Spain
| |
Collapse
|
19
|
Tjalvin G, Svanes Ø, Igland J, Bertelsen RJ, Benediktsdóttir B, Dharmage S, Forsberg B, Holm M, Janson C, Jõgi NO, Johannessen A, Malinovschi A, Pape K, Real FG, Sigsgaard T, Torén K, Vindenes HK, Zock JP, Schlünssen V, Svanes C. Maternal preconception occupational exposure to cleaning products and disinfectants and offspring asthma. J Allergy Clin Immunol 2021; 149:422-431.e5. [PMID: 34674855 DOI: 10.1016/j.jaci.2021.08.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/08/2021] [Accepted: 08/31/2021] [Indexed: 02/02/2023]
Abstract
BACKGROUND Emerging research suggests health effects in offspring after parental chemical exposures before conception. Many future mothers are exposed to potent chemicals at work, but potential offspring health effects are hardly investigated. OBJECTIVE We sought to investigate childhood asthma in relation to mother's occupational exposure to cleaning products and disinfectants before conception. METHODS The multicenter Respiratory Health In Northern Europe/Respiratory Health In Northern Europe, Spain and Australia generation study investigated asthma and wheeze starting at age less than 10 years in 3318 mother-offspring pairs. From an asthma-specific Job-Exposure Matrix and mothers' occupational history, we defined maternal occupational exposure to indoor cleaning agents (cleaning products/detergents and disinfectants) starting before conception, in the 2-year period around conception and pregnancy, or after birth. Never-employed mothers were excluded. Exposed groups include cleaners, health care workers, cooks, and so forth. Associations were analyzed using mixed-effects logistic regression and ordinary logistic regression with clustered robust SEs and adjustment for maternal education. RESULTS Maternal occupational exposure to indoor cleaning starting preconception and continuing (n = 610) was associated with offspring's childhood asthma: odds ratio 1.56 (95% CI, 1.05-2.31), childhood asthma with nasal allergies: 1.77 (1.13-2.77), and childhood wheeze and/or asthma: 1.71 (95% CI, 1.19-2.44). Exposure starting around conception and pregnancy (n = 77) was associated with increased childhood wheeze and/or asthma: 2.25 (95% CI, 1.03-4.91). Exposure starting after birth was not associated with asthma outcomes (1.13 [95% CI, 0.71-1.80], 1.15 [95% CI, 0.67-1.97], 1.08 [95% CI, 0.69-1.67]). CONCLUSIONS Mother's occupational exposure to indoor cleaning agents starting before conception, or around conception and pregnancy, was associated with more childhood asthma and wheeze in offspring. Considering potential implications for vast numbers of women in childbearing age using cleaning agents, and their children, further research is imperative.
Collapse
Affiliation(s)
- Gro Tjalvin
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway; Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway.
| | - Øistein Svanes
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Jannicke Igland
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway; Department of Health and Caring Sciences, Faculty of Health and Social Sciences, Western Norway University of Applied Sciences, Bergen, Norway
| | - Randi Jacobsen Bertelsen
- Department of Clinical Science, University of Bergen, Bergen, Norway; Oral Health Center of Expertise in Western Norway, Bergen, Norway
| | - Bryndís Benediktsdóttir
- Medical Faculty, University of Iceland, Reykjavík, Iceland; Department of Sleep, Landspitali University Hospital Reykjavík, Reykjavík, Iceland
| | - Shyamali Dharmage
- Allergy and Lung Health Unit, University of Melbourne, Melbourne, Australia
| | - Bertil Forsberg
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Mathias Holm
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Christer Janson
- Department of Medical Sciences: Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Nils Oskar Jõgi
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway; Tartu University Lung Clinic, Tartu, Estonia
| | - Ane Johannessen
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Andrei Malinovschi
- Department of Medical Sciences: Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Kathrine Pape
- National Research Centre for the Working Environment, Aarhus, Denmark; Department of Public Health, Aarhus University, Environment, Work and Health, Danish Ramazzini Centre, Aarhus, Denmark
| | - Francisco Gomez Real
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Torben Sigsgaard
- Department of Public Health, Aarhus University, Environment, Work and Health, Danish Ramazzini Centre, Aarhus, Denmark
| | - Kjell Torén
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hilde Kristin Vindenes
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Jan-Paul Zock
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Vivi Schlünssen
- National Research Centre for the Working Environment, Aarhus, Denmark; Department of Public Health, Aarhus University, Environment, Work and Health, Danish Ramazzini Centre, Aarhus, Denmark
| | - Cecilie Svanes
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway; Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| |
Collapse
|
20
|
A Study on the Behavior Patterns of Liquid Aerosols Using Disinfectant Chloromethylisothiazolinone/Methylisothiazolinone Solution. Molecules 2021; 26:molecules26195725. [PMID: 34641269 PMCID: PMC8510451 DOI: 10.3390/molecules26195725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/10/2021] [Accepted: 09/17/2021] [Indexed: 11/17/2022] Open
Abstract
This study evaluates the behavioral characteristics of components (methylisothiazolinone (MIT) and chloromethylisothiazolinone (CMIT)) contained in disinfectant solutions when they convert to liquid aerosols. The analytical method for MIT and CMIT quantitation was established and optimized using sorbent tube/thermal desorber-gas chromatography-mass spectrometry system; their behavioral characteristics are discussed using the quantitative results of these aerosols under different liquid aerosol generation conditions. MIT and CMIT showed different behavioral characteristics depending on the aerosol mass concentration and sampling time (sampling volume). When the disinfectant solution was initially aerosolized, MIT and CMIT were primarily collected on glass filter (MIT = 91.8 ± 10.6% and CMIT = 90.6 ± 5.18%), although when the generation and filter sampling volumes of the aerosols increased to 30 L, the relative proportions collected on the filter decreased (MIT = 79.0 ± 12.0% and CMIT = 39.7 ± 8.35%). Although MIT and CMIT had relatively high vapor pressure, in liquid aerosolized state, they primarily accumulated on the filter and exhibited particulate behavior. Their relative proportions in the aerosol were different from those in disinfectant solution. In the aerosol with mass concentration of ≤5 mg m-3, the relative proportion deviations of MIT and CMIT were large; when the mass concentration of the aerosol increased, their relative proportions constantly converged at a lower level than those in the disinfectant solution. Hence, it can be concluded that the behavioral characteristics and relative proportions need to be considered to perform the quantitative analysis of the liquid aerosols and evaluate various toxic effects using the quantitative data.
Collapse
|
21
|
Yep A, Nation JM, Moreno R, Reyes H, Torres A, De Smet C. Nuestra Ciencia: Transforming microbiology for Spanish-speaking elementary and college students. Integr Comp Biol 2021; 61:1066-1077. [PMID: 34050752 DOI: 10.1093/icb/icab117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
This forward-looking perspective describes the university-elementary bilingual partnership program Nuestra Ciencia. This program aims to simultaneously tackle two parallel sets of challenges, the first related to recruitment and retention of Latinx into STEM fields, and the second related to generalized microbiology misconceptions. Latinxs are severely underrepresented in STEM fields, in part because they face systemic barriers and typically arrive at college with a weaker science foundation from their K-12 education and thus are less likely to be drawn to STEM majors. Beyond grappling with the science content, Latinx students reach college with assumptions about who belongs in science practices and professions, which in turn negatively affect their representation in STEM careers. Misconceptions also plague microbiology education, and most students reach college with deep-seated yet inaccurate ideas about the microbial world, such as the ways in which vaccines and antibiotics work. Unfortunately, lack of microbiology literacy has a direct impact on personal choices that can affect individuals but also the success of public health and environmental policies. Nuestra Ciencia addresses both sets of problems, as we work with interdisciplinary groups of undergraduates to develop engaging experiments for elementary classrooms that illustrate microbiology concepts, and then visit bilingual classrooms to lead the experiments in Spanish. Lessons have accompanying resources in Spanish and English for teachers and students, including background information, handouts, and assessment tools. In this manuscript, we outline the background, goals and components of the program, review activities developed for elementary students, and share potential impact and lessons learned. Additionally, we explore future directions and outreach activities, especially in relation to online learning.
Collapse
Affiliation(s)
- Alejandra Yep
- Associate Professor, Biological Sciences, California Polytechnic State University, San Luis Obispo, CA
| | - Jasmine McBeath Nation
- Assistant Professor, Liberal Studies, California Polytechnic State University, San Luis Obispo, CA 93407, 805-756-2990
| | - Ruby Moreno
- BEACoN Undergraduate Researcher, Department of Biomedical Engineering, California Polytechnic State University, San Luis Obispo, CA
| | - Hector Reyes
- Undergraduate Research Assistant, Department of Psychology, California Polytechnic State University, San Luis Obispo, CA
| | - Adrian Torres
- BEACoN Undergraduate Researcher, Department of Political Science, California Polytechnic State University, San Luis Obispo, CA
| | - Chanel De Smet
- Undergraduate Research Assistant, Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA
| |
Collapse
|
22
|
Kampf G, Jatzwauk L. [Is Disinfection of Public Surfaces useful for the Prevention of SARS-CoV-2 Infections?]. DAS GESUNDHEITSWESEN 2021; 83:180-185. [PMID: 33540430 PMCID: PMC8043670 DOI: 10.1055/a-1335-4549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Measures to control SARS-CoV-2 often include the regular disinfection of public surfaces. The frequency of SARS-CoV-2 detection on surfaces in the surrounding of confirmed cases was evaluated in this systematic review. Overall, 26 studies showed 0 and 100% rates of contamination with SARS-CoV-2 RNA on surfaces in the surrounding of patients. Seven studies with at least 100 samples mostly showed detection rates between 1.4 and 19%. Two other studies did not detect infectious SARS-CoV-2 on any surface. Similar results were obtained from surfaces in the surrounding of confirmed SARS- and influenza-patients. A contamination of public surfaces with infectious virus is considerably less likely because there are much less potential viral spreaders around a surface, the contact time between a person and the surface is much shorter, and the asymptomatic carriers typically have no symptoms. In addition, a hand contact with a contaminated surface transfers only a small part of the viral load. A simple cleaning reduces the number of infectious viruses already by 2 log10-steps. That is why public surfaces should in general be cleaned because the wide use of biocidal agents for surface disinfection further increases the microbial selection pressure without an expectable health benefit.
Collapse
Affiliation(s)
- Günter Kampf
- Institut für Hygiene und Umweltmedizin, Universitätsmedizin Greifswald, Greifswald, Deutschland
| | - Lutz Jatzwauk
- Krankenhaushygiene, Universitätsklinikum Carl Gustav Carus, Dresden, Deutschland
| |
Collapse
|
23
|
Deehan EC, Colin-Ramirez E, Triador L, Madsen KL, Prado CM, Field CJ, Ball GDC, Tan Q, Orsso C, Dinu I, Pakseresht M, Rubin D, Sharma AM, Tun H, Walter J, Newgard CB, Freemark M, Wine E, Haqq AM. Efficacy of metformin and fermentable fiber combination therapy in adolescents with severe obesity and insulin resistance: study protocol for a double-blind randomized controlled trial. Trials 2021; 22:148. [PMID: 33596993 PMCID: PMC7890810 DOI: 10.1186/s13063-021-05060-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/20/2021] [Indexed: 02/08/2023] Open
Abstract
Background Accumulating evidence suggests that the metabolic effects of metformin and fermentable fibers are mediated, in part, through diverging or overlapping effects on the composition and metabolic functions of the gut microbiome. Pre-clinical animal models have established that the addition of fiber to metformin monotherapy improves glucose tolerance. However, possible synergistic effects of combination therapy (metformin plus fiber) have not been investigated in humans. Moreover, the underlying mechanisms of synergy have yet to be elucidated. The aim of this study is to compare in adolescents with obesity the metabolic effects of metformin and fermentable fibers in combination with those of metformin or fiber alone. We will also determine if therapeutic responses correlate with compositional and functional features of the gut microbiome. Methods This is a parallel three-armed, double-blinded, randomized controlled trial. Adolescents (aged 12–18 years) with obesity, insulin resistance (IR), and a family history of type 2 diabetes mellitus (T2DM) will receive either metformin (850 mg p.o. twice/day), fermentable fibers (35 g/day), or a combination of metformin plus fiber for 12 months. Participants will be seen at baseline, 3, 6, and 12 months, with a phone follow-up at 1 and 9 months. Primary and secondary outcomes will be assessed at baseline, 6, and 12 months. The primary outcome is change in IR estimated by homeostatic model assessment of IR; key secondary outcomes include changes in the Matsuda index, oral disposition index, body mass index z-score, and fat mass to fat-free mass ratio. To gain mechanistic insight, endpoints that reflect host-microbiota interactions will also be assessed: obesity-related immune, metabolic, and satiety markers; humoral metabolites; and fecal microbiota composition, short-chain fatty acids, and bile acids. Discussion This study will compare the potential metabolic benefits of fiber with those of metformin in adolescents with obesity, determine if metformin and fiber act synergistically to improve IR, and elucidate whether the metabolic benefits of metformin and fiber associate with changes in fecal microbiota composition and the output of health-related metabolites. This study will provide insight into the potential role of the gut microbiome as a target for enhancing the therapeutic efficacy of emerging treatments for T2DM prevention. Trial registration ClinicalTrials.gov NCT04578652. Registered on 8 October 2020. Supplementary Information The online version contains supplementary material available at 10.1186/s13063-021-05060-8.
Collapse
Affiliation(s)
- Edward C Deehan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | | | - Lucila Triador
- Department of Pediatrics, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Karen L Madsen
- Department of Medicine, University of Alberta, Edmonton, T6G 2C2, AB, Canada
| | - Carla M Prado
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Geoff D C Ball
- Department of Pediatrics, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Qiming Tan
- Department of Pediatrics, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Camila Orsso
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Irina Dinu
- School of Public Health, University of Alberta, Edmonton, T6G 1C9, AB, Canada
| | - Mohammadreza Pakseresht
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | - Daniela Rubin
- California State University Fullerton, Fullerton, USA
| | - Arya M Sharma
- Department of Medicine, University of Alberta, Edmonton, T6G 2C2, AB, Canada
| | - Hein Tun
- University of Hong Kong School of Public Health, Hong Kong, China
| | - Jens Walter
- DNational University of Ireland University College Cork, University College Cork, Cork, Ireland
| | | | - Michael Freemark
- Duke University Medical Center, Duke University Hospital, Durham, NC, USA
| | - Eytan Wine
- Department of Pediatrics and Physiology, University of Alberta, Edmonton, T6G 1C9, BA, Canada
| | - Andrea M Haqq
- Department of Pediatrics, University of Alberta, Edmonton, T6G 2E1, AB, Canada.
| |
Collapse
|
24
|
Obesity, Early Life Gut Microbiota, and Antibiotics. Microorganisms 2021; 9:microorganisms9020413. [PMID: 33671180 PMCID: PMC7922584 DOI: 10.3390/microorganisms9020413] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/09/2021] [Accepted: 02/15/2021] [Indexed: 02/07/2023] Open
Abstract
Obesity is a major public health problem that continues to be one of the leading risk factors for premature death. Early life is a critical period of time when the gut microbiota and host metabolism are developing in tandem and significantly contribute to long-term health outcomes. Dysbiosis of the gut microbiota, particularly in early life, can have detrimental effects on host health and increase the susceptibility of developing obesity later in life. Antibiotics are an essential lifesaving treatment; however, their use in early life may not be without risk. Antibiotics are a leading cause of intestinal dysbiosis, and early life administration is associated with obesity risk. The following review explores the relevant literature that simultaneously examines antibiotic-induced dysbiosis and obesity risk. Current evidence suggests that disruptions to the composition and maturation of the gut microbiota caused by antibiotic use in early life are a key mechanism linking the association between antibiotics and obesity. Without compromising clinical practice, increased consideration of the long-term adverse effects of antibiotic treatment on host health, particularly when used in early life is warranted. Novel adjunct interventions should be investigated (e.g., prebiotics) to help mitigate metabolic risk when antibiotic treatment is clinically necessary.
Collapse
|
25
|
Finlay BB, Amato KR, Azad M, Blaser MJ, Bosch TCG, Chu H, Dominguez-Bello MG, Ehrlich SD, Elinav E, Geva-Zatorsky N, Gros P, Guillemin K, Keck F, Korem T, McFall-Ngai MJ, Melby MK, Nichter M, Pettersson S, Poinar H, Rees T, Tropini C, Zhao L, Giles-Vernick T. The hygiene hypothesis, the COVID pandemic, and consequences for the human microbiome. Proc Natl Acad Sci U S A 2021; 118:e2010217118. [PMID: 33472859 PMCID: PMC8017729 DOI: 10.1073/pnas.2010217118] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Indexed: 12/12/2022] Open
Abstract
The COVID-19 pandemic has the potential to affect the human microbiome in infected and uninfected individuals, having a substantial impact on human health over the long term. This pandemic intersects with a decades-long decline in microbial diversity and ancestral microbes due to hygiene, antibiotics, and urban living (the hygiene hypothesis). High-risk groups succumbing to COVID-19 include those with preexisting conditions, such as diabetes and obesity, which are also associated with microbiome abnormalities. Current pandemic control measures and practices will have broad, uneven, and potentially long-term effects for the human microbiome across the planet, given the implementation of physical separation, extensive hygiene, travel barriers, and other measures that influence overall microbial loss and inability for reinoculation. Although much remains uncertain or unknown about the virus and its consequences, implementing pandemic control practices could significantly affect the microbiome. In this Perspective, we explore many facets of COVID-19-induced societal changes and their possible effects on the microbiome, and discuss current and future challenges regarding the interplay between this pandemic and the microbiome. Recent recognition of the microbiome's influence on human health makes it critical to consider both how the microbiome, shaped by biosocial processes, affects susceptibility to the coronavirus and, conversely, how COVID-19 disease and prevention measures may affect the microbiome. This knowledge may prove key in prevention and treatment, and long-term biological and social outcomes of this pandemic.
Collapse
Affiliation(s)
- B Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada;
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
| | - Katherine R Amato
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Department of Anthropology, Northwestern University, Evanston, IL 60208
| | - Meghan Azad
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Manitoba Interdisciplinary Lactation Centre, Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Martin J Blaser
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Center for Advanced Biotechnology and Medicine at Rutgers Biomedical and Health Sciences, Rutgers University, Piscataway, NJ 08854-8021
| | - Thomas C G Bosch
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Zoologisches Institut, University of Kiel, 24118 Kiel, Germany
| | - Hiutung Chu
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Department of Pathology, University of California San Diego, La Jolla, CA 92093
| | - Maria Gloria Dominguez-Bello
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ 08901
| | - Stanislav Dusko Ehrlich
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Metagenopolis Unit, French National Institute for Agricultural Research, 78350 Jouy-en-Josas, France
| | - Eran Elinav
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Department of Immunology, Weizmann Institute of Science, Rehovot 761000, Israel
- Cancer-Microbiome Division, Deutsches Krebsforschungszentrum, 69120 Heidelberg, Germany
| | - Naama Geva-Zatorsky
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Technion Integrated Cancer Center, Department of Cell Biology and Cancer Science, Technion-Israel Institute of Technology, Haifa 3525433, Israel
| | - Philippe Gros
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Karen Guillemin
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403
| | - Frédéric Keck
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Centre National de la Recherche Scientifique, 75016 Paris, France
- Laboratoire d'Anthropologie Sociale, Collège de France, 75005 Paris, France
| | - Tal Korem
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Department of Systems Biology, Irving Cancer Research Center, Columbia University, New York, NY 10032
- Department of Obstetrics and Gynecology, Irving Cancer Research Center, Columbia University, New York, NY 10032
| | - Margaret J McFall-Ngai
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Pacific Biosciences Research Center, University of Hawai'i at Manoa, Honolulu, HI 96822
| | - Melissa K Melby
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Department of Anthropology, University of Delaware, Newark, DE 19711
| | - Mark Nichter
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Department of Anthropology, University of Arizona, Tucson, AZ 85721
| | - Sven Pettersson
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Lee Kong Chian School of Medicine, Nanyang Technological University, 637715 Singapore
| | - Hendrik Poinar
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Department of Anthropology, McMaster University, Hamilton, ON L8S 4M4, Canada
| | - Tobias Rees
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Transformations of the Human Program, Berggruen Institute, Los Angeles, CA 90013
| | - Carolina Tropini
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Liping Zhao
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ 08901
| | - Tamara Giles-Vernick
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada;
- Anthropology & Ecology of Disease Emergence, Institut Pasteur, 75015 Paris, France
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW Evidence for adverse respiratory effects of occupational exposure to disinfectants and cleaning products (DCPs) has grown in the last two decades. The relationship between DCPs and asthma is well documented but questions remain regarding specific causal agents. Beyond asthma, associations between DCPs and COPD or chronic rhinitis are plausible and have been examined recently. The purpose of this review is to summarize recent advances on the effect of occupational exposure to DCP and chronic airway diseases. RECENT FINDINGS Recent epidemiological studies have often focused on healthcare workers and are characterized by efforts to improve assessment of exposure to specific DCPs. Despite increasing knowledge on the effect of DCPs on asthma, the burden of work-related asthma caused by DCPs has not decreased in the past decade, emphasizing the need to strengthen prevention efforts. Novel data suggest an association between occupational exposure to DCPs and other chronic airway diseases, such as rhinitis, COPD, and poor lung function. SUMMARY Epidemiological and experimental data showed that many chemicals contained in DCPs are likely to cause airway damage, indicating that prevention strategies should target multiple products. Further research is needed to evaluate the impact of DCP exposure on occupational airway diseases beyond asthma.
Collapse
|
27
|
Can we prevent allergic disease? Understanding the links between the early life microbiome and allergic diseases of childhood. Curr Opin Pediatr 2020; 32:790-797. [PMID: 33027216 DOI: 10.1097/mop.0000000000000956] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW The microbiome and immune system are intrinsically linked, and during infancy these crucial biological systems undergo a concurrent and expansive maturation process. As these maturation processes progress, some children develop a sequence of IgE-mediated immune disorders termed the 'Allergic March', and unfortunately the prevalence of these lifelong and burdensome allergic conditions has increased over the past half century. Our current treatment strategies are unable to prevent or cure components of the Allergic March. However, recent discoveries have enhanced our mechanistic understanding of early-life microbiota-immune interactions with exciting implications for preventing these allergic disorders. RECENT FINDINGS The current review will detail recent literature regarding perinatal factors (e.g. birth mode, antibiotic exposure, breastmilk seeding of the microbiota, built environment) that shape the infant gut microbiota composition. Furthermore, we will discuss new findings that have highlighted immune cells which are particularly sensitive to microbial influences in utero and during the early-life window of development. SUMMARY As our understanding of the dynamic relationship between the developing infant microbiota and immune system grows, a priority toward preserving critical early-life interactions may provide life-long protection to these diseases in the future.
Collapse
|
28
|
Drall KM, Field CJ, Haqq AM, de Souza RJ, Tun HM, Morales-Lizcano NP, Konya TB, Guttman DS, Azad MB, Becker AB, Lefebvre DL, Mandhane PJ, Moraes TJ, Sears MR, Turvey SE, Subbarao P, Scott JA, Kozyrskyj AL. Vitamin D supplementation in pregnancy and early infancy in relation to gut microbiota composition and C. difficile colonization: implications for viral respiratory infections. Gut Microbes 2020; 12:1799734. [PMID: 32779963 PMCID: PMC7524344 DOI: 10.1080/19490976.2020.1799734] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In Canada and the US, the infant diet is supplemented with vitamin D via supplement drops or formula. Pregnant and nursing mothers often take vitamin D supplements. Since little is known about the impact of this supplementation on infant gut microbiota, we undertook a study to determine the association between maternal and infant vitamin D supplementation, infant gut microbiota composition and Clostridioides difficile colonization in 1,157 mother-infant pairs of the CHILD (Canadian Healthy Infant Longitudinal Development) Cohort Study over 2009-2012. Logistic and MaAsLin regression were employed to assess associations between vitamin D supplementation, and C. difficile colonization, or other gut microbiota, respectively. Sixty-five percent of infants received a vitamin D supplement. Among all infants, infant vitamin D supplementation was associated with a lower abundance of genus Megamonas (q = 0.01) in gut microbiota. Among those exclusively breastfed, maternal prenatal supplementation was associated with lower abundance of Bilophila (q = 0.01) and of Lachnospiraceae (q = 0.02) but higher abundance of Haemophilus (q = 0.02). There were no differences in microbiota composition with vitamin D supplementation among partially and not breastfed infants. Neither infant nor maternal vitamin D supplementation were associated with C. difficile colonization, after adjusting for breastfeeding status and other factors. However, maternal consumption of vitamin-D fortified milk reduced the likelihood of C. difficile colonization in infants (adjustedOR: 0.40, 95% CI: 0.19-0.82). The impact of this compositional difference on later childhood health, especially defense against viral respiratory infection, may go beyond the expected effects of vitamin D supplements and remains to be ascertained.
Collapse
Affiliation(s)
- Kelsea M. Drall
- Departments of Pediatrics, Obstetrics & Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Catherine J. Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Andrea M. Haqq
- Departments of Pediatrics, Obstetrics & Gynecology, University of Alberta, Edmonton, AB, Canada,Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Russell J. de Souza
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada,Population Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Hein M. Tun
- Departments of Pediatrics, Obstetrics & Gynecology, University of Alberta, Edmonton, AB, Canada,HKU-Pasteur Research Pole, School of Public Health, Hong Kong University, Hong Kong SAR, China
| | | | - Theodore B. Konya
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - David S. Guttman
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada,Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, ON, Canada
| | - Meghan B. Azad
- Department of Pediatrics & Child Health, Children’s Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Allan B. Becker
- Department of Pediatrics & Child Health, Children’s Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | | | - Piush J. Mandhane
- Departments of Pediatrics, Obstetrics & Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Theo J. Moraes
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ONCanada
| | - Malcolm R. Sears
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Stuart E. Turvey
- Department of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Padmaja Subbarao
- Department of Medicine, McMaster University, Hamilton, ON, Canada,Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ONCanada
| | - James A. Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Anita L Kozyrskyj
- Departments of Pediatrics, Obstetrics & Gynecology, University of Alberta, Edmonton, AB, Canada,School of Public Health, University of Alberta, Edmonton, Canada,CONTACT : Anita L Kozyrskyj 3-527 Edmonton Clinic Health Academy, Edmonton, ABT6G 1C9, Canada
| |
Collapse
|
29
|
Parks J, Takaro TK. Exposure to cleaning products and childhood asthma: more than just a link? Expert Rev Respir Med 2020; 14:1185-1188. [PMID: 32990115 DOI: 10.1080/17476348.2020.1813572] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Jaclyn Parks
- Faculty of Health Sciences, Simon Fraser University , Burnaby, BC, Canada
| | - Tim K Takaro
- Faculty of Health Sciences, Simon Fraser University , Burnaby, BC, Canada
| |
Collapse
|
30
|
Developmental Origins of Health and Disease: Impact of environmental dust exposure in modulating microbiome and its association with non-communicable diseases. J Dev Orig Health Dis 2020; 11:545-556. [PMID: 32536356 DOI: 10.1017/s2040174420000549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Non-communicable diseases (NCDs) including obesity, diabetes, and allergy are chronic, multi-factorial conditions that are affected by both genetic and environmental factors. Over the last decade, the microbiome has emerged as a possible contributor to the pathogenesis of NCDs. Microbiome profiles were altered in patients with NCDs, and shift in microbial communities was associated with improvement in these health conditions. Since the genetic component of these diseases cannot be altered, the ability to manipulate the microbiome holds great promise for design of novel therapies in the prevention and treatment of NCDs. Together, the Developmental Origins of Health and Disease concept and the microbial hypothesis propose that early life exposure to environmental stimuli will alter the development and composition of the human microbiome, resulting in health consequences. Recent studies indicated that the environment we are exposed to in early life is instrumental in shaping robust immune development, possibly through modulation of the human microbiome (skin, airway, and gut). Despite much research into human microbiome, the origin of their constituent microbiota remains unclear. Dust (also known as particulate matter) is a key determinant of poor air quality in the modern urban environment. It is ubiquitous and serves as a major source and reservoir of microbial communities that modulates the human microbiome, contributing to health and disease. There are evidence that reported significant associations between environmental dust and NCDs. In this review, we will focus on the impact of dust exposure in shaping the human microbiome and its possible contribution to the development of NCDs.
Collapse
|
31
|
Cowan CSM, Dinan TG, Cryan JF. Annual Research Review: Critical windows - the microbiota-gut-brain axis in neurocognitive development. J Child Psychol Psychiatry 2020; 61:353-371. [PMID: 31773737 DOI: 10.1111/jcpp.13156] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/26/2019] [Accepted: 10/09/2019] [Indexed: 02/06/2023]
Abstract
The gut microbiota is a vast, complex, and fascinating ecosystem of microorganisms that resides in the human gastrointestinal tract. As an integral part of the microbiota-gut-brain axis, it is now being recognized that the microbiota is a modulator of brain and behavior, across species. Intriguingly, periods of change in the microbiota coincide with the development of other body systems and particularly the brain. We hypothesize that these times of parallel development are biologically relevant, corresponding to 'sensitive periods' or 'critical windows' in the development of the microbiota-gut-brain axis. Specifically, signals from the microbiota during these periods are hypothesized to be crucial for establishing appropriate communication along the axis throughout the life span. In other words, the microbiota is hypothesized to act like an expected input to calibrate the development of the microbiota-gut-brain axis. The absence or disruption of the microbiota during specific developmental windows would therefore be expected to have a disproportionate effect on specific functions or potentially for regulation of the system as a whole. Evidence for microbial modulation of neurocognitive development and neurodevelopmental risk is discussed in light of this hypothesis, finishing with a focus on the challenges that lay ahead for the future study of the microbiota-gut-brain axis during development.
Collapse
Affiliation(s)
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
32
|
Parks J, McCandless L, Dharma C, Brook J, Turvey SE, Mandhane P, Becker AB, Kozyrskyj AL, Azad MB, Moraes TJ, Lefebvre DL, Sears MR, Subbarao P, Scott J, Takaro TK. Association of use of cleaning products with respiratory health in a Canadian birth cohort. CMAJ 2020; 192:E154-E161. [PMID: 32071106 PMCID: PMC7030878 DOI: 10.1503/cmaj.190819] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Comprehensive longitudinal studies are important for understanding the complex risk factors, pathways, exposures and interactions that lead to the development and persistence of asthma. We aimed to examine associations between use of household cleaning products in early life and childhood respiratory and allergic disease using data from the Canadian Healthy Infant Longitudinal Development (CHILD) Cohort Study. METHODS We summed responses from parental questionnaires that indicated the frequency of use of 26 household cleaning products in the homes of 2022 children from this birth cohort when they were 3-4 months of age to create a cumulative Frequency of Use Score (FUS). We used multivariable logistic regression models to assess whether frequent compared with less frequent use was associated with recurrent wheeze, atopy or asthma diagnosis, as defined by the questionnaire and clinical assessments at age 3 years. Data were collected between 2008 and 2015. RESULTS Children in homes with a higher frequency of use of cleaning products in infancy, as determined by an interquartile range increase, had higher odds of recurrent wheeze (adjusted odds ratio [OR] 1.35, 95% confidence interval [CI] 1.11-1.64), recurrent wheeze with atopy (adjusted OR 1.49, 95% CI 1.02-2.16) and asthma diagnosis (adjusted OR 1.37, 95% CI 1.09-1.70), but no increase in the odds of atopy at age 3 years (adjusted OR 1.14, 95% CI 0.96-1.35). Compared with the lowest tertile of FUS exposure, infants in the highest tertile had higher odds of acquiring asthma. Stratification of the results showed that females had higher ORs than males for all outcomes, although the p values for this sex difference did not reach statistical significance. INTERPRETATION Frequent use of household cleaning products in early life was associated with an increased risk for childhood wheeze and asthma but not atopy at age 3 years. Our findings add to the understanding of how early life exposures to cleaning products may be associated with the development of allergic airway disease and help to identify household behaviours as a potential area for intervention.
Collapse
Affiliation(s)
- Jaclyn Parks
- Faculty of Health Sciences (Parks, McCandless, Takaro), Simon Fraser University, Vancouver, BC; Department of Medicine (Dharma, Lefebvre, Sears), McMaster University, Hamilton, Ont.; Dalla Lana School of Public Health (Brook, Scott), University of Toronto, Toronto, Ont.; Department of Pediatrics (Turvey), University of British Columbia, Vancouver, BC; Department of Pediatrics (Mandhane, Kozyrskyj), University of Alberta, Edmonton, Alta.; Department of Pediatrics & Child Health (Becker, Azad), University of Manitoba, Winnipeg, Man.; Department of Pediatrics (Moraes, Subbarao), University of Toronto and Hospital for Sick Children, Toronto, Ont
| | - Lawrence McCandless
- Faculty of Health Sciences (Parks, McCandless, Takaro), Simon Fraser University, Vancouver, BC; Department of Medicine (Dharma, Lefebvre, Sears), McMaster University, Hamilton, Ont.; Dalla Lana School of Public Health (Brook, Scott), University of Toronto, Toronto, Ont.; Department of Pediatrics (Turvey), University of British Columbia, Vancouver, BC; Department of Pediatrics (Mandhane, Kozyrskyj), University of Alberta, Edmonton, Alta.; Department of Pediatrics & Child Health (Becker, Azad), University of Manitoba, Winnipeg, Man.; Department of Pediatrics (Moraes, Subbarao), University of Toronto and Hospital for Sick Children, Toronto, Ont
| | - Christoffer Dharma
- Faculty of Health Sciences (Parks, McCandless, Takaro), Simon Fraser University, Vancouver, BC; Department of Medicine (Dharma, Lefebvre, Sears), McMaster University, Hamilton, Ont.; Dalla Lana School of Public Health (Brook, Scott), University of Toronto, Toronto, Ont.; Department of Pediatrics (Turvey), University of British Columbia, Vancouver, BC; Department of Pediatrics (Mandhane, Kozyrskyj), University of Alberta, Edmonton, Alta.; Department of Pediatrics & Child Health (Becker, Azad), University of Manitoba, Winnipeg, Man.; Department of Pediatrics (Moraes, Subbarao), University of Toronto and Hospital for Sick Children, Toronto, Ont
| | - Jeffrey Brook
- Faculty of Health Sciences (Parks, McCandless, Takaro), Simon Fraser University, Vancouver, BC; Department of Medicine (Dharma, Lefebvre, Sears), McMaster University, Hamilton, Ont.; Dalla Lana School of Public Health (Brook, Scott), University of Toronto, Toronto, Ont.; Department of Pediatrics (Turvey), University of British Columbia, Vancouver, BC; Department of Pediatrics (Mandhane, Kozyrskyj), University of Alberta, Edmonton, Alta.; Department of Pediatrics & Child Health (Becker, Azad), University of Manitoba, Winnipeg, Man.; Department of Pediatrics (Moraes, Subbarao), University of Toronto and Hospital for Sick Children, Toronto, Ont
| | - Stuart E Turvey
- Faculty of Health Sciences (Parks, McCandless, Takaro), Simon Fraser University, Vancouver, BC; Department of Medicine (Dharma, Lefebvre, Sears), McMaster University, Hamilton, Ont.; Dalla Lana School of Public Health (Brook, Scott), University of Toronto, Toronto, Ont.; Department of Pediatrics (Turvey), University of British Columbia, Vancouver, BC; Department of Pediatrics (Mandhane, Kozyrskyj), University of Alberta, Edmonton, Alta.; Department of Pediatrics & Child Health (Becker, Azad), University of Manitoba, Winnipeg, Man.; Department of Pediatrics (Moraes, Subbarao), University of Toronto and Hospital for Sick Children, Toronto, Ont
| | - Piush Mandhane
- Faculty of Health Sciences (Parks, McCandless, Takaro), Simon Fraser University, Vancouver, BC; Department of Medicine (Dharma, Lefebvre, Sears), McMaster University, Hamilton, Ont.; Dalla Lana School of Public Health (Brook, Scott), University of Toronto, Toronto, Ont.; Department of Pediatrics (Turvey), University of British Columbia, Vancouver, BC; Department of Pediatrics (Mandhane, Kozyrskyj), University of Alberta, Edmonton, Alta.; Department of Pediatrics & Child Health (Becker, Azad), University of Manitoba, Winnipeg, Man.; Department of Pediatrics (Moraes, Subbarao), University of Toronto and Hospital for Sick Children, Toronto, Ont
| | - Allan B Becker
- Faculty of Health Sciences (Parks, McCandless, Takaro), Simon Fraser University, Vancouver, BC; Department of Medicine (Dharma, Lefebvre, Sears), McMaster University, Hamilton, Ont.; Dalla Lana School of Public Health (Brook, Scott), University of Toronto, Toronto, Ont.; Department of Pediatrics (Turvey), University of British Columbia, Vancouver, BC; Department of Pediatrics (Mandhane, Kozyrskyj), University of Alberta, Edmonton, Alta.; Department of Pediatrics & Child Health (Becker, Azad), University of Manitoba, Winnipeg, Man.; Department of Pediatrics (Moraes, Subbarao), University of Toronto and Hospital for Sick Children, Toronto, Ont
| | - Anita L Kozyrskyj
- Faculty of Health Sciences (Parks, McCandless, Takaro), Simon Fraser University, Vancouver, BC; Department of Medicine (Dharma, Lefebvre, Sears), McMaster University, Hamilton, Ont.; Dalla Lana School of Public Health (Brook, Scott), University of Toronto, Toronto, Ont.; Department of Pediatrics (Turvey), University of British Columbia, Vancouver, BC; Department of Pediatrics (Mandhane, Kozyrskyj), University of Alberta, Edmonton, Alta.; Department of Pediatrics & Child Health (Becker, Azad), University of Manitoba, Winnipeg, Man.; Department of Pediatrics (Moraes, Subbarao), University of Toronto and Hospital for Sick Children, Toronto, Ont
| | - Meghan B Azad
- Faculty of Health Sciences (Parks, McCandless, Takaro), Simon Fraser University, Vancouver, BC; Department of Medicine (Dharma, Lefebvre, Sears), McMaster University, Hamilton, Ont.; Dalla Lana School of Public Health (Brook, Scott), University of Toronto, Toronto, Ont.; Department of Pediatrics (Turvey), University of British Columbia, Vancouver, BC; Department of Pediatrics (Mandhane, Kozyrskyj), University of Alberta, Edmonton, Alta.; Department of Pediatrics & Child Health (Becker, Azad), University of Manitoba, Winnipeg, Man.; Department of Pediatrics (Moraes, Subbarao), University of Toronto and Hospital for Sick Children, Toronto, Ont
| | - Theo J Moraes
- Faculty of Health Sciences (Parks, McCandless, Takaro), Simon Fraser University, Vancouver, BC; Department of Medicine (Dharma, Lefebvre, Sears), McMaster University, Hamilton, Ont.; Dalla Lana School of Public Health (Brook, Scott), University of Toronto, Toronto, Ont.; Department of Pediatrics (Turvey), University of British Columbia, Vancouver, BC; Department of Pediatrics (Mandhane, Kozyrskyj), University of Alberta, Edmonton, Alta.; Department of Pediatrics & Child Health (Becker, Azad), University of Manitoba, Winnipeg, Man.; Department of Pediatrics (Moraes, Subbarao), University of Toronto and Hospital for Sick Children, Toronto, Ont
| | - Diana L Lefebvre
- Faculty of Health Sciences (Parks, McCandless, Takaro), Simon Fraser University, Vancouver, BC; Department of Medicine (Dharma, Lefebvre, Sears), McMaster University, Hamilton, Ont.; Dalla Lana School of Public Health (Brook, Scott), University of Toronto, Toronto, Ont.; Department of Pediatrics (Turvey), University of British Columbia, Vancouver, BC; Department of Pediatrics (Mandhane, Kozyrskyj), University of Alberta, Edmonton, Alta.; Department of Pediatrics & Child Health (Becker, Azad), University of Manitoba, Winnipeg, Man.; Department of Pediatrics (Moraes, Subbarao), University of Toronto and Hospital for Sick Children, Toronto, Ont
| | - Malcolm R Sears
- Faculty of Health Sciences (Parks, McCandless, Takaro), Simon Fraser University, Vancouver, BC; Department of Medicine (Dharma, Lefebvre, Sears), McMaster University, Hamilton, Ont.; Dalla Lana School of Public Health (Brook, Scott), University of Toronto, Toronto, Ont.; Department of Pediatrics (Turvey), University of British Columbia, Vancouver, BC; Department of Pediatrics (Mandhane, Kozyrskyj), University of Alberta, Edmonton, Alta.; Department of Pediatrics & Child Health (Becker, Azad), University of Manitoba, Winnipeg, Man.; Department of Pediatrics (Moraes, Subbarao), University of Toronto and Hospital for Sick Children, Toronto, Ont
| | - Padmaja Subbarao
- Faculty of Health Sciences (Parks, McCandless, Takaro), Simon Fraser University, Vancouver, BC; Department of Medicine (Dharma, Lefebvre, Sears), McMaster University, Hamilton, Ont.; Dalla Lana School of Public Health (Brook, Scott), University of Toronto, Toronto, Ont.; Department of Pediatrics (Turvey), University of British Columbia, Vancouver, BC; Department of Pediatrics (Mandhane, Kozyrskyj), University of Alberta, Edmonton, Alta.; Department of Pediatrics & Child Health (Becker, Azad), University of Manitoba, Winnipeg, Man.; Department of Pediatrics (Moraes, Subbarao), University of Toronto and Hospital for Sick Children, Toronto, Ont
| | - James Scott
- Faculty of Health Sciences (Parks, McCandless, Takaro), Simon Fraser University, Vancouver, BC; Department of Medicine (Dharma, Lefebvre, Sears), McMaster University, Hamilton, Ont.; Dalla Lana School of Public Health (Brook, Scott), University of Toronto, Toronto, Ont.; Department of Pediatrics (Turvey), University of British Columbia, Vancouver, BC; Department of Pediatrics (Mandhane, Kozyrskyj), University of Alberta, Edmonton, Alta.; Department of Pediatrics & Child Health (Becker, Azad), University of Manitoba, Winnipeg, Man.; Department of Pediatrics (Moraes, Subbarao), University of Toronto and Hospital for Sick Children, Toronto, Ont
| | - Tim K Takaro
- Faculty of Health Sciences (Parks, McCandless, Takaro), Simon Fraser University, Vancouver, BC; Department of Medicine (Dharma, Lefebvre, Sears), McMaster University, Hamilton, Ont.; Dalla Lana School of Public Health (Brook, Scott), University of Toronto, Toronto, Ont.; Department of Pediatrics (Turvey), University of British Columbia, Vancouver, BC; Department of Pediatrics (Mandhane, Kozyrskyj), University of Alberta, Edmonton, Alta.; Department of Pediatrics & Child Health (Becker, Azad), University of Manitoba, Winnipeg, Man.; Department of Pediatrics (Moraes, Subbarao), University of Toronto and Hospital for Sick Children, Toronto, Ont.
| |
Collapse
|
33
|
Impact of the early-life skin microbiota on the development of canine atopic dermatitis in a high-risk breed birth cohort. Sci Rep 2020; 10:1044. [PMID: 31974513 PMCID: PMC6978374 DOI: 10.1038/s41598-020-57798-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 01/08/2020] [Indexed: 01/10/2023] Open
Abstract
Canine atopic dermatitis (CAD) is a prevalent inflammatory skin disease of dogs worldwide. Certain breeds such as the West Highland White Terriers (WHWT) are predisposed to suffer from CAD. Microbial dysbiosis is known to play a significant role in the pathogenesis of the disease, which is similar to its human counterpart, atopic dermatitis (AD). To date, no large cohort-study has been conducted in a predisposed dog breed to study the impact of the early-life microbiota on the development of CAD, as well as the possible implication of factors such as hygiene and access to the outdoors. In this study skin samples of 143 WHWT, including 109 puppies up to three weeks old and 34 parent dogs, from 17 breeders, were subjected to 16S rRNA gene and ITS2 amplicon sequencing to disclose the bacterial and fungal oral and skin microbiota, respectively. The oral samples served as a control group to confirm differences between haired and mucosal surfaces. The cutaneous microbiota differed between sample sites and age of the dogs. The season of sampling, geographical origin as well as hygiene status of the household and the access to the outdoors shaped the skin microbiota of the puppies significantly. However, we found that the individual early-life microbiota did not predispose for the later development of CAD.
Collapse
|
34
|
Underwood MA, Mukhopadhyay S, Lakshminrusimha S, Bevins CL. Neonatal intestinal dysbiosis. J Perinatol 2020; 40:1597-1608. [PMID: 32968220 PMCID: PMC7509828 DOI: 10.1038/s41372-020-00829-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/17/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022]
Abstract
The initial colonization of the neonatal intestinal tract is influenced by delivery mode, feeding, the maternal microbiota, and a host of environmental factors. After birth, the composition of the infant's microbiota undergoes a series of significant changes particularly in the first weeks and months of life ultimately developing into a more stable and diverse adult-like population in childhood. Intestinal dysbiosis is an alteration in the intestinal microbiota associated with disease and appears to be common in neonates. The consequences of intestinal dysbiosis are uncertain, but strong circumstantial evidence and limited confirmations of causality suggest that dysbiosis early in life can influence the health of the infant acutely, as well as contribute to disease susceptibility later in life.
Collapse
Affiliation(s)
- Mark A. Underwood
- grid.27860.3b0000 0004 1936 9684Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA USA
| | - Sagori Mukhopadhyay
- grid.25879.310000 0004 1936 8972Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA USA
| | - Satyan Lakshminrusimha
- grid.27860.3b0000 0004 1936 9684Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA USA
| | - Charles L. Bevins
- grid.27860.3b0000 0004 1936 9684Department of Medical Microbiology and Immunology, UC Davis School of Medicine, Davis, CA USA
| |
Collapse
|
35
|
Dumas O, Le Moual N. Damaging effects of household cleaning products on the lungs. Expert Rev Respir Med 2020; 14:1-4. [PMID: 31682770 DOI: 10.1080/17476348.2020.1689123] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/01/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Orianne Dumas
- Inserm, U1168, VIMA: Aging and Chronic Diseases, Epidemiological and Public Health Approaches, F-94807, Villejuif, France
- Univ Versailles St-Quentin-en-Yvelines, UMR-S 1168, F-78180, Montigny le Bretonneux, France
| | - Nicole Le Moual
- Inserm, U1168, VIMA: Aging and Chronic Diseases, Epidemiological and Public Health Approaches, F-94807, Villejuif, France
- Univ Versailles St-Quentin-en-Yvelines, UMR-S 1168, F-78180, Montigny le Bretonneux, France
| |
Collapse
|
36
|
Renson A, Herd P, Dowd JB. Sick Individuals and Sick (Microbial) Populations: Challenges in Epidemiology and the Microbiome. Annu Rev Public Health 2019; 41:63-80. [PMID: 31635533 DOI: 10.1146/annurev-publhealth-040119-094423] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The human microbiome represents a new frontier in understanding the biology of human health. While epidemiology in this area is still in its infancy, its scope will likely expand dramatically over the coming years. To rise to the challenge, we argue that epidemiology should capitalize on its population perspective as a critical complement to molecular microbiome research, allowing for the illumination of contextual mechanisms that may vary more across populations rather than among individuals. We first briefly review current research on social context and the gut microbiome, focusing specifically on socioeconomic status (SES) and race/ethnicity. Next, we reflect on the current state of microbiome epidemiology through the lens of one specific area, the association of the gut microbiome and metabolic disorders. We identify key methodological shortcomings of current epidemiological research in this area, including extensive selection bias, the use of noncompositionally robust measures, and a lack of attention to social factors as confounders or effect modifiers.
Collapse
Affiliation(s)
- Audrey Renson
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina 27599, USA;
| | - Pamela Herd
- McCourt School of Public Policy, Georgetown University, Washington, DC 20057, USA;
| | - Jennifer B Dowd
- Department of Global Health and Social Medicine, King's College London, London WC2B 4BG, United Kingdom; .,Current affiliation: Leverhulme Center for Demographic Science, University of Oxford, Oxford OX1 1JD, United Kingdom;
| |
Collapse
|
37
|
Ryan PM, Stanton C, Ross RP, Kelly AL, Dempsey E, Ryan CA. Paediatrician's perspective of infant gut microbiome research: current status and challenges. Arch Dis Child 2019; 104:701-705. [PMID: 31113768 DOI: 10.1136/archdischild-2019-316891] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 01/16/2023]
Abstract
Due to its innately intriguing nature and recent genomic technological advances, gut microbiome research has been at the epicentre of medical research for over a decade now. Despite the degree of publicisation, a comprehensive understanding and, therefore, acceptance of the area as a whole may be somewhat lacking within the broader medical community. This paper summarises the main analytical techniques and tools currently applied to compositional microbiome research. In addition, we outline five major lessons learnt from a decade of infant microbiome research, along with the current research gaps. Finally, we aim to provide an introduction and general guidelines relating to infant gut microbiome research for the practising paediatrician.
Collapse
Affiliation(s)
| | - Catherine Stanton
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Alan L Kelly
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
| | - Eugene Dempsey
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.,Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - C Anthony Ryan
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.,Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| |
Collapse
|
38
|
Zhang M, Differding MK, Benjamin-Neelon SE, Østbye T, Hoyo C, Mueller NT. Association of prenatal antibiotics with measures of infant adiposity and the gut microbiome. Ann Clin Microbiol Antimicrob 2019; 18:18. [PMID: 31226994 PMCID: PMC6587281 DOI: 10.1186/s12941-019-0318-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 06/06/2019] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Prenatal antibiotic exposure has been associated with an altered infant gut microbiome composition and higher risk of childhood obesity, but no studies have examined if prenatal antibiotics simultaneously alter the gut microbiome and adiposity in infants. METHOD In this prospective study (Nurture: recruitment 2013-2015 in North Carolina, United States), we examined in 454 infants the association of prenatal antibiotic exposure (by any prenatal antibiotic exposure; by trimester of pregnancy; by number of courses; by type of antibiotics) with infant age- and sex-specific weight-for-length z score (WFL-z) and skinfold thicknesses (subscapular, triceps, abdominal) at 12 months of age. In a subsample, we also examined whether prenatal antibiotic exposure was associated with alterations in the infant gut microbiome at ages 3 and 12 months. RESULTS Compared to infants not exposed to prenatal antibiotics, infants who were exposed to any prenatal antibiotics had 0.21 (95% confidence interval [CI] 0.02, 0.41) higher WFL-z at 12 months, and 0.28 (95% CI 0.02, 0.55) higher WFL-z if they were exposed to antibiotics in the second trimester, after adjustment for potential confounders, birth weight, and gestational age. We also observed a dose-dependent association (P-value for trend = 0.006) with infants exposed to ≥ 3 courses having 0.41 (95% CI 0.13, 0.68) higher WFL-z at 12 months. After further adjustment for delivery method, only second-trimester antibiotic exposure remained associated with higher infant WFL-z (0.27, 95% CI 0.003, 0.54) and subscapular skinfold thickness (0.49 mm, 95% CI 0.11, 0.88) at 12 months. Infants exposed to second-trimester antibiotics versus not had differential abundance of 13 bacterial amplicon sequence variants (ASVs) at age 3 months and 17 ASVs at 12 months (false discovery rate adjusted P-value < 0.05). CONCLUSIONS Prenatal antibiotic exposure in the second trimester was associated with an altered infant gut microbiome composition at 3 and 12 months and with higher infant WFL-z and subscapular skinfold thickness at 12 months.
Collapse
Affiliation(s)
- Mingyu Zhang
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | - Moira K. Differding
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | - Sara E. Benjamin-Neelon
- Department of Health, Behavior and Society, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | - Truls Østbye
- Department of Community and Family Medicine, Duke University Medical Center, Durham, NC USA
| | - Cathrine Hoyo
- Department of Biological Sciences and Center for Human Health and the Environment, North Carolina State University, Raleigh, NC USA
| | - Noel T. Mueller
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, MD USA
| |
Collapse
|
39
|
Baranowski T, Motil KJ, Moreno JP. Behavioral Research Agenda in a Multietiological Approach to Child Obesity Prevention. Child Obes 2019; 15:223-226. [PMID: 30925082 PMCID: PMC6622575 DOI: 10.1089/chi.2019.0052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Tom Baranowski
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX.,Address correspondence to: Tom Baranowski, PhD, Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street, Houston, TX 77030
| | - Kathleen J. Motil
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Jennette P. Moreno
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| |
Collapse
|
40
|
Hale VL, Tan CL, Niu K, Yang Y, Zhang Q, Knight R, Amato KR. Gut microbiota in wild and captive Guizhou snub-nosed monkeys, Rhinopithecus brelichi. Am J Primatol 2019; 81:e22989. [PMID: 31106872 DOI: 10.1002/ajp.22989] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/14/2019] [Accepted: 04/21/2019] [Indexed: 12/30/2022]
Abstract
Many colobine species-including the endangered Guizhou snub-nosed monkey (Rhinopithecus brelichi) are difficult to maintain in captivity and frequently exhibit gastrointestinal (GI) problems. GI problems are commonly linked to alterations in the gut microbiota, which lead us to examine the gut microbial communities of wild and captive R. brelichi. We used high-throughput sequencing of the 16S rRNA gene to compare the gut microbiota of wild (N = 7) and captive (N = 8) R. brelichi. Wild monkeys exhibited increased gut microbial diversity based on the Chao1 but not Shannon diversity metric and greater relative abundances of bacteria in the Lachnospiraceae and Ruminococcaceae families. Microbes in these families digest complex plant materials and produce butyrate, a short chain fatty acid critical to colonocyte health. Captive monkeys had greater relative abundances of Prevotella and Bacteroides species, which degrade simple sugars and carbohydrates, like those present in fruits and cornmeal, two staples of the captive R. brelichi diet. Captive monkeys also had a greater abundance of Akkermansia species, a microbe that can thrive in the face of host malnutrition. Taken together, these findings suggest that poor health in captive R. brelichi may be linked to diet and an altered gut microbiota.
Collapse
Affiliation(s)
- Vanessa L Hale
- Biological Sciences, Purdue University, West Lafayette, Indiana
| | - Chia L Tan
- LVDI International, San Marcos, California.,Nonhuman Primate Conservation and Research Institute, Tongren University, Tongren, Guizhou, China
| | - Kefeng Niu
- Institute of Eastern-Himalaya Biodiversity Research, Dali University, Dali, Yunnan, China
| | - Yeqin Yang
- Nonhuman Primate Conservation and Research Institute, Tongren University, Tongren, Guizhou, China
| | - Qikun Zhang
- Hangzhou KaiTai Biotechnology Co., Ltd, Hangzhou, China
| | - Rob Knight
- Pediatrics, University of California San Diego, La Jolla, California.,Computer Science and Engineering, University of California San Diego, La Jolla, California
| | | |
Collapse
|
41
|
Core Fucosylation of Maternal Milk N-Glycan Evokes B Cell Activation by Selectively Promoting the l-Fucose Metabolism of Gut Bifidobacterium spp. and Lactobacillus spp. mBio 2019; 10:mBio.00128-19. [PMID: 30940702 PMCID: PMC6445936 DOI: 10.1128/mbio.00128-19] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
This study provides novel evidence for the critical role of maternal milk protein glycosylation in shaping early-life gut microbiota and promoting B cell activation of neonates. The special core-fucosylated oligosaccharides might be promising prebiotics for the personalized nutrition of infants. The maternal milk glycobiome is crucial for shaping the gut microbiota of infants. Although high core fucosylation catalyzed by fucosyltransferase 8 (Fut8) is a general feature of human milk glycoproteins, its role in the formation of a healthy microbiota has not been evaluated. In this study, we found that the core-fucosylated N-glycans in milk of Chinese mothers selectively promoted the colonization of specific gut microbial groups, such as Bifidobacterium spp. and Lactobacillus spp. in their breast-fed infants during lactation. Compared with Fut8+/+ (WT) mouse-fed neonates, the offspring fed by Fut8+/− maternal mice had a distinct gut microbial profile, which was featured by a significant reduction of Lactobacillus spp., Bacteroides spp., and Bifidobacterium spp. and increased abundance of members of the Lachnospiraceae NK4A136 group and Akkermansia spp. Moreover, these offspring mice showed a lower proportion of splenic CD19+ CD69+ B lymphocytes and attenuated humoral immune responses upon ovalbumin (OVA) immunization. In vitro studies demonstrated that the chemically synthesized core-fucosylated oligosaccharides possessed the ability to promote the growth of tested Bifidobacterium and Lactobacillus strains in minimal medium. The resulting L-fucose metabolites, lactate and 1,2-propanediol, could promote the activation of B cells via the B cell receptor (BCR)-mediated signaling pathway.
Collapse
|
42
|
Baranowski T, Motil KJ, Moreno JP. Multi-etiological Perspective on Child Obesity Prevention. Curr Nutr Rep 2019; 8:10.1007/s13668-019-0256-3. [PMID: 30649714 PMCID: PMC6635107 DOI: 10.1007/s13668-019-0256-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW The simple energy balance model of obesity is inconsistent with the available findings on obesity etiology, prevention, and treatment. Yet, the most commonly stated causes of pediatric obesity are predicated on this model. A more comprehensive biological model is needed upon which to base behavioral interventions aimed at obesity prevention. In this light, alternative etiologies are little investigated and thereby poorly understood. RECENT FINDINGS Three candidate alternate etiologies are briefly presented: infectobesity, the gut microbiome, and circadian rhythms. Behavioral child obesity preventive investigators need to collaborate with biological colleagues to more intensively analyze the behavioral aspects of these etiologies and to generate innovative procedures for preventing a multi-etiological problem, e.g., group risk analysis, triaging for likely causes of obesity.
Collapse
Affiliation(s)
- Tom Baranowski
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street, Houston, TX, 77030, USA.
| | - Kathleen J Motil
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street, Houston, TX, 77030, USA
| | - Jennette P Moreno
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Street, Houston, TX, 77030, USA
| |
Collapse
|
43
|
Vo TD, Lynch BS, Roberts A. Dietary Exposures to Common Emulsifiers and Their Impact on the Gut Microbiota: Is There a Cause for Concern? Compr Rev Food Sci Food Saf 2018; 18:31-47. [DOI: 10.1111/1541-4337.12410] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/15/2018] [Accepted: 10/21/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Trung D. Vo
- the Intertek Scientific & Regulatory Consultancy; 2233 Argentia Road, Suite 201 Mississauga Ontario Canada L5N 2X7
| | - Barry S. Lynch
- the Intertek Scientific & Regulatory Consultancy; 2233 Argentia Road, Suite 201 Mississauga Ontario Canada L5N 2X7
| | - Ashley Roberts
- the Intertek Scientific & Regulatory Consultancy; 2233 Argentia Road, Suite 201 Mississauga Ontario Canada L5N 2X7
| |
Collapse
|
44
|
Postnatal exposure to household disinfectants, infant gut microbiota and subsequent risk of overweight in children. CMAJ 2018; 190:E1341. [PMID: 30420393 DOI: 10.1503/cmaj.181432] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
45
|
Differding MK, Mueller NT. Are household disinfectants microbially mediated obesogens? CMAJ 2018; 190:E1095-E1096. [PMID: 30224441 PMCID: PMC6141251 DOI: 10.1503/cmaj.181134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Affiliation(s)
- Moira K Differding
- Department of Epidemiology (Differding, Mueller), and Welch Center for Prevention, Epidemiology and Clinical Research (Mueller), Johns Hopkins Bloomberg School of Public Health, Baltimore, Md
| | - Noel T Mueller
- Department of Epidemiology (Differding, Mueller), and Welch Center for Prevention, Epidemiology and Clinical Research (Mueller), Johns Hopkins Bloomberg School of Public Health, Baltimore, Md.
| |
Collapse
|