51
|
McLoughlin IJ, Wright EM, Tagg JR, Jain R, Hale JDF. Skin Microbiome-The Next Frontier for Probiotic Intervention. Probiotics Antimicrob Proteins 2021; 14:630-647. [PMID: 34383234 DOI: 10.1007/s12602-021-09824-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2021] [Indexed: 01/16/2023]
Abstract
The skin is the largest organ in the human body, and it orchestrates many functions that are fundamentally important for our survival. Although the skin might appear to present a relatively inhospitable or even hostile environment, a multitude of commensals and also some potentially pathogenic microorganisms have successfully adapted to survive and/or thrive within the diverse ecological niches created by the skin's topographical architecture. Dysbiosis within these microbial populations can result in the emergence and pathological progression of skin diseases. Unsurprisingly, this has led to a new focus of research both for the medical dermatology and cosmetic industries that is concerned with modulation of the skin microbiome to help address common microbially mediated or modulated conditions such as acne, body odour, and atopic dermatitis. This review presents an overview of our current understanding of the complex relationship of the skin with its microbiome and then introduces the concept of probiotic intervention for the management of microbial dysbiosis within the skin ecosystem.
Collapse
Affiliation(s)
| | - Eva M Wright
- School of Pharmacy, University of Otago, PO Box 56, Dunedin, New Zealand
| | - John R Tagg
- Blis Technologies, 81 Glasgow St, South Dunedin, 9012, Dunedin, New Zealand
| | - Rohit Jain
- Blis Technologies, 81 Glasgow St, South Dunedin, 9012, Dunedin, New Zealand
| | - John D F Hale
- Blis Technologies, 81 Glasgow St, South Dunedin, 9012, Dunedin, New Zealand.
| |
Collapse
|
52
|
Clarke SLN, Mageean KS, Maccora I, Harrison S, Simonini G, Sharp GC, Relton CL, Ramanan AV. Moving from nature to nurture: a systematic review and meta-analysis of environmental factors associated with juvenile idiopathic arthritis. Rheumatology (Oxford) 2021; 61:514-530. [PMID: 34382060 PMCID: PMC8824412 DOI: 10.1093/rheumatology/keab627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/10/2021] [Accepted: 07/21/2021] [Indexed: 11/23/2022] Open
Abstract
Objectives JIA is the most common paediatric rheumatic disease, thought to be influenced by both genetics and the environment. Identifying environmental factors associated with disease risk will improve knowledge of disease mechanism and ultimately benefit patients. This review aimed to collate and synthesize the current evidence of environmental factors associated with JIA. Methods Four databases (MEDLINE, Embase, Web of Science and Cumulative Index to Nursing and Allied Health Literature) were searched from inception to January 2020. Study quality was rated using the Newcastle-Ottawa Scale. Pooled estimates for each environmental factor were generated using a random-effects, inverse-variance method, where possible. The remaining environmental factors were synthesized in narrative form. Results This review includes 66 environmental factors from 39 studies (11 cohort and 28 case-control studies) over 45 years. Study sample sizes ranged from 41 to 1.9 million participants. Eight environmental factors from ten studies were meta-analysed. Caesarean section delivery was associated with increased JIA risk [pooled odds ratio (OR) 1.11, 95% CI: 1.01, 1.22]. Conversely, presence (vs absence) of siblings (pooled OR 0.60, 95% CI: 0.44, 0.81) and maternal prenatal smoking (pooled OR 0.70, 95% CI: 0.58, 0.84) were associated with decreased JIA risk. Conclusion This review identifies several environmental factors associated with JIA and demonstrates the huge breadth of environmental research undertaken over five decades. We also highlight the challenges of combining data collected over this period due to limited between study comparability, evolution in healthcare and social practices, and changing environment, which warrant consideration when planning future studies.
Collapse
Affiliation(s)
- Sarah L N Clarke
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.,Department of Paediatric Rheumatology, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Katie S Mageean
- Department of Paediatric Rheumatology, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Ilaria Maccora
- Rheumatology Unit, A Meyer Children Hospital, NEUROFARBA Department, University of Florence, Florence, Italy
| | - Sean Harrison
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Gabriele Simonini
- Rheumatology Unit, A Meyer Children Hospital, NEUROFARBA Department, University of Florence, Florence, Italy
| | - Gemma C Sharp
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Athimalaipet V Ramanan
- Department of Paediatric Rheumatology, University Hospitals Bristol NHS Foundation Trust, Bristol, UK.,Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
53
|
Haddad EN, Comstock SS. Archive for Research in Child Health (ARCH) and Baby Gut: Study Protocol for a Remote, Prospective, Longitudinal Pregnancy and Birth Cohort to Address Microbiota Development and Child Health. Methods Protoc 2021; 4:mps4030052. [PMID: 34449678 PMCID: PMC8395764 DOI: 10.3390/mps4030052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/16/2021] [Accepted: 07/24/2021] [Indexed: 02/05/2023] Open
Abstract
The infant gut microbiome is shaped by numerous factors such as diet and the maternal microbiota and is also associated with later atopy and obesity. The Archive for Research in Child Health and Baby Gut (ARCHBG) cohort was established in 2015 to (1) understand how the development of the infant gut microbiota is associated with atopy, obesity, and gastrointestinal disease and (2) characterize the associations of maternal pre-pregnancy BMI and infant diet with the development of the gut microbiota. Study participants for ARCHBG are convenience samples recruited through two pipelines in Lansing and Traverse City, Michigan: (1) Archive for Research in Child Health (ARCHGUT) and (2) BABYGUT. A total of (n = 51) mother–infant dyads have been enrolled to date. This prospective cohort study collects maternal pre-pregnancy fecal samples, maternal data, child fecal samples at four timepoints (one week, six months, 12 months, and 24 months), and child data up to five years of age. All samples and data are collected remotely by mail, phone, or drop-off at select locations. Of all participants enrolled, 76.5% (n = 39) of infants have a complete record of stool samples. At least 88.2% (n = 45) of fecal samples were submitted at each timepoint. ARCHBG will allow for a nuanced understanding of the temporal development of the infant gut microbiome and numerous child health outcomes.
Collapse
|
54
|
Chinnappan M, Harris-Tryon TA. Novel mechanisms of microbial crosstalk with skin innate immunity. Exp Dermatol 2021; 30:1484-1495. [PMID: 34252227 DOI: 10.1111/exd.14429] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/28/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022]
Abstract
Skin is an organ with a dynamic ecosystem that harbours pathogenic and commensal microbes, which constantly communicate amongst each other and with the host immune system. Evolutionarily, skin and its microbiota have evolved to remain in homeostasis. However, frequently this homeostatic relationship is disturbed by a variety of factors such as environmental stress, diet, genetic mutations, and the microbiome itself. Commensal microbes also play a major role in the maintenance of microbial homeostasis. In addition to their ability to limit pathogens, many skin commensals such as Staphylococcus epidermidis and Cutibacterium acnes have recently been implicated in disease pathogenesis either by directly modulating the host immune components or by supporting the expansion of other pathogenic microbes. Likewise, opportunistic skin pathogens such as Staphylococcus aureus and Staphylococcus lugdunensis are able to breach the skin and cause disease. Though much has been established about the microbiota's function in skin immunity, we are in a time where newer mechanistic insights rapidly redefine our understanding of the host/microbial interface in the skin. In this review, we provide a concise summary of recent advances in our understanding of the interplay between host defense strategies and the skin microbiota.
Collapse
Affiliation(s)
- Mahendran Chinnappan
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tamia A Harris-Tryon
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
55
|
Ser HL, Letchumanan V, Goh BH, Wong SH, Lee LH. The Use of Fecal Microbiome Transplant in Treating Human Diseases: Too Early for Poop? Front Microbiol 2021; 12:519836. [PMID: 34054740 PMCID: PMC8155486 DOI: 10.3389/fmicb.2021.519836] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/07/2021] [Indexed: 12/15/2022] Open
Abstract
Fecal microbiome transplant (FMT) has gained popularity over the past few years, given its success in treating several gastrointestinal diseases. At the same time, microbial populations in the gut have been shown to have more physiological effects than we expected as "habitants" of the gut. The imbalance in the gut microbiome or dysbiosis, particularly when there are excessive harmful pathogens, can trigger not just infections but can also result in the development of common diseases, such as cancer and cardiometabolic diseases. By using FMT technology, the dysbiosis of the gut microbiome in patients can be resolved by administering fecal materials from a healthy donor. The current review summarizes the history and current uses of FMT before suggesting potential ideas for its high-quality application in clinical settings.
Collapse
Affiliation(s)
- Hooi-Leng Ser
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Vengadesh Letchumanan
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Bey-Hing Goh
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Sunny Hei Wong
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
56
|
Fan HY, Tung YT, Yang YCSH, Hsu JB, Lee CY, Chang TH, Su ECY, Hsieh RH, Chen YC. Maternal Vegetable and Fruit Consumption during Pregnancy and Its Effects on Infant Gut Microbiome. Nutrients 2021; 13:1559. [PMID: 34063157 PMCID: PMC8148194 DOI: 10.3390/nu13051559] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 12/20/2022] Open
Abstract
Maternal nutrition intake during pregnancy may affect the mother-to-child transmission of bacteria, resulting in gut microflora changes in the offspring, with long-term health consequences in later life. Longitudinal human studies are lacking, as only a small amount of studies showing the effect of nutrition intake during pregnancy on the gut microbiome of infants have been performed, and these studies have been mainly conducted on animals. This pilot study explores the effects of high or low fruit and vegetable gestational intake on the infant microbiome. We enrolled pregnant women with a complete 3-day dietary record and received postpartum follow-up. The 16S rRNA gene sequence was used to characterize the infant gut microbiome at 2 months (n = 39). Principal coordinate analysis ordination revealed that the infant gut microbiome clustered differently for high and low maternal fruit and vegetable consumption (p < 0.001). The linear discriminant analysis effect size and feature selection identified 6 and 17 taxa from both the high and low fruit and vegetable consumption groups. Among the 23 abundant taxa, we observed that six maternal intake nutrients were associated with nine taxa (e.g., Erysipelatoclostridium, Isobaculum, Lachnospiraceae, Betaproteobacteria, Burkholderiaceae, Sutterella, Clostridia, Clostridiales, and Lachnoclostridium). The amount of gestational fruit and vegetable consumption is associated with distinct changes in the infant gut microbiome at 2 months of age. Therefore, strategies involving increased fruit and vegetable consumption during pregnancy should be employed for modifying the gut microbiome early in life.
Collapse
Affiliation(s)
- Hsien-Yu Fan
- Department of Family Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan;
- Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Yu-Tang Tung
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan;
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
| | - Yu-Chen S. H. Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei 110, Taiwan;
| | - Justin BoKai Hsu
- Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan;
| | - Cheng-Yang Lee
- Office of Information Technology, Taipei Medical University, Taipei 110, Taiwan; (C.-Y.L.); (T.-H.C.)
| | - Tzu-Hao Chang
- Office of Information Technology, Taipei Medical University, Taipei 110, Taiwan; (C.-Y.L.); (T.-H.C.)
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan;
| | - Emily Chia-Yu Su
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan;
- Clinical Big Data Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Rong-Hong Hsieh
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan;
| | - Yang-Ching Chen
- Department of Family Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan;
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan;
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan;
- Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
57
|
Lauby SC, Fleming AS, McGowan PO. Beyond maternal care: The effects of extra-maternal influences within the maternal environment on offspring neurodevelopment and later-life behavior. Neurosci Biobehav Rev 2021; 127:492-501. [PMID: 33905789 DOI: 10.1016/j.neubiorev.2021.04.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/08/2021] [Accepted: 04/17/2021] [Indexed: 01/26/2023]
Abstract
The early-life maternal environment has a profound and persistent effect on offspring neuroendocrine function, neurotransmitter systems, and behavior. Studies using rodent models suggest that early-life maternal care can influence the 'developmental programming' of offspring in part through altered epigenetic regulation of specific genes. The exploration of epigenetic regulation of these genes as a biological mechanism has been important to our understanding of how animals adapt to their environments and how these developmental trajectories may be altered. However, other non-maternal factors have been shown to act directly, or to interact with maternal care, to influence later-life phenotype. Based on accumulating evidence, including our research, we discuss other important influences on the developmental programming of offspring. We highlight early-life variations in temperature exposure and offspring genotype x environment interactions as prominent examples. We conclude with recommendations for future investigations on how early-life maternal care and extra-maternal influences lead to persistent changes in the brain and behavior of the offspring throughout development.
Collapse
Affiliation(s)
- Samantha C Lauby
- Department of Biological Sciences, University of Toronto Scarborough Campus, Scarborough, ON, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Alison S Fleming
- Department of Psychology, University of Toronto, Toronto, ON, Canada; Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada.
| | - Patrick O McGowan
- Department of Biological Sciences, University of Toronto Scarborough Campus, Scarborough, ON, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada; Department of Psychology, University of Toronto, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
58
|
Sturov NV, Popov SV, Zhukov VA. Modern approaches to the correction of the gut microbiota. MEDITSINSKIY SOVET = MEDICAL COUNCIL 2021:136-143. [DOI: 10.21518/2079-701x-2021-4-136-143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
The article presents modern data on the formation, structure, functions and possibilities of correction of the gut microbiota. The gut microbiota is a collection of living organisms that inhabit the human intestine and form a complex microecological system that performs many functions. It is known that the composition and state of the gut microbiota is influenced by both environmental factors, such as diet and lifestyle, and the human body, including genetic predisposition. A violation in this system (dysbiosis) can provoke the development of a number of diseases and pathological conditions, in which the correction of the gut microbiota may be a promising therapeutic strategy. The most common methods of correcting dysbiosis are dieting, the use of pro-and prebiotics, and fecal microbiota transplantation. The diet affects the qualitative and quantitative composition and functions of the gut microbiota, the activity of its individual representatives. Probiotics are used to modulate, preserve the gut microbiota in dysbiosis, as well as to prevent its development. Fecal microbiota transplantation is performed by transferring the microbiota from a healthy donor. This method is one of the most effective ways to treat Clostridium difficile infection. This review article also presents the results of fecal microbiota transplantation in patients with inflammatory bowel disease and hepatic encephalopathy. It is shown that after transplantation, there is a rapid change in the composition of the gut microbiota, which becomes similar to the microbiota of a healthy donor. Each of these methods of correction demonstrates a different degree of influence on the gut microbiota, and their therapeutic effectiveness depends on the direct characteristics of the methods used, as well as the specific disease and requires further study.
Collapse
|
59
|
Skowron K, Bauza-Kaszewska J, Kraszewska Z, Wiktorczyk-Kapischke N, Grudlewska-Buda K, Kwiecińska-Piróg J, Wałecka-Zacharska E, Radtke L, Gospodarek-Komkowska E. Human Skin Microbiome: Impact of Intrinsic and Extrinsic Factors on Skin Microbiota. Microorganisms 2021; 9:543. [PMID: 33808031 PMCID: PMC7998121 DOI: 10.3390/microorganisms9030543] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 02/28/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
The skin is the largest organ of the human body and it protects the body from the external environment. It has become the topic of interest of researchers from various scientific fields. Microorganisms ensure the proper functioning of the skin. Of great importance, are the mutual relations between such microorganisms and their responses to environmental impacts, as dysbiosis may contribute to serious skin diseases. Molecular methods, used for microorganism identification, allow us to gain a better understanding of the skin microbiome. The presented article contains the latest reports on the skin microbiota in health and disease. The review discusses the relationship between a properly functioning microbiome and the body's immune system, as well as the impact of internal and external factors on the human skin microbiome.
Collapse
Affiliation(s)
- Krzysztof Skowron
- Department of Microbiology, Nicolaus Copernicus University in Toruń, L. Rydygier Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland; (Z.K.); (N.W.-K.); (K.G.-B.); (J.K.-P.); (E.G.-K.)
| | - Justyna Bauza-Kaszewska
- Department of Microbiology and Food Technology, UTP University of Science and Technology, 85-029 Bydgoszcz, Poland;
| | - Zuzanna Kraszewska
- Department of Microbiology, Nicolaus Copernicus University in Toruń, L. Rydygier Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland; (Z.K.); (N.W.-K.); (K.G.-B.); (J.K.-P.); (E.G.-K.)
| | - Natalia Wiktorczyk-Kapischke
- Department of Microbiology, Nicolaus Copernicus University in Toruń, L. Rydygier Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland; (Z.K.); (N.W.-K.); (K.G.-B.); (J.K.-P.); (E.G.-K.)
| | - Katarzyna Grudlewska-Buda
- Department of Microbiology, Nicolaus Copernicus University in Toruń, L. Rydygier Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland; (Z.K.); (N.W.-K.); (K.G.-B.); (J.K.-P.); (E.G.-K.)
| | - Joanna Kwiecińska-Piróg
- Department of Microbiology, Nicolaus Copernicus University in Toruń, L. Rydygier Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland; (Z.K.); (N.W.-K.); (K.G.-B.); (J.K.-P.); (E.G.-K.)
| | - Ewa Wałecka-Zacharska
- Department of Food Hygiene and Consumer Health, Wrocław University of Environmental and Life Sciences, 31 C.K. Norwida St., 50-375 Wrocław, Poland;
| | - Laura Radtke
- Faculty of Civil and Environmental Engineering and Architecture, UTP University of Science and Technology in Bydgoszcz, Al. prof. S. Kaliskiego 7, 85-796 Bydgoszcz, Poland;
| | - Eugenia Gospodarek-Komkowska
- Department of Microbiology, Nicolaus Copernicus University in Toruń, L. Rydygier Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland; (Z.K.); (N.W.-K.); (K.G.-B.); (J.K.-P.); (E.G.-K.)
| |
Collapse
|
60
|
Dumitriu D, Emeruwa UN, Hanft E, Liao GV, Ludwig E, Walzer L, Arditi B, Saslaw M, Andrikopoulou M, Scripps T, Baptiste C, Khan A, Breslin N, Rubenstein D, Simpson LL, Kyle MH, Friedman AM, Hirsch DS, Miller RS, Fernández CR, Fuchs KM, Keown MK, Glassman ME, Stephens A, Gupta A, Sultan S, Sibblies C, Whittier S, Abreu W, Akita F, Penn A, D’Alton ME, Orange JS, Goffman D, Saiman L, Stockwell MS, Gyamfi-Bannerman C. Outcomes of Neonates Born to Mothers With Severe Acute Respiratory Syndrome Coronavirus 2 Infection at a Large Medical Center in New York City. JAMA Pediatr 2021; 175:157-167. [PMID: 33044493 PMCID: PMC7551222 DOI: 10.1001/jamapediatrics.2020.4298] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
IMPORTANCE Limited data on vertical and perinatal transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and health outcomes of neonates born to mothers with symptomatic or asymptomatic coronavirus disease 2019 (COVID-19) are available. Studies are needed to inform evidence-based infection prevention and control (IP&C) policies. OBJECTIVE To describe the outcomes of neonates born to mothers with perinatal SARS-CoV-2 infection and the IP&C practices associated with these outcomes. DESIGN, SETTING, AND PARTICIPANTS This retrospective cohort analysis reviewed the medical records for maternal and newborn data for all 101 neonates born to 100 mothers positive for or with suspected SARS-CoV-2 infection from March 13 to April 24, 2020. Testing for SARS-CoV-2 was performed using Cobas (Roche Diagnostics) or Xpert Xpress (Cepheid) assays. Newborns were admitted to well-baby nurseries (WBNs) (82 infants) and neonatal intensive care units (NICUs) (19 infants) in 2 affiliate hospitals at a large academic medical center in New York, New York. Newborns from the WBNs roomed-in with their mothers, who were required to wear masks. Direct breastfeeding after appropriate hygiene was encouraged. EXPOSURES Perinatal exposure to maternal asymptomatic/mild vs severe/critical COVID-19. MAIN OUTCOMES AND MEASURES The primary outcome was newborn SARS-CoV-2 testing results. Maternal COVID-19 status was classified as asymptomatic/mildly symptomatic vs severe/critical. Newborn characteristics and clinical courses were compared across maternal COVID-19 severity. RESULTS In total, 141 tests were obtained from 101 newborns (54 girls [53.5%]) on 0 to 25 days of life (DOL-0 to DOL-25) (median, DOL-1; interquartile range [IQR], DOL-1 to DOL-3). Two newborns had indeterminate test results, indicative of low viral load (2.0%; 95% CI, 0.2%-7.0%); 1 newborn never underwent retesting but remained well on follow-up, and the other had negative results on retesting. Maternal severe/critical COVID-19 was associated with newborns born approximately 1 week earlier (median gestational age, 37.9 [IQR, 37.1-38.4] vs 39.1 [IQR, 38.3-40.2] weeks; P = .02) and at increased risk of requiring phototherapy (3 of 10 [30.0%] vs 6 of 91 [7.0%]; P = .04) compared with newborns of mothers with asymptomatic/mild COVID-19. Fifty-five newborns were followed up in a new COVID-19 Newborn Follow-up Clinic at DOL-3 to DOL-10 and remained well. Twenty of these newborns plus 3 newborns followed up elsewhere had 32 nonroutine encounters documented at DOL-3 to DOL-25, and none had evidence of SARS-CoV-2 infection, including 6 with negative retesting results. CONCLUSIONS AND RELEVANCE No clinical evidence of vertical transmission was identified in 101 newborns of mothers positive for or with suspected SARS-CoV-2 infection, despite most newborns rooming-in and direct breastfeeding practices.
Collapse
Affiliation(s)
- Dani Dumitriu
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York,NewYork–Presbyterian Hospital, New York, New York,Division of Developmental Neuroscience, Department of Psychiatry, Columbia University Irving Medical Center, New York, New York,Sackler Institute, Zuckerman Institute, and the Columbia Population Research Center, Columbia University, New York, New York
| | - Ukachi N. Emeruwa
- NewYork–Presbyterian Hospital, New York, New York,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York
| | - Erin Hanft
- Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Grace V. Liao
- Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Elizabeth Ludwig
- NewYork–Presbyterian Hospital, New York, New York,Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York
| | - Lauren Walzer
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York,NewYork–Presbyterian Hospital, New York, New York
| | - Brittany Arditi
- NewYork–Presbyterian Hospital, New York, New York,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York
| | - Minna Saslaw
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York,NewYork–Presbyterian Hospital, New York, New York
| | - Maria Andrikopoulou
- NewYork–Presbyterian Hospital, New York, New York,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York
| | - Tessa Scripps
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York,NewYork–Presbyterian Hospital, New York, New York
| | - Caitlin Baptiste
- NewYork–Presbyterian Hospital, New York, New York,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York
| | - Adrita Khan
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York,NewYork–Presbyterian Hospital, New York, New York
| | - Noelle Breslin
- NewYork–Presbyterian Hospital, New York, New York,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York
| | - David Rubenstein
- NewYork–Presbyterian Hospital, New York, New York,Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Lynn L. Simpson
- NewYork–Presbyterian Hospital, New York, New York,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York
| | - Margaret H. Kyle
- Division of Developmental Neuroscience, Department of Psychiatry, Columbia University Irving Medical Center, New York, New York
| | - Alexander M. Friedman
- NewYork–Presbyterian Hospital, New York, New York,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York
| | - Daniel S. Hirsch
- NewYork–Presbyterian Hospital, New York, New York,Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Russell S. Miller
- NewYork–Presbyterian Hospital, New York, New York,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York
| | - Cristina R. Fernández
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York,NewYork–Presbyterian Hospital, New York, New York
| | - Karin M. Fuchs
- NewYork–Presbyterian Hospital, New York, New York,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York
| | - M. Kathleen Keown
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York,NewYork–Presbyterian Hospital, New York, New York
| | - Melissa E. Glassman
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York,NewYork–Presbyterian Hospital, New York, New York
| | - Ashley Stephens
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York,NewYork–Presbyterian Hospital, New York, New York
| | - Archana Gupta
- NewYork–Presbyterian Hospital, New York, New York,Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Sally Sultan
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York,NewYork–Presbyterian Hospital, New York, New York
| | - Caroline Sibblies
- NewYork–Presbyterian Hospital, New York, New York,Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Susan Whittier
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York
| | - Wanda Abreu
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York,NewYork–Presbyterian Hospital, New York, New York
| | - Francis Akita
- NewYork–Presbyterian Hospital, New York, New York,Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Anna Penn
- NewYork–Presbyterian Hospital, New York, New York,Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Mary E. D’Alton
- NewYork–Presbyterian Hospital, New York, New York,Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York
| | - Jordan S. Orange
- NewYork–Presbyterian Hospital, New York, New York,Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Dena Goffman
- NewYork–Presbyterian Hospital, New York, New York,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York,Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York
| | - Lisa Saiman
- NewYork–Presbyterian Hospital, New York, New York,Division of Pediatric Infectious Diseases, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Melissa S. Stockwell
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York,NewYork–Presbyterian Hospital, New York, New York,Department of Population and Family Health, Mailman School of Public Health, Columbia University Irving Medical Center, New York, New York
| | - Cynthia Gyamfi-Bannerman
- NewYork–Presbyterian Hospital, New York, New York,Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
61
|
Vijaya Chandra SH, Srinivas R, Dawson TL, Common JE. Cutaneous Malassezia: Commensal, Pathogen, or Protector? Front Cell Infect Microbiol 2021; 10:614446. [PMID: 33575223 PMCID: PMC7870721 DOI: 10.3389/fcimb.2020.614446] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/04/2020] [Indexed: 12/19/2022] Open
Abstract
The skin microbial community is a multifunctional ecosystem aiding prevention of infections from transient pathogens, maintenance of host immune homeostasis, and skin health. A better understanding of the complex milieu of microbe-microbe and host-microbe interactions will be required to define the ecosystem's optimal function and enable rational design of microbiome targeted interventions. Malassezia, a fungal genus currently comprising 18 species and numerous functionally distinct strains, are lipid-dependent basidiomycetous yeasts and integral components of the skin microbiome. The high proportion of Malassezia in the skin microbiome makes understanding their role in healthy and diseased skin crucial to development of functional skin health knowledge and understanding of normal, healthy skin homeostasis. Over the last decade, new tools for Malassezia culture, detection, and genetic manipulation have revealed not only the ubiquity of Malassezia on skin but new pathogenic roles in seborrheic dermatitis, psoriasis, Crohn's disease, and pancreatic ductal carcinoma. Application of these tools continues to peel back the layers of Malassezia/skin interactions, including clear examples of pathogenicity, commensalism, and potential protective or beneficial activities creating mutualism. Our increased understanding of host- and microbe-specific interactions should lead to identification of key factors that maintain skin in a state of healthy mutualism or, in turn, initiate pathogenic changes. These approaches are leading toward development of new therapeutic targets and treatment options. This review discusses recent developments that have expanded our understanding of Malassezia's role in the skin microbiome, with a focus on its multiple roles in health and disease as commensal, pathogen, and protector.
Collapse
Affiliation(s)
| | - Ramasamy Srinivas
- Skin Research Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Thomas L Dawson
- Skin Research Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Drug Discovery, College of Pharmacy, Medical University of South Carolina, Charleston, SC, United States
| | - John E Common
- Skin Research Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| |
Collapse
|
62
|
Kaur G, Behl T, Bungau S, Kumar A, Uddin MS, Mehta V, Zengin G, Mathew B, Shah MA, Arora S. Dysregulation of the Gut-Brain Axis, Dysbiosis and Influence of Numerous Factors on Gut Microbiota Associated Parkinson's Disease. Curr Neuropharmacol 2021; 19:233-247. [PMID: 32504503 PMCID: PMC8033978 DOI: 10.2174/1570159x18666200606233050] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/27/2020] [Accepted: 06/02/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) has been one of the substantial social, medical concerns and, burdens of the present time. PD is a gradually devastating neurodegenerative disorder of the neurological function marked with α-synucleinopathy affecting numerous regions of the brain-gut axis, as well as the central, enteric, and autonomic nervous system. Its etiology is a widely disputed topic. OBJECTIVE This review emphasizes to find out the correlation among the microbial composition and the observable disturbances in the metabolites of the microbial species and its impact on the immune response, which may have a concrete implication on the occurrence, persistence and, pathophysiology of PD via the gut-brain axis. METHODS An in-depth research and the database was developed from the available peer-reviewed articles to date (March 2020) utilizing numerous search engines like PubMed, MEDLINE and, other internet sources. RESULTS Progressively increasing shreds of evidence have proved the fact that dysbiosis in the gut microbiome plays a central role in many neurological disorders, such as PD. Indeed, a disordered microbiome-gut-brain axis in PD could be focused on gastrointestinal afflictions that manifest primarily several years prior to the diagnosis, authenticating a concept wherein the pathological pathway progresses from the intestine reaching the brain. CONCLUSION The microbiota greatly affects the bidirectional interaction between the brain and the gut via synchronized neurological, immunological, and neuroendocrine mechanisms. It can be concluded that a multitude of factors discussed in this review steadily induce the onset of dysbacteriosis that may exacerbate the etiologic mechanism of Parkinson's disease.
Collapse
Affiliation(s)
| | - Tapan Behl
- Address correspondence to this author at the Chitkara College of Pharmacy, Chitkara University, Punjab, India; Tel: +91-8527517931;, E-mails: ;
| | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Hoang DM, Levy EI, Vandenplas Y. The impact of Caesarean section on the infant gut microbiome. Acta Paediatr 2021; 110:60-67. [PMID: 33405258 DOI: 10.1111/apa.15501] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 07/19/2020] [Accepted: 07/22/2020] [Indexed: 12/14/2022]
Abstract
AIM Description of the impact of caesarean section on the infant gut microbiome, infant health and solutions to restore the dysbiosis. METHODS We searched PubMed and Google Scholar for relevant articles. Only articles published in English were selected. Separate searches were performed for each topic. We selected 60 articles published between 1999 and 2019 by extracting subject headings and key words of interest for this topic. RESULTS Caesarean section is an obstetrical procedure that is increasing in prevalence worldwide. On top of the maternal and neonatal risks that this procedure carries, it also induces a dysbiosis of the infant intestinal microbiome possibly challenging the health outcome for the infant. Antibiotics administered during Caesarean deliveries contribute to the development of the gut microbiome. Nonetheless, breastfeeding and several interventions such as vaginal seeding and supplementation with probiotics, prebiotics and synbiotics may contribute to the restoration of the dysbiosis. CONCLUSION Caesarean section is associated with the development of an imbalance of the infant gut microbiome. Long-term consequences of this imbalance are debated. Besides breastfeeding, other strategies to restore this dysbiosis need further studies before they can be recommended.
Collapse
Affiliation(s)
- Delphine M. Hoang
- KidZ Health Castle Universitair Ziekenhuis Brussel Vrije Universiteit Brussel Brussels Belgium
| | - Elvira I. Levy
- KidZ Health Castle Universitair Ziekenhuis Brussel Vrije Universiteit Brussel Brussels Belgium
| | - Yvan Vandenplas
- KidZ Health Castle Universitair Ziekenhuis Brussel Vrije Universiteit Brussel Brussels Belgium
| |
Collapse
|
64
|
Royal C, Gray C. Allergy Prevention: An Overview of Current Evidence. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2020; 93:689-698. [PMID: 33380931 PMCID: PMC7757062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background: There has been a rapid rise in allergic disorders across the globe. This has increased research into the determinants of allergy development, to identify factors that may be manipulated to mitigate risk. An opportune window in immunological development appears to exist in early life whereby certain exposures may promote or prevent the development of an allergic disposition. Furthermore, factors that affect the composition and diversity of the microbiome in early life have been explored. In this review, we discuss current literature and recommendations relating to exposures that may prevent allergy development or promote tolerance. Risk factors and recommendations: Delivery by caesarean section, omission of breastfeeding, vitamin D insufficiency, and environmental exposures, such as cigarette smoke exposure, all increase the risk of an allergic predisposition. Dietary diversity during pregnancy, lactation, and in infancy is protective. Breastfeeding for at least 4 months reduces the risk of eczema. Recommendations for food-allergen exposure has shifted from delayed introduction to early introduction as a tolerance-inducing strategy. Supplements such as probiotics and vitamins during pregnancy and infancy have yet to produce conclusive results for allergy prevention. Emollient use in infancy has not been shown to be protective against eczema or food allergy.
Collapse
Affiliation(s)
- Candice Royal
- To whom all correspondence should be addressed:
Candice Royal, Paediatric Allergology, Kidsallergy Paediatric and Allergy
Centre, Cape Town, South Africa;
| | | |
Collapse
|
65
|
Lutz H, Vangelatos A, Gottel N, Osculati A, Visona S, Finley SJ, Gilbert JA, Javan GT. Effects of Extended Postmortem Interval on Microbial Communities in Organs of the Human Cadaver. Front Microbiol 2020; 11:569630. [PMID: 33363519 PMCID: PMC7752770 DOI: 10.3389/fmicb.2020.569630] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/16/2020] [Indexed: 12/20/2022] Open
Abstract
Human thanatomicrobiota studies have shown that microorganisms inhabit and proliferate externally and internally throughout the body and are the primary mediators of putrefaction after death. Yet little is known about the source and diversity of the thanatomicrobiome or the underlying factors leading to delayed decomposition exhibited by reproductive organs. The use of the V4 hypervariable region of bacterial 16S rRNA gene sequences for taxonomic classification ("barcoding") and phylogenetic analyses of human postmortem microbiota has recently emerged as a possible tool in forensic microbiology. The goal of this study was to apply a 16S rRNA barcoding approach to investigate variation among different organs, as well as the extent to which microbial associations among different body organs in human cadavers can be used to predict forensically important determinations, such as cause and time of death. We assessed microbiota of organ tissues including brain, heart, liver, spleen, prostate, and uterus collected at autopsy from criminal casework of 40 Italian cadavers with times of death ranging from 24 to 432 h. Both the uterus and prostate had a significantly higher alpha diversity compared to other anatomical sites, and exhibited a significantly different microbial community composition from non-reproductive organs, which we found to be dominated by the bacterial orders MLE1-12, Saprospirales, and Burkholderiales. In contrast, reproductive organs were dominated by Clostridiales, Lactobacillales, and showed a marked decrease in relative abundance of MLE1-12. These results provide insight into the observation that the uterus and prostate are the last internal organs to decay during human decomposition. We conclude that distinct community profiles of reproductive versus non-reproductive organs may help guide the application of forensic microbiology tools to investigations of human cadavers.
Collapse
Affiliation(s)
- Holly Lutz
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States.,Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA, United States
| | | | - Neil Gottel
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States.,Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA, United States
| | - Antonio Osculati
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Silvia Visona
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Sheree J Finley
- Physical Sciences Department, Forensic Science Programs, Alabama State University, Montgomery, AL, United States
| | - Jack A Gilbert
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States.,Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA, United States
| | - Gulnaz T Javan
- Physical Sciences Department, Forensic Science Programs, Alabama State University, Montgomery, AL, United States
| |
Collapse
|
66
|
Kyle MH, Glassman ME, Khan A, Fernández CR, Hanft E, Emeruwa UN, Scripps T, Walzer L, Liao GV, Saslaw M, Rubenstein D, Hirsch DS, Keown MK, Stephens A, Mollicone I, Bence ML, Gupta A, Sultan S, Sibblies C, Whittier S, Abreu W, Akita F, Penn A, Orange JS, Saiman L, Welch MG, Gyamfi-Bannerman C, Stockwell MS, Dumitriu D. A review of newborn outcomes during the COVID-19 pandemic. Semin Perinatol 2020; 44:151286. [PMID: 32826081 PMCID: PMC7376345 DOI: 10.1016/j.semperi.2020.151286] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
As the COVID-19 pandemic continues to spread worldwide, it is crucial that we determine populations that are at-risk and develop appropriate clinical care policies to protect them. While several respiratory illnesses are known to seriously impact pregnant women and newborns, preliminary data on the novel SARS-CoV-2 Coronavirus suggest that these groups are no more at-risk than the general population. Here, we review the available literature on newborns born to infected mothers and show that newborns of mothers with positive/suspected SARS-CoV-2 infection rarely acquire the disease or show adverse clinical outcomes. With this evidence in mind, it appears that strict postnatal care policies, including separating mothers and newborns, discouraging breastfeeding, and performing early bathing, may be more likely to adversely impact newborns than they are to reduce the low risk of maternal transmission of SARS-CoV-2 or the even lower risk of severe COVID-19 disease in otherwise healthy newborns.
Collapse
Affiliation(s)
- Margaret H Kyle
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA; Division of Developmental Neuroscience, Psychiatry Department, Columbia University Irving Medical Center, New York, NY USA
| | - Melissa E Glassman
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA; NewYork-Presbyterian Hospital, New York, NY USA
| | - Adrita Khan
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA; NewYork-Presbyterian Hospital, New York, NY USA
| | - Cristina R Fernández
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA; NewYork-Presbyterian Hospital, New York, NY USA
| | - Erin Hanft
- Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA
| | - Ukachi N Emeruwa
- NewYork-Presbyterian Hospital, New York, NY USA; Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY USA
| | - Tessa Scripps
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA; NewYork-Presbyterian Hospital, New York, NY USA
| | - Lauren Walzer
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA; NewYork-Presbyterian Hospital, New York, NY USA
| | - Grace V Liao
- Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA
| | - Minna Saslaw
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA; NewYork-Presbyterian Hospital, New York, NY USA
| | - David Rubenstein
- NewYork-Presbyterian Hospital, New York, NY USA; Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA
| | - Daniel S Hirsch
- NewYork-Presbyterian Hospital, New York, NY USA; Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA
| | - M Kathleen Keown
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA; NewYork-Presbyterian Hospital, New York, NY USA
| | - Ashley Stephens
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA; NewYork-Presbyterian Hospital, New York, NY USA
| | - Isabelle Mollicone
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA
| | - Mary L Bence
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA
| | - Archana Gupta
- NewYork-Presbyterian Hospital, New York, NY USA; Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA
| | - Sally Sultan
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA; NewYork-Presbyterian Hospital, New York, NY USA
| | - Caroline Sibblies
- NewYork-Presbyterian Hospital, New York, NY USA; Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA
| | - Susan Whittier
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY USA
| | - Wanda Abreu
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA; NewYork-Presbyterian Hospital, New York, NY USA
| | - Francis Akita
- NewYork-Presbyterian Hospital, New York, NY USA; Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA
| | - Anna Penn
- NewYork-Presbyterian Hospital, New York, NY USA; Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA
| | - Jordan S Orange
- NewYork-Presbyterian Hospital, New York, NY USA; Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA
| | - Lisa Saiman
- NewYork-Presbyterian Hospital, New York, NY USA; Division of Pediatric Infectious Diseases, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA
| | - Martha G Welch
- Division of Developmental Neuroscience, Psychiatry Department, Columbia University Irving Medical Center, New York, NY USA; Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY USA; Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA
| | - Cynthia Gyamfi-Bannerman
- NewYork-Presbyterian Hospital, New York, NY USA; Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY USA
| | - Melissa S Stockwell
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA; NewYork-Presbyterian Hospital, New York, NY USA; Department of Population and Family Health, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY USA
| | - Dani Dumitriu
- Division of Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY USA; Division of Developmental Neuroscience, Psychiatry Department, Columbia University Irving Medical Center, New York, NY USA; NewYork-Presbyterian Hospital, New York, NY USA; Sackler Institute, Zuckerman Institute, and the Columbia Population Research Center, Columbia University, New York, NY USA.
| |
Collapse
|
67
|
Ramadugu K, Bhaumik D, Luo T, Gicquelais RE, Lee KH, Stafford EB, Marrs CF, Neiswanger K, McNeil DW, Marazita ML, Foxman B. Maternal Oral Health Influences Infant Salivary Microbiome. J Dent Res 2020; 100:58-65. [PMID: 32859139 DOI: 10.1177/0022034520947665] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Oral microbiomes vary in cariogenic potential; these differences may be established early in life. A major concern is whether mothers transmit cariogenic bacteria to their children. Here we characterize early salivary microbiome development and the potential associations of that development with route of delivery, breastfeeding, and mother's oral health, and we evaluate transmission of microbes between mother and child. We analyzed saliva and metadata from the Center for Oral Health Research in Appalachia. For this cohort study, we sequenced the V6 region of the 16S rRNA gene and used quantitative polymerase chain reaction to detect Streptococcus mitis, Streptococcus sobrinus, Streptococcus mutans, Streptococcus oralis, and Candida albicans in the saliva from mothers and their infants, collected at 2, 9, and 12 mo (Pennsylvania site) and 2, 12, and 24 mo (West Virginia site). Breastfed children had lower relative abundances of Prevotella and Veillonella. If mothers had decayed, missing, or filled teeth, children had greater abundances of Veillonella and Actinomyces. There was little evidence of maternal transmission of selected microbes. At 12 mo, children's microbiomes were more similar to other children's than to their mothers'. Infants' salivary microbiomes became more adult-like with age but still differed with mothers' microbiomes at 12 mo. There was little evidence supporting transmission of selected microbes from mothers to children, but risk of colonization was associated with tooth emergence. Children are likely to acquire cariogenic bacteria from a variety of sources, including foods and contact with other children and adults.
Collapse
Affiliation(s)
- K Ramadugu
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - D Bhaumik
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - T Luo
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - R E Gicquelais
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - K H Lee
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - E B Stafford
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - C F Marrs
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - K Neiswanger
- Center for Craniofacial and Dental Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - D W McNeil
- Center for Oral Health Research in Appalachia, Department of Psychology, Department of Dental Practice and Rural Health, West Virginia University, Morgantown, WV, USA
| | - M L Marazita
- Center for Craniofacial and Dental Genetics, Department of Oral Biology, Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - B Foxman
- Center of Molecular and Clinical Epidemiology of Infectious Diseases, Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
68
|
Scazzocchio B, Minghetti L, D’Archivio M. Interaction between Gut Microbiota and Curcumin: A New Key of Understanding for the Health Effects of Curcumin. Nutrients 2020; 12:E2499. [PMID: 32824993 PMCID: PMC7551052 DOI: 10.3390/nu12092499] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/10/2020] [Accepted: 08/17/2020] [Indexed: 12/20/2022] Open
Abstract
Curcumin, a lipophilic polyphenol contained in the rhizome of Curcuma longa (turmeric), has been used for centuries in traditional Asian medicine, and nowadays it is widely used in food as dietary spice worldwide. It has received considerable attention for its pharmacological activities, which appear to act primarily through anti-inflammatory and antioxidant mechanisms. For this reason, it has been proposed as a tool for the management of many diseases, among which are gastrointestinal and neurological diseases, diabetes, and several types of cancer. However, the pharmacology of curcumin remains to be elucidated; indeed, a discrepancy exists between the well-documented in vitro and in vivo activities of curcumin and its poor bioavailability and chemical instability that should limit any therapeutic effect. Recently, it has been hypothesized that curcumin could exert direct regulative effects primarily in the gastrointestinal tract, where high concentrations of this polyphenol have been detected after oral administration. Consequently, it might be hypothesized that curcumin directly exerts its regulatory effects on the gut microbiota, thus explaining the paradox between its low systemic bioavailability and its wide pharmacological activities. It is well known that the microbiota has several important roles in human physiology, and its composition can be influenced by a multitude of environmental and lifestyle factors. Accordingly, any perturbations in gut microbiome profile or dysbiosis can have a key role in human disease progression. Interestingly, curcumin and its metabolites have been shown to influence the microbiota. It is worth noting that from the interaction between curcumin and microbiota two different phenomena arise: the regulation of intestinal microflora by curcumin and the biotransformation of curcumin by gut microbiota, both of them potentially crucial for curcumin activity. This review summarizes the most recent studies on this topic, highlighting the strong connection between curcumin and gut microbiota, with the final aim of adding new insight into the potential mechanisms by which curcumin exerts its effects.
Collapse
Affiliation(s)
- Beatrice Scazzocchio
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Luisa Minghetti
- Research Coordination and Support Service, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Massimo D’Archivio
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy;
| |
Collapse
|
69
|
Hoffman KW, Lee JJ, Corcoran CM, Kimhy D, Kranz TM, Malaspina D. Considering the Microbiome in Stress-Related and Neurodevelopmental Trajectories to Schizophrenia. Front Psychiatry 2020; 11:629. [PMID: 32719625 PMCID: PMC7350783 DOI: 10.3389/fpsyt.2020.00629] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Early life adversity and prenatal stress are consistently associated with an increased risk for schizophrenia, although the exact pathogenic mechanisms linking the exposures with the disease remain elusive. Our previous view of the HPA stress axis as an elegant but simple negative feedback loop, orchestrating adaptation to stressors among the hypothalamus, pituitary, and adrenal glands, needs to be updated. Research in the last two decades shows that important bidirectional signaling between the HPA axis and intestinal mucosa modulates brain function and neurochemistry, including effects on glucocorticoid hormones and brain-derived neurotrophic factor (BDNF). The intestinal microbiome in earliest life, which is seeded by the vaginal microbiome during delivery, programs the development of the HPA axis in a critical developmental window, determining stress sensitivity and HPA function as well as immune system development. The crosstalk between the HPA and the Microbiome Gut Brain Axis (MGBA) is particularly high in the hippocampus, the most consistently disrupted neural region in persons with schizophrenia. Animal models suggest that the MGBA remains influential on behavior and physiology across developmental stages, including the perinatal window, early childhood, adolescence, and young adulthood. Understanding the role of the microbiome on critical risk related stressors may enhance or transform of understanding of the origins of schizophrenia and offer new approaches to increase resilience against stress effects for preventing and treating schizophrenia.
Collapse
Affiliation(s)
- Kevin W. Hoffman
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jakleen J. Lee
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Cheryl M. Corcoran
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- James J. Peters VA Medical Center, Mental Illness Research, Education and Clinical Centers (MIRECC), New York, NY, United States
| | - David Kimhy
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- James J. Peters VA Medical Center, Mental Illness Research, Education and Clinical Centers (MIRECC), New York, NY, United States
| | - Thorsten M. Kranz
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Dolores Malaspina
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
70
|
Nandakumar V, Aly H. Microbiota and chronic lung disease in preterm infants. Where is the truth? J Perinatol 2020; 40:983-984. [PMID: 32277163 DOI: 10.1038/s41372-020-0666-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/11/2020] [Accepted: 03/31/2020] [Indexed: 11/10/2022]
Affiliation(s)
| | - Hany Aly
- Department of Neonatology, Cleveland Clinic Children's, Cleveland, OH, USA.
| |
Collapse
|
71
|
Richards M, Ferber J, Li DK, Darrow LA. Cesarean delivery and the risk of allergic rhinitis in children. Ann Allergy Asthma Immunol 2020; 125:280-286.e5. [PMID: 32387533 DOI: 10.1016/j.anai.2020.04.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND Cesarean delivery (C-section) may influence the infant microbiome and affect immune system development and subsequent risk for allergic rhinitis (AR). OBJECTIVE To investigate the association between C-section and AR at ages 6, 8, and 10 years. METHODS Data were collected prospectively through Kaiser Permanente Northern Californias (KPNC) integrated healthcare system. Children were eligible if they were born in a KPNC hospital and remained in the KPNC system for minimum 6 years (n = 117,768 age 6; n = 75,115 age 8; n = 40,332 age 10). Risk ratios (RR) for C-section and AR were estimated at each follow-up age and adjusted for important covariates, including intrapartum antibiotics, pre-pregnancy body mass index, maternal allergic morbidities, and breastfeeding. Subanalyses considered information on C-section indication, labor, and membrane rupture. RESULTS After adjusting for confounders, we did not observe an association between C-section and AR at follow-up ages 6, 8, or 10 years (RR [CI]: 6 years, 0.98 [0.91, 1.04]; 8 years, 1.00 [0.95, 1.07]; 10 years, 1.03 [0.96, 1.10]). In stratified analyses, there was limited evidence that C-section increases the risk of AR in certain subgroups (eg, children of non-atopic mothers, second or higher birth order children), but most estimated risk ratios were consistent with no association. Estimated associations were unaffected by participant attrition, missing data, or intrapartum antibiotics. CONCLUSION C-section delivery was not associated with AR at follow-up ages of 6, 8, or 10 years in a large contemporary US cohort.
Collapse
Affiliation(s)
- Megan Richards
- School of Community Health Sciences, University of Nevada, Reno, Nevada.
| | - Jeannette Ferber
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - De-Kun Li
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Lyndsey A Darrow
- School of Community Health Sciences, University of Nevada, Reno, Nevada
| |
Collapse
|
72
|
Tang Q, Jin G, Wang G, Liu T, Liu X, Wang B, Cao H. Current Sampling Methods for Gut Microbiota: A Call for More Precise Devices. Front Cell Infect Microbiol 2020; 10:151. [PMID: 32328469 PMCID: PMC7161087 DOI: 10.3389/fcimb.2020.00151] [Citation(s) in RCA: 259] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 03/23/2020] [Indexed: 12/14/2022] Open
Abstract
The development of next-generation sequencing technology has enabled researchers to explore and understand the gut microbiome from a broader and deeper perspective. However, the results of different studies on gut microbiota are highly variable even in the same disease, which makes it difficult to guide clinical diagnosis and treatment. The ideal sampling method should be non-invasive, involve little cross-contamination or bowel preparation, and collect gut microbiota at different sites. Currently, sequencing technologies are usually based on samples collected from feces, mucosal biopsy, intestinal fluid, etc. However, different parts of the gastrointestinal tract possess various physiological characteristics that are essential for particular species of living microbiota. Moreover, current sampling methods are somewhat defective. For example, fecal samples are just a proxy for intestinal microbiota, while biopsies are invasive for patients and not suitable for healthy controls. In this review, we summarize the current sampling methods and their advantages and shortcomings. New sampling technologies, such as the Brisbane Aseptic Biopsy Device and the intelligent capsule, are also mentioned to inspire the development of future precise description methods of the gut microbiome.
Collapse
Affiliation(s)
- Qiang Tang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Tianjin Institute of Digestive Disease, General Hospital, Tianjin Medical University, Tianjin, China
| | - Ge Jin
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Tianjin Institute of Digestive Disease, General Hospital, Tianjin Medical University, Tianjin, China
| | - Gang Wang
- Tianjin Institute of Digestive Disease, General Hospital, Tianjin Medical University, Tianjin, China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Tianjin Institute of Digestive Disease, General Hospital, Tianjin Medical University, Tianjin, China
| | - Xiang Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Tianjin Institute of Digestive Disease, General Hospital, Tianjin Medical University, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China.,Tianjin Institute of Digestive Disease, General Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
73
|
Is the Newborn Microbiome Disrupted by Routine Newborn Suctioning? An Exploratory Approach for Policy Development. J Perinat Neonatal Nurs 2020; 34:231-238. [PMID: 32697543 DOI: 10.1097/jpn.0000000000000499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
An invasive procedure commonly used in the perinatal setting is newborn suctioning at birth. Routine newborn suctioning, without indication, is not recommended by the American Academy of Pediatrics, the American Heart Association, and the European Resuscitation Council for the newborn with spontaneous respirations, adequate crying, and good muscle tone. Whether to suction a newborn is a difficult practice decision made daily by the perinatal nurse. A primary element of this practice decision is the consideration that newborn suctioning has the potential to affect health outcomes. Furthermore, routine newborn suctioning may be disrupting the newborn microbiome by removing commensal bacteria and potentially, negatively affecting newborn health. This article will explore the connection between the practice of routine newborn suctioning in the perinatal setting and the potential to disrupt the newborn microbiome. The methods employed and reported in this article consisted of review of literature and review of clinical guidelines and a descriptive study to determine the scope of practice of newborn suctioning. The premise of whether the practice of routine newborn suctioning at birth may be disrupting the newborn microbiome will also be considered. Finally, developing a microbiome-centric perspective will be explored.
Collapse
|
74
|
Laboratory Analysis Techniques for the Perinatal Microbiome: Implications for Studies of Probiotic Interventions. J Perinat Neonatal Nurs 2020; 34:239-250. [PMID: 32697544 PMCID: PMC9528808 DOI: 10.1097/jpn.0000000000000496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The microbiome is composed of many organisms and is impacted by an intricate exchange between genetics and environmental factors. The perinatal microbiome influences both the developing fetus and the pregnant person. The purpose of this article is to describe the tests that are currently available for laboratory analysis of the perinatal microbiome in relationship to probiotic interventions. This article focuses on the bacterial component of the microbiome. Although adverse outcomes associated with the perinatal microbiome have been studied, a comprehensive understanding of the physiologic perinatal microbiome is still emerging. Early efforts to influence the perinatal microbiome through probiotics are currently under investigation. Unique terminology is defined, and the microbial composition of perinatal microbiota is summarized. The outcomes of studies of antenatal probiotics are summarized. Microbiome testing and analysis are defined and compared. Implications for perinatal care and probiotics research are presented.
Collapse
|
75
|
Haikal C, Chen QQ, Li JY. Microbiome changes: an indicator of Parkinson's disease? Transl Neurodegener 2019; 8:38. [PMID: 31890161 PMCID: PMC6929290 DOI: 10.1186/s40035-019-0175-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/16/2019] [Indexed: 12/20/2022] Open
Abstract
Parkinson’s disease is characterized by dopaminergic neuron loss and intracellular inclusions composed mainly of alpha synuclein (α-syn), but the mechanism of pathogenesis is still obscure. In recent years, more attention has been given to the gut as a key player in the initiation and progression of PD pathology. Several studies characterizing changes in the microbiome, particularly the gut microbiome, have been conducted. Although many studies found a decrease in the bacterial family Prevotellaceae and in butyrate-producing bacterial genera such as Roseburia and Faecalibacteria, and an increase in the genera Akkermansia many of the studies reported contradictory findings. In this review, we highlight the findings from the different studies and reflect on the future of microbiome studies in PD research.
Collapse
Affiliation(s)
- Caroline Haikal
- 1Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, 221 84 Lund, Sweden
| | - Qian-Qian Chen
- 2Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, Liaoning China
| | - Jia-Yi Li
- 1Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, BMC A10, 221 84 Lund, Sweden.,3Institute of Health Sciences, China Medical University, Shenyang, 110112 China
| |
Collapse
|
76
|
Van Daele E, Knol J, Belzer C. Microbial transmission from mother to child: improving infant intestinal microbiota development by identifying the obstacles. Crit Rev Microbiol 2019; 45:613-648. [DOI: 10.1080/1040841x.2019.1680601] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Emmy Van Daele
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Jan Knol
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Gut Biology and Microbiology, Danone Nutricia Research, Utrecht, The Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
77
|
Roman P, Cardona D, Sempere L, Carvajal F. Microbiota and organophosphates. Neurotoxicology 2019; 75:200-208. [PMID: 31560873 DOI: 10.1016/j.neuro.2019.09.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 09/22/2019] [Accepted: 09/22/2019] [Indexed: 02/08/2023]
Abstract
Organophosphates (OPs) are important toxic compounds commonly used for a variety of purposes in agriculture, industry and household settings. Consumption of these compounds affects several central nervous system functions. Some of the most recognised consequences of organophosphate pesticide exposure in humans include neonatal developmental abnormalities, endocrine disruption, neurodegeneration, neuroinflammation and cancer. In addition, neurobehavioral and emotional deficits following OP exposure have been reported. It would be of great value to discover a therapeutic strategy which produces a protective effect against these neurotoxic compounds. Moreover, a growing body of preclinical data suggests that the microbiota may affect metabolism and neurotoxic outcomes through exposure to OPs. The human gut is colonised by a broad variety of microorganisms. This huge number of bacteria and other microorganisms which survive by colonising the gastrointestinal tract is defined as "gut microbiota". The gut microbiome plays a profound role in metabolic processing, energy production, immune and cognitive development and homeostasis. The effects are not only localized in the gut, but also influence many other organs, such as the brain through the microbiome-gut-brain axis. Therefore, given the gut microbiota's key role in host homeostasis, this microbiota may be altered or modified temporarily by factors such as antibiotics, diet and toxins such as pesticides. The aim of this review is to examine scientific articles concerning the impact of microbiota in OP toxicity. Studies focussed on the possible contribution the microbiota has on variable host pharmacokinetic responses such as absorption and biotransformation of xenobiotics will be evaluated. Microbiome manipulation by antibiotic or probiotic administration and faecal transplantation are experimental approaches recently proposed as treatments for several diseases. Finally, microbiota manipulation as a possible therapeutic strategy in order to reduce OP toxicity will be discussed.
Collapse
Affiliation(s)
- Pablo Roman
- Departamento de Enfermería, Fisioterapia y Medicina, Universidad de Almería, La Cañada, 04120 Almería, Spain; Health Research Center, University of Almería, Spain; Health Sciences Research Group (CTS-451), University of Almería, Spain
| | - Diana Cardona
- Departamento de Enfermería, Fisioterapia y Medicina, Universidad de Almería, La Cañada, 04120 Almería, Spain; Health Research Center, University of Almería, Spain; Research Center for Agricultural and Food Biotechnology BITAL, Universidad de Almería, Spain.
| | - Lluis Sempere
- NeuroCritical Care Unit, Virgen del Rocio University Hospital, IBIS/CSIC/University of Seville, Spain
| | - Francisca Carvajal
- Departamento de Psicología, Universidad de Almería, La Cañada, 04120 Almería, Spain; Health Research Center, University of Almería, Spain
| |
Collapse
|
78
|
Reza MM, Finlay BB, Pettersson S. Gut microbes, ageing & organ function: a chameleon in modern biology? EMBO Mol Med 2019; 11:e9872. [PMID: 31410991 PMCID: PMC6728600 DOI: 10.15252/emmm.201809872] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 05/27/2019] [Accepted: 06/25/2019] [Indexed: 12/15/2022] Open
Abstract
All species, including humans, are cohabited by a myriad of microbial species, which massively influences body function in a diet‐, exercise‐ and age‐dependent manner. The microbiome composition differs between individuals, partly due to the polymorphic immune system, as well as the environment, making the microbe–host interplay unique in each one of us. Ageing is a gradual loss of function in part due to reduced repair mechanisms and accumulation of tissue damage through mechanisms largely unknown. Accumulating evidence suggests that our indigenous microbes, a known major regulator of human physiology, are also connected to regulate the ageing process through signalling pathways and metabolites though the biological mechanisms are unknown. At an ageing meeting in Singapore in 2018, investigators discussed the current understanding of microbe regulation and its impact on healthy ageing. This review summarizes the highlights from the meeting and conveys some of the new ideas that emerged around gut microbes and the biology of ageing. While highly speculative, an idea emerged in which gut microbes constantly respond and evolve to environmental cues, as part of an ageing process, thus serving as a second messenger to support and attenuate organ decline in a diet‐, gender‐ and age‐dependent manner.
Collapse
Affiliation(s)
- Musarrat Maisha Reza
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden.,School of Biological Sciences, Nanyang Technological University, Singapore City, Singapore
| | - B Brett Finlay
- Michael Smith Laboratories and the Departments of Biochemistry and Molecular Biology, and Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Sven Pettersson
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore City, Singapore.,Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore City, Singapore
| |
Collapse
|
79
|
|