101
|
Miranda-Comas G, Petering RC, Zaman N, Chang R. Implications of the Gut Microbiome in Sports. Sports Health 2022; 14:894-898. [PMID: 35034531 PMCID: PMC9631033 DOI: 10.1177/19417381211060006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
CONTEXT Two-thirds of an individual's gut microbiota is unique and influenced by dietary and exercise habits, age, sex, genetics, ethnicity, antibiotics, health, and disease. It plays important roles in nutrient and vitamin metabolism, inflammatory modulation, immune system function, and overall health of an individual. Specifically, in sports it may help decrease recovery time and improve athletic performance. EVIDENCE ACQUISITION PubMed and Medline databases were used for the literature search. Bibliographies based on the original search were utilized to pursue further literature search. STUDY DESIGN Clinical review. LEVEL OF EVIDENCE Level 4. RESULTS Diet and exercise play very important roles in the composition of the gut microbiota in the athletic and nonathletic individual. Ingestion of carbohydrates during and after exercise seems to have an anti-inflammatory effect postexercise. Supplementation with probiotic seems to aid in recovery after exercise, too, especially restoring the "normal" gut microbiota. Physically active individuals of all levels have more alpha diversity and "health-promoting gut species" in their microbiome than nonactive individuals, along with higher concentrations of short-chain fatty acids (SCFA) and SCFA-producing organisms. However, exercise interventions should be longer than 8 weeks to see these positive characteristics. Immune function is highly influenced by the gut microbiota's response to exercise. A transient immune dysfunction occurs after prolonged high-intensity exercise, which correlates with microbiota dysregulation. Nevertheless, long-term exposure to exercise will enhance the immune response and lead to positive changes in the gut microbiota. CONCLUSION Although the exact mechanisms of the effects that diet, exercise, and genetics have on the gut microbiota remain largely unknown, there is evidence that suggests overall health benefits. In the athletic population, these benefits can ultimately lead to performance improvement.
Collapse
Affiliation(s)
- Gerardo Miranda-Comas
- Department of Rehabilitation and
Human Performance, Icahn School of Medicine at Mount Sinai, New York, New
York,Gerardo
Miranda-Comas, MD, 1510 Ashford Avenue, Apartment 802, San Juan, PR
00911 ()
(Twitter: @SportsMDgmirand)
| | - Ryan C. Petering
- Department of Family Medicine,
Oregon Health & Science University School of Medicine, Portland,
Oregon
| | - Nadia Zaman
- PM&R, Tufts University School
of Medicine, Boston, Massachusetts
| | - Richard Chang
- Department of Rehabilitation and
Human Performance, Icahn School of Medicine at Mount Sinai, New York, New
York
| |
Collapse
|
102
|
Nearing JT, Douglas GM, Hayes MG, MacDonald J, Desai DK, Allward N, Jones CMA, Wright RJ, Dhanani AS, Comeau AM, Langille MGI. Microbiome differential abundance methods produce different results across 38 datasets. Nat Commun 2022; 13:342. [PMID: 35039521 PMCID: PMC8763921 DOI: 10.1038/s41467-022-28034-z] [Citation(s) in RCA: 350] [Impact Index Per Article: 116.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
Identifying differentially abundant microbes is a common goal of microbiome studies. Multiple methods are used interchangeably for this purpose in the literature. Yet, there are few large-scale studies systematically exploring the appropriateness of using these tools interchangeably, and the scale and significance of the differences between them. Here, we compare the performance of 14 differential abundance testing methods on 38 16S rRNA gene datasets with two sample groups. We test for differences in amplicon sequence variants and operational taxonomic units (ASVs) between these groups. Our findings confirm that these tools identified drastically different numbers and sets of significant ASVs, and that results depend on data pre-processing. For many tools the number of features identified correlate with aspects of the data, such as sample size, sequencing depth, and effect size of community differences. ALDEx2 and ANCOM-II produce the most consistent results across studies and agree best with the intersect of results from different approaches. Nevertheless, we recommend that researchers should use a consensus approach based on multiple differential abundance methods to help ensure robust biological interpretations. Many microbiome differential abundance methods are available, but it lacks systematic comparison among them. Here, the authors compare the performance of 14 differential abundance testing methods on 38 16S rRNA gene datasets with two sample groups, and show ALDEx2 and ANCOM-II produce the most consistent results.
Collapse
Affiliation(s)
- Jacob T Nearing
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada.
| | - Gavin M Douglas
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Molly G Hayes
- Department of Mathematics and Statistics, Dalhousie University, Halifax, NS, Canada
| | - Jocelyn MacDonald
- Department of Computer Science, Dalhousie University, Halifax, NS, Canada
| | - Dhwani K Desai
- Integrated Microbiome Resource, Dalhousie University, Halifax, NS, Canada
| | - Nicole Allward
- Department of Civil and Resource Engineering, Dalhousie University, Halifax, NS, Canada
| | - Casey M A Jones
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Robyn J Wright
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Akhilesh S Dhanani
- Integrated Microbiome Resource, Dalhousie University, Halifax, NS, Canada
| | - André M Comeau
- Integrated Microbiome Resource, Dalhousie University, Halifax, NS, Canada
| | - Morgan G I Langille
- Integrated Microbiome Resource, Dalhousie University, Halifax, NS, Canada.,Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
103
|
Aguilar-Lopez M, Dinsmoor AM, Ho TTB, Donovan SM. A systematic review of the factors influencing microbial colonization of the preterm infant gut. Gut Microbes 2022; 13:1-33. [PMID: 33818293 PMCID: PMC8023245 DOI: 10.1080/19490976.2021.1884514] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Prematurity coupled with the necessary clinical management of preterm (PT) infants introduces multiple factors that can interfere with microbial colonization. This study aimed to review the perinatal, physiological, pharmacological, dietary, and environmental factors associated with gut microbiota of PT infants. A total of 587 articles were retrieved from a search of multiple databases. Sixty studies were included in the review after removing duplicates and articles that did not meet the inclusion criteria. Review of this literature revealed that evidence converged on the effect of postnatal age, mode of delivery, use of antibiotics, and consumption of human milk in the composition of gut microbiota of PT infants. Less evidence was found for associations with race, sex, use of different fortifiers, macronutrients, and other medications. Future studies with rich metadata are needed to further explore the impact of the PT exposome on the development of the microbiota in this high-risk population.
Collapse
Affiliation(s)
- Miriam Aguilar-Lopez
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, USA
| | - Andrew M. Dinsmoor
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, USA
| | - Thao T. B. Ho
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, USA
| | - Sharon M. Donovan
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, USA,Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, USA,CONTACT Sharon M. Donovan Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, 339 Bevier Hall 905 S. Goodwin Avenue, Urbana, IL61801, USA
| |
Collapse
|
104
|
Tu J, Wang Y, Jin L, Huang W. Bile acids, gut microbiota and metabolic surgery. Front Endocrinol (Lausanne) 2022; 13:929530. [PMID: 36072923 PMCID: PMC9441571 DOI: 10.3389/fendo.2022.929530] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Metabolic surgery, or bariatric surgery, is currently the most effective approach for treating obesity and its complications. Vertical sleeve gastrectomy (VSG) and Roux-en-Y gastric bypass (RYGB) are the top two types of commonly performed metabolic surgery now. The precise mechanisms of how the surgeries work are still unclear, therefore much research has been conducted in this area. Gut hormones such as GLP-1 and PYY have been studied extensively in the context of metabolic surgery because they both participate in satiety and glucose homeostasis. Bile acids, whose functions cover intestinal lipid absorption and various aspects of metabolic regulation via the action of FXR, TGR5, and other bile acid receptors, have also been actively investigated as potential mediators of metabolic surgery. Additionally, gut microbiota and their metabolites have also been studied because they can affect metabolic health. The current review summarizes and compares the recent scientific progress made on identifying the mechanisms of RYGB and VSG. One of the long-term goals of metabolic/bariatric surgery research is to develop new pharmacotherapeutic options for the treatment of obesity and diabetes. Because obesity is a growing health concern worldwide, there is a dire need in developing novel non-invasive treatment options.
Collapse
Affiliation(s)
- Jui Tu
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
- Irell & Manella Graduate School of Biomedical Science, City of Hope National Medical Center, Duarte, CA, United States
| | - Yangmeng Wang
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Lihua Jin
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
- Irell & Manella Graduate School of Biomedical Science, City of Hope National Medical Center, Duarte, CA, United States
- *Correspondence: Wendong Huang,
| |
Collapse
|
105
|
Peeters J, Thas O, Shkedy Z, Kodalci L, Musisi C, Owokotomo OE, Dyczko A, Hamad I, Vangronsveld J, Kleinewietfeld M, Thijs S, Aerts J. Exploring the Microbiome Analysis and Visualization Landscape. FRONTIERS IN BIOINFORMATICS 2021; 1:774631. [PMID: 36303773 PMCID: PMC9580862 DOI: 10.3389/fbinf.2021.774631] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 10/29/2021] [Indexed: 02/02/2023] Open
Abstract
Research on the microbiome has boomed recently, which resulted in a wide range of tools, packages, and algorithms to analyze microbiome data. Here we investigate and map currently existing tools that can be used to perform visual analysis on the microbiome, and associate the including methods, visual representations and data features to the research objectives currently of interest in microbiome research. The analysis is based on a combination of a literature review and workshops including a group of domain experts. Both the reviewing process and workshops are based on domain characterization methods to facilitate communication and collaboration between researchers from different disciplines. We identify several research questions related to microbiomes, and describe how different analysis methods and visualizations help in tackling them.
Collapse
Affiliation(s)
- Jannes Peeters
- CENSTAT, Data Science Institute (DSI), Hasselt University, Diepenbeek, Belgium
- *Correspondence: Jannes Peeters ,
| | - Olivier Thas
- CENSTAT, Data Science Institute (DSI), Hasselt University, Diepenbeek, Belgium
| | - Ziv Shkedy
- CENSTAT, Data Science Institute (DSI), Hasselt University, Diepenbeek, Belgium
| | - Leyla Kodalci
- CENSTAT, Data Science Institute (DSI), Hasselt University, Diepenbeek, Belgium
| | - Connie Musisi
- CENSTAT, Data Science Institute (DSI), Hasselt University, Diepenbeek, Belgium
| | | | - Aleksandra Dyczko
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), Hasselt University, Diepenbeek, Belgium
- Department of Immunology and Infection, Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| | - Ibrahim Hamad
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), Hasselt University, Diepenbeek, Belgium
- Department of Immunology and Infection, Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| | - Jaco Vangronsveld
- Center for Environmental Sciences, Environmental Biology, Hasselt University, Diepenbeek, Belgium
- Department of Plant Physiology and Biophysics, Faculty of Biology and Biotechnology, Maria Curie–Skłodowska University, Lublin, Poland
| | - Markus Kleinewietfeld
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), Hasselt University, Diepenbeek, Belgium
- Department of Immunology and Infection, Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| | - Sofie Thijs
- Center for Environmental Sciences, Environmental Biology, Hasselt University, Diepenbeek, Belgium
| | - Jan Aerts
- CENSTAT, Data Science Institute (DSI), Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
106
|
Teaw S, Hinchcliff M, Cheng M. A review and roadmap of the skin, lung and gut microbiota in systemic sclerosis. Rheumatology (Oxford) 2021; 60:5498-5508. [PMID: 33734316 PMCID: PMC8643452 DOI: 10.1093/rheumatology/keab262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 11/12/2022] Open
Abstract
As our understanding of the genetic underpinnings of SSc increases, questions regarding the environmental trigger(s) that induce and propagate SSc in the genetically predisposed individual emerge. The interplay between the environment, the immune system, and the microbial species that inhabit the patient's skin and gastrointestinal tract is a pathobiological frontier that is largely unexplored in SSc. The purpose of this review is to provide an overview of the methodologies, experimental study results and future roadmap for elucidating the relationship between the SSc host and his/her microbiome.
Collapse
Affiliation(s)
- Shannon Teaw
- Yale School of Medicine, Department of Medicine Section of Rheumatology, Allergy & Immunology, New Haven, CT, USA
| | - Monique Hinchcliff
- Yale School of Medicine, Department of Medicine Section of Rheumatology, Allergy & Immunology, New Haven, CT, USA
| | - Michelle Cheng
- Yale School of Medicine, Department of Medicine Section of Rheumatology, Allergy & Immunology, New Haven, CT, USA
| |
Collapse
|
107
|
Oral microbiome associated with lymph node metastasis in oral squamous cell carcinoma. Sci Rep 2021; 11:23176. [PMID: 34848792 PMCID: PMC8633319 DOI: 10.1038/s41598-021-02638-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/19/2021] [Indexed: 01/09/2023] Open
Abstract
Oral microbiota can alter cancer susceptibility and progression by modulating metabolism and inflammation. We assessed the association between the oral microbiome and lymph node (LN) metastasis in oral squamous cell carcinoma (OSCC). We collected a total of 54 saliva samples from patients with OSCC before surgery. LN metastasis was assessed based on postoperative pathological examination. We used QIIME2, linear discriminant analysis effect size (LEfSe), and PICRUSt2 methods to analyze microbial dysbiosis. A random forest classifier was used to assess whether the oral microbiome could predict LN metastasis. Among the 54 OSCC samples, 20 had LN metastasis, and 34 had no evidence of metastasis. There was a significant difference in β-diversity between the metastasis and no metastasis groups. Through LEfSe analysis, the metastasis group was enriched in the genera Prevotella, Stomatobaculum, Bifidobacterium, Peptostreptococcaceae, Shuttleworthia and Finegoldia. Pathways related to signal peptidase II were predominant in the no metastasis group. The RF model showed a modestly high accuracy for predicting metastasis. Differences in microbial community composition and functions were observed in the oral microbiome of patients with OSCC with and without LN metastasis. However, the finding that specific taxa may be associated with LN metastasis should be verified in a further prospective study.
Collapse
|
108
|
Fischer JAJ, Karakochuk CD. Feasibility of an At-Home Adult Stool Specimen Collection Method in Rural Cambodia. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182312430. [PMID: 34886156 PMCID: PMC8656988 DOI: 10.3390/ijerph182312430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 11/22/2022]
Abstract
The human microbiome has received significant attention over the past decade regarding its potential impact on health. Epidemiological and intervention studies often rely on at-home stool collection methods designed for high-resource settings, such as access to an improved toilet with a modern toilet seat. However, this is not always appropriate or applicable to low-resource settings. Therefore, the design of a user-friendly stool collection kit for low-resource rural settings is needed. We describe the development, assembly, and user experience of a simple and low-cost at-home stool collection kit for women living in rural Cambodia as part of a randomized controlled trial in 2020. Participants were provided with the stool collection kit and detailed verbal instruction. Enrolled women (n = 480) provided two stool specimens (at the start of the trial and after 12 weeks) at their home and brought them to the health centre that morning in a sterile collection container. User specimen collection compliance was high, with 90% (n = 434) of women providing a stool specimen at the end of the trial (after 12 weeks). This feasible and straightforward method has strong potential for similar or adapted use among adults residing in other rural or low-resource contexts.
Collapse
Affiliation(s)
- Jordie A. J. Fischer
- Food, Nutrition, and Health, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada;
- Healthy Starts, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Crystal D. Karakochuk
- Food, Nutrition, and Health, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada;
- Healthy Starts, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Correspondence:
| |
Collapse
|
109
|
Di Pierro F. Gut Microbiota Parameters Potentially Useful in Clinical Perspective. Microorganisms 2021; 9:microorganisms9112402. [PMID: 34835527 PMCID: PMC8623243 DOI: 10.3390/microorganisms9112402] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022] Open
Abstract
Interest in gut microbiota analyses is at an all-time high. Gut microbiota is thought to relate to an increasing range of diseases of interest to physicians and nutritionists. Overweight, obesity, response to diet, metabolic syndrome, low grade inflammation, diabetes and colon neoplasms could maybe be observed in microbiota if affordable markers were available. Possible biomarkers like the Firmicutes/Bacteroidetes ratio, the Gram-positive/Gram-negative ratio, the Prevotella/Bacteroides ratio, and the Fusobacterium nucleatum/Faecalibacterium prausnitzii ratio are here reviewed in a narrative way in the attempt to highlight their possible future role in routine practice and clinically relevant diagnostics.
Collapse
Affiliation(s)
- Francesco Di Pierro
- Digestive Endoscopy & Gastroenterology, Fondazione Poliambulanza, 25124 Brescia, Italy;
- UNICAM, Camerino University, 62032 Camerino, Italy
- Scientific Department, Velleja Research, 20124 Milan, Italy
| |
Collapse
|
110
|
Yau YY, Wasinger VC, Hirten RP, Chuang E, Huntsman M, Stylli J, Shimizu J, Yajnik V, Smith J, Lee SN, Singh S, Wahl C, Leong RW, Sands BE. Current Trends in IBD-Development of Mucosal-Based Biomarkers and a Novel Minimally Invasive Recoverable Sampling System. Inflamm Bowel Dis 2021; 27:S17-S24. [PMID: 34791290 PMCID: PMC9214562 DOI: 10.1093/ibd/izab179] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Indexed: 12/16/2022]
Abstract
Despite recent developments in therapy for inflammatory bowel diseases (IBDs), there have been limited advances in diagnostic tools available to aid in disease management. A growing body of evidence suggests that there are important host-microbe interactions at the mucosal interface that modulate the inflammatory response in patients with IBD. Additionally, the importance of mucosal integrity and its disruption appears to be important in the pathophysiology and perpetuation of the disease. The ability to characterize this interface may provide valuable information for both disease monitoring and identification of new treatment targets. Endoscopy remains the primary tool for disease monitoring, and mucosal healing is the primary therapeutic target in IBD treatment. However, establishing mucosal healing requires repetitive endoscopic procedures, and endoscopy is limited by factors such as invasiveness, cost, and risk of adverse events. Moreover, the use of a bowel preparation for colonoscopies alters the mucus layer and thus perturbs evaluation of the host-microbe interaction. Stool sampling may also be inaccurate because it reflects the end state of metabolites and proteins, failing to take into account the degradation or alteration of substrates of interest by bacterial proteases and other enzymes during passage through the colon. A novel sampling capsule, called the Recoverable Sampling System (RSS), is being developed as a complementary tool to colonoscopy. The RSS is intended to be a platform for noninvasive autonomous sampling, preservation, handling, and storage of analytes of interest found in the gastrointestinal fluids. A proprietary preservative contained within the chambers of the capsule has been developed to stabilize DNA and proteins for ex vivo microbiome and metabolomics analyses. Surrogate markers such as SPP24 and GUCA2a have been identified to correlate with gut health, intestinal permeability, and inflammation and could be locally sampled by the RSS. The potential clinical utility of an RSS device is broad and would likely be able to guide therapy by allowing for more frequent disease monitoring, aiding in disease characterization, and facilitating in the identification of novel therapeutic targets.
Collapse
Affiliation(s)
| | | | - Robert P Hirten
- Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emil Chuang
- Address correspondence to: Emil Chuang, MB, BS, Progenity, Inc, San Diego, CA, USA ()
| | | | - Jack Stylli
- Georgetown University, School of Medicine, Washington D.C., USA
| | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Abstract
Several human intestinal microbiota studies suggest that bacteriophages, viruses infecting bacteria, play a role in gut homeostasis. Currently, bacteriophages are considered a tool to precisely engineer the intestinal microbiota, but they have also attracted considerable attention as a possible solution to fight against bacterial pathogens resistant to antibiotics. These two applications necessitate bacteriophages to reach and kill their bacterial target within the gut environment. Unfortunately, exploitable clinical data in this field are scarce. Here, we review the administration of bacteriophages to target intestinal bacteria in mammalian experimental models. While bacteriophage amplification in the gut was often confirmed, we found that in most studies, it had no significant impact on the load of the targeted bacteria. In particular, we observed that the outcome of bacteriophage treatments is linked to the behavior of the target bacteria toward each animal model. Treatment efficacy ranges from poor in asymptomatic intestinal carriage to high in intestinal disease. This broad range of efficacy underlines the difficulties to reach a consensus on the impact of bacteriophages in the gut and calls for deeper investigations of key parameters that influence the success of such interventions before launching clinical trials.
Collapse
|
112
|
Sun J, Huang T, Debelius JW, Fang F. Gut microbiome and amyotrophic lateral sclerosis: A systematic review of current evidence. J Intern Med 2021; 290:758-788. [PMID: 34080741 DOI: 10.1111/joim.13336] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS), characterized by a loss of motor neurons in the brain and spinal cord, is a relatively rare but currently incurable neurodegenerative disease. The global incidence of ALS is estimated as 1.75 per 100,000 person-years and the global prevalence is estimated as 4.1-8.4 per 100,000 individuals. Contributions from outside the central nervous system to the etiology of ALS have been increasingly recognized. Gut microbiome is one of the most quickly growing fields of research for ALS. In this article, we performed a comprehensive review of the results from existing animal and human studies, to provide an up-to-date summary of the current research on gut microbiome and ALS. In brief, we found relatively consistent results from animal studies, suggesting an altered gut microbiome composition in experimental ALS. Publication bias might however be a concern. Findings from human studies are largely inconclusive. A few animal and human studies demonstrated the usefulness of intervention with microbial-derived metabolites in modulating the disease progression of ALS. We discussed potential methodological concerns in these studies, including study design, statistical power, handling process of biospecimens and sequencing data, as well as statistical methods and interpretation of results. Finally, we made a few proposals for continued microbiome research in ALS, with the aim to provide valid, reproducible, and translatable findings.
Collapse
Affiliation(s)
- Jiangwei Sun
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tingting Huang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Justine W Debelius
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Fang Fang
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
113
|
Abdullatif VA, Sur RL, Eshaghian E, Gaura KA, Goldman B, Panchatsharam PK, Williams NJ, Abbott JE. Efficacy of Probiotics as Prophylaxis for Urinary Tract Infections in Premenopausal Women: A Systematic Review and Meta-Analysis. Cureus 2021; 13:e18843. [PMID: 34671514 PMCID: PMC8523083 DOI: 10.7759/cureus.18843] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2021] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION Although antibiotic therapy has been the mainstay of prophylaxis and treatment of urinary tract infections (UTIs), antibacterial resistance has led to increased incidence of infections and healthcare spending in both community-acquired and nosocomial UTIs. This has led to an active exploration of alternative remedies for both the prophylaxis and treatment of UTIs, especially in women with recurrent urinary tract infections. Probiotic supplementation is one novel intervention that has been studied as a prophylactic measure in patients with UTIs. The current systematic review and meta-analysis was conducted to evaluate the efficacy of probiotics for prophylaxis in UTIs in premenopausal women. METHODS Detailed search strategies for each electronic database were developed for PubMed, EMBASE, and Scopus to identify relevant literature published between 2001-2021. RevMan 5.3 statistical software was used to analyze data in studies. The random-effects model was used for pooling the data. The risk of bias and study quality were assessed using Cochrane Collaboration's tool for assessing risk of bias in included studies. The scope of focus for this review was premenopausal adult women with a history of one or more UTI. The intervention consisted of a probiotic regimen for which the goal was to enhance the defensive microflora of the urogenital tract. Studies comparing a probiotic regimen to a placebo regimen were included. These studies' primary outcome was the proportion of women with at least one symptomatic bacterial UTI in each group (i.e., UTI recurrence rate) in the 12-month period following probiotic intervention. This study extends the work of researchers who systematically investigated the scientific literature on probiotics in the prevention of urinary tract infections with a particular focus on premenopausal women. RESULTS After screening, three parallel-group randomized-controlled trials (RCTs) were included. We estimated the overall pooled data of these three studies with a total of 284 participants to have met the predefined inclusion criteria and were therefore included in this review. The results demonstrated that probiotics did not have a significant effect in the prophylaxis of UTIs. (Risk Ratio (RR): 0.59 confidence interval (CI): 0.26, 1.33), Heterogeneity: Chi² = 6.63, df = 2 (p = 0.04); I² =70%, Test for overall effect: Z = 1.27 (p = 0.20). Conclusions: Probiotics did not demonstrate a significant benefit in reducing UTI recurrence compared to placebo in premenopausal women. However, more conclusive data is needed to determine the effect that probiotics have on strengthening the urogenital microbial barrier against pathogenic bacteria and protecting against UTI recurrence.
Collapse
Affiliation(s)
| | - Roger L Sur
- Urology, UC San Diego Health, San Diego, USA
| | - Eli Eshaghian
- Research, Western University of Health Sciences, Pomona, USA
| | - Kellie A Gaura
- Research, Western University of Health Sciences, Pomona, USA
| | | | | | | | - Joel E Abbott
- Urology, Pacific West Urology, Las Vegas, USA
- Research, Western University of Health Sciences, Pomona, USA
| |
Collapse
|
114
|
Grewal S, Reuvers JRD, Abis GSA, Otten RHJ, Kazemier G, Stockmann HBAC, van Egmond M, Oosterling SJ. Oral Antibiotic Prophylaxis Reduces Surgical Site Infection and Anastomotic Leakage in Patients Undergoing Colorectal Cancer Surgery. Biomedicines 2021; 9:biomedicines9091184. [PMID: 34572371 PMCID: PMC8471843 DOI: 10.3390/biomedicines9091184] [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: 07/17/2021] [Revised: 09/01/2021] [Accepted: 09/05/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Surgical-site infection (SSI) and anastomotic leakage (AL) are major complications following surgical resection of colorectal carcinoma (CRC). The beneficial effect of prophylactic oral antibiotics (OABs) on AL in particular is inconsistent. We investigated the impact of OABs on AL rates and on SSI. METHODS A systematic review and meta-analysis of recent RCTs and cohort studies was performed including patients undergoing elective CRC surgery, receiving OABs with or without mechanical bowel preparation (MBP). Primary outcomes were rates of SSI and AL. Secondarily, rates of SSI and AL were compared in broad-spectrum OABs and selective OABs (selective decontamination of the digestive tract (SDD)) subgroups. RESULTS Eight studies (seven RCTs and one cohort study) with a total of 2497 patients were included. Oral antibiotics combined with MBP was associated with a significant reduction in SSI (RR = 0.46, 95% confidence interval (CI) 0.31-0.69), I2 = 1.03%) and AL rates (RR = 0.58, 95% CI 0.37-0.91, I2 = 0.00%), compared to MBP alone. A subgroup analysis demonstrated that SDD resulted in a significant reduction in AL rates compared to broad-spectrum OABs (RR = 0.52, 95% CI 0.30 to 0.91), I2 = 0.00%). CONCLUSION OABs in addition to MBP reduces SSI and AL rates in patients undergoing elective CRC surgery and, more specifically, SDD appears to be more effective compared to broad-spectrum OABs in reducing AL.
Collapse
Affiliation(s)
- Simran Grewal
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centers, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (J.R.D.R.); (M.v.E.)
- Department of Surgery, Cancer Center Amsterdam, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
- Correspondence:
| | - J. Reinder D. Reuvers
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centers, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (J.R.D.R.); (M.v.E.)
- Department of Surgery, Cancer Center Amsterdam, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
| | - Gabor S. A. Abis
- Department of Surgery, Spaarne Gasthuis, Boerhaavelaan 22, 2035 RC Haarlem, The Netherlands; (G.S.A.A.); (H.B.A.C.S.); (S.J.O.)
| | - René H. J. Otten
- Medical Library, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
| | - Geert Kazemier
- Department of Surgery, Cancer Center Amsterdam, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
| | - Hein B. A. C. Stockmann
- Department of Surgery, Spaarne Gasthuis, Boerhaavelaan 22, 2035 RC Haarlem, The Netherlands; (G.S.A.A.); (H.B.A.C.S.); (S.J.O.)
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centers, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands; (J.R.D.R.); (M.v.E.)
- Department of Surgery, Cancer Center Amsterdam, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
| | - Steven J. Oosterling
- Department of Surgery, Spaarne Gasthuis, Boerhaavelaan 22, 2035 RC Haarlem, The Netherlands; (G.S.A.A.); (H.B.A.C.S.); (S.J.O.)
| |
Collapse
|
115
|
Mjaess G, Karam A, Aoun F, Albisinni S, Roumeguère T. Fecal microbiota transplantation for immunotherapy-resistant urological tumors: Is it time? An update of the recent literature. Cancer 2021; 128:14-19. [PMID: 34494666 DOI: 10.1002/cncr.33893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/26/2021] [Accepted: 06/03/2021] [Indexed: 12/19/2022]
Affiliation(s)
- Georges Mjaess
- Department of Urology, University Clinics of Brussels, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium.,Hotel-Dieu de France, University of Saint Joseph, Beirut, Lebanon
| | - Aya Karam
- Hotel-Dieu de France, University of Saint Joseph, Beirut, Lebanon
| | - Fouad Aoun
- Hotel-Dieu de France, University of Saint Joseph, Beirut, Lebanon.,Department of Urology, Institut Jules Bordet, Brussels, Belgium
| | - Simone Albisinni
- Department of Urology, University Clinics of Brussels, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Thierry Roumeguère
- Department of Urology, University Clinics of Brussels, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium.,Department of Urology, Institut Jules Bordet, Brussels, Belgium
| |
Collapse
|
116
|
Maan H, Gilhar O, Porat Z, Kolodkin-Gal I. Bacillus subtilis Colonization of Arabidopsis thaliana Roots Induces Multiple Biosynthetic Clusters for Antibiotic Production. Front Cell Infect Microbiol 2021; 11:722778. [PMID: 34557426 PMCID: PMC8454505 DOI: 10.3389/fcimb.2021.722778] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/16/2021] [Indexed: 12/01/2022] Open
Abstract
Beneficial and probiotic bacteria play an important role in conferring immunity of their hosts to a wide range of bacterial, viral, and fungal diseases. Bacillus subtilis is a Gram-positive bacterium that protects the plant from various pathogens due to its capacity to produce an extensive repertoire of antibiotics. At the same time, the plant microbiome is a highly competitive niche, with multiple microbial species competing for space and resources, a competition that can be determined by the antagonistic potential of each microbiome member. Therefore, regulating antibiotic production in the rhizosphere is of great importance for the elimination of pathogens and establishing beneficial host-associated communities. In this work, we used B. subtilis as a model to investigate the role of plant colonization in antibiotic production. Flow cytometry and imaging flow cytometry (IFC) analysis supported the notion that Arabidopsis thaliana specifically induced the transcription of the biosynthetic clusters for the non-ribosomal peptides surfactin, bacilysin, plipastatin, and the polyketide bacillaene. IFC was more robust in quantifying the inducing effects of A. thaliana, considering the overall heterogeneity of the population. Our results highlight IFC as a useful tool to study the effect of association with a plant host on bacterial gene expression. Furthermore, the common regulation of multiple biosynthetic clusters for antibiotic production by the plant can be translated to improve the performance and competitiveness of beneficial members of the plant microbiome.
Collapse
Affiliation(s)
- Harsh Maan
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Omri Gilhar
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ziv Porat
- Flow Cytometry Unit, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Ilana Kolodkin-Gal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
117
|
Chen LL, Abbaspour A, Mkoma GF, Bulik CM, Rück C, Djurfeldt D. Gut Microbiota in Psychiatric Disorders: A Systematic Review. Psychosom Med 2021; 83:679-692. [PMID: 34117156 PMCID: PMC8428865 DOI: 10.1097/psy.0000000000000959] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 02/04/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE This systematic review sought to comprehensively summarize gut microbiota research in psychiatric disorders following PRISMA guidelines. METHODS Literature searches were performed on databases using keywords involving gut microbiota and psychiatric disorders. Articles in English with human participants up until February 13, 2020, were reviewed. Risk of bias was assessed using a modified Newcastle-Ottawa Scale for microbiota studies. RESULTS Sixty-nine of 4231 identified studies met the inclusion criteria for extraction. In most studies, gut microbiota composition differed between individuals with psychiatric disorders and healthy controls; however, limited consistency was observed in the taxonomic profiles. At the genus level, the most replicated findings were higher abundance of Bifidobacterium and lower abundance of Roseburia and Faecalibacterium among patients with psychiatric disorders. CONCLUSIONS Gut bacteria that produce short-chain fatty acids, such as Roseburia and Faecalibacterium, could be less abundant in patients with psychiatric disorders, whereas commensal genera, for example, Bifidobacterium, might be more abundant compared with healthy controls. However, most included studies were hampered by methodological shortcomings including small sample size, unclear diagnostics, failure to address confounding factors, and inadequate bioinformatic processing, which might contribute to inconsistent results. Based on our findings, we provide recommendations to improve quality and comparability of future microbiota studies in psychiatry.
Collapse
|
118
|
Ma E, Maskarinec G, Lim U, Boushey CJ, Wilkens LR, Setiawan VW, Le Marchand L, Randolph TW, Jenkins IC, Curtis KR, Lampe JW, Hullar MA. Long-term association between diet quality and characteristics of the gut microbiome in the multiethnic cohort study. Br J Nutr 2021; 128:1-10. [PMID: 34369335 PMCID: PMC8825880 DOI: 10.1017/s0007114521002968] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As past usual diet quality may affect gut microbiome (GM) composition, we examined the association of the Healthy Eating Index (HEI)-2015 assessed 21 and 9 years before stool collection with measures of fecal microbial composition in a subset of the Multiethnic Cohort. A total of 5936 participants completed a validated quantitative FFQ (QFFQ) at cohort entry (Q1, 1993-1996), 5280 at follow-up (Q3, 2003-2008) and 1685 also at a second follow-up (Adiposity Phenotype Study (APS), 2013-2016). All participants provided a stool sample in 2013-2016. Fecal microbial composition was obtained from 16S rRNA gene sequencing (V1-V3 regions). HEI-2015 scores were computed based on each QFFQ. Using linear regression adjusted for relevant covariates, we calculated associations of HEI-2015 scores with gut microbial diversity and 152 individual genera. The mean HEI-2015 scores increased from Q1 (67 (sd 10)) to Q3 (71 (sd 11)) and APS (72 (sd 10)). Alpha diversity assessed by the Shannon Index was significantly higher with increasing tertiles of HEI-2015. Of the 152 bacterial genera tested, seven (Anaerostipes, Coprococcus_2, Eubacterium eligens, Lachnospira, Lachnospiraceae_ND3007, Ruminococcaceae_UCG-013 and Ruminococcus_1) were positively and five (Collinsella, Parabacteroides, Ruminiclostridium_5, Ruminococcus gnavus and Tyzzerella) were inversely associated with HEI-2015 assessed in Q1, Q3 and APS. The estimates of change per unit of the HEI-2015 score associated with the abundance of these twelve genera were consistent across the three questionnaires. The quality of past diet, assessed as far as ∼20 years before stool collection, is equally predictive of GM composition as concurrently assessed diet, indicative of the long-term consistency of this relation.
Collapse
Affiliation(s)
- Erica Ma
- University of Hawai’i Cancer Center, Honolulu, HI
| | | | - Unhee Lim
- University of Hawai’i Cancer Center, Honolulu, HI
| | | | | | - V. Wendy Setiawan
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA
| | | | | | | | | | | | | |
Collapse
|
119
|
Kumari R, Palaniyandi S, Hildebrandt GC. The microbiome-the revealing of a long time unbeknownst factor for outcome in murine models of graft-versus-host disease. Bone Marrow Transplant 2021; 56:1777-1783. [PMID: 34052837 DOI: 10.1038/s41409-021-01325-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 04/13/2021] [Accepted: 04/21/2021] [Indexed: 02/04/2023]
Affiliation(s)
- Reena Kumari
- Division of Hematology & Blood and Marrow Transplantation, Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Senthilnathan Palaniyandi
- Division of Hematology & Blood and Marrow Transplantation, Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Gerhard Carl Hildebrandt
- Division of Hematology & Blood and Marrow Transplantation, Markey Cancer Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
120
|
Vyhnalova T, Danek Z, Gachova D, Linhartova PB. The Role of the Oral Microbiota in the Etiopathogenesis of Oral Squamous Cell Carcinoma. Microorganisms 2021; 9:microorganisms9081549. [PMID: 34442627 PMCID: PMC8400438 DOI: 10.3390/microorganisms9081549] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 02/07/2023] Open
Abstract
Dysbiosis in the oral environment may play a role in the etiopathogenesis of oral squamous cell carcinoma (OSCC). This review aims to summarize the current knowledge about the association of oral microbiota with OSCC and to describe possible etiopathogenetic mechanisms involved in processes of OSCC development and progression. Association studies included in this review were designed as case–control/case studies, analyzing the bacteriome, mycobiome, and virome from saliva, oral rinses, oral mucosal swabs, or oral mucosal tissue samples (deep and superficial) and comparing the results in healthy individuals to those with OSCC and/or with premalignant lesions. Changes in relative abundances of specific bacteria (e.g., Porphyromonas gingivalis, Fusobacterium nucleatum, Streptococcus sp.) and fungi (especially Candida sp.) were associated with OSCC. Viruses can also play a role; while the results of studies investigating the role of human papillomavirus in OSCC development are controversial, Epstein–Barr virus was positively correlated with OSCC. The oral microbiota has been linked to tumorigenesis through a variety of mechanisms, including the stimulation of cell proliferation, tumor invasiveness, angiogenesis, inhibition of cell apoptosis, induction of chronic inflammation, or production of oncometabolites. We also advocate for the necessity of performing a complex analysis of the microbiome in further studies and of standardizing the sampling procedures by establishing guidelines to support future meta-analyses.
Collapse
Affiliation(s)
- Tereza Vyhnalova
- Environmental Genomics Research Group, RECETOX, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic; (T.V.); (D.G.); (P.B.L.)
- Department of Maxillofacial Surgery, Faculty of Medicine, Masaryk University, Jihlavská 20, 62500 Brno, Czech Republic
| | - Zdenek Danek
- Environmental Genomics Research Group, RECETOX, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic; (T.V.); (D.G.); (P.B.L.)
- Department of Maxillofacial Surgery, Faculty of Medicine, Masaryk University, Jihlavská 20, 62500 Brno, Czech Republic
- Department of Maxillofacial Surgery, University Hospital Brno, Jihlavská 20, 62500 Brno, Czech Republic
- Correspondence: ; Tel.: +420-777-550-596
| | - Daniela Gachova
- Environmental Genomics Research Group, RECETOX, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic; (T.V.); (D.G.); (P.B.L.)
| | - Petra Borilova Linhartova
- Environmental Genomics Research Group, RECETOX, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic; (T.V.); (D.G.); (P.B.L.)
- Department of Maxillofacial Surgery, Faculty of Medicine, Masaryk University, Jihlavská 20, 62500 Brno, Czech Republic
- Institute of Medical Genetics and Genomics, Faculty of Medicine, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| |
Collapse
|
121
|
Estaki M, Jiang L, Bokulich NA, McDonald D, González A, Kosciolek T, Martino C, Zhu Q, Birmingham A, Vázquez-Baeza Y, Dillon MR, Bolyen E, Caporaso JG, Knight R. QIIME 2 Enables Comprehensive End-to-End Analysis of Diverse Microbiome Data and Comparative Studies with Publicly Available Data. ACTA ACUST UNITED AC 2021; 70:e100. [PMID: 32343490 PMCID: PMC9285460 DOI: 10.1002/cpbi.100] [Citation(s) in RCA: 233] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
QIIME 2 is a completely re‐engineered microbiome bioinformatics platform based on the popular QIIME platform, which it has replaced. QIIME 2 facilitates comprehensive and fully reproducible microbiome data science, improving accessibility to diverse users by adding multiple user interfaces. QIIME 2 can be combined with Qiita, an open‐source web‐based platform, to re‐use available data for meta‐analysis. The following basic protocol describes how to install QIIME 2 on a single computer and analyze microbiome sequence data, from processing of raw DNA sequence reads through generating publishable interactive figures. These interactive figures allow readers of a study to interact with data with the same ease as its authors, advancing microbiome science transparency and reproducibility. We also show how plug‐ins developed by the community to add analysis capabilities can be installed and used with QIIME 2, enhancing various aspects of microbiome analyses—e.g., improving taxonomic classification accuracy. Finally, we illustrate how users can perform meta‐analyses combining different datasets using readily available public data through Qiita. In this tutorial, we analyze a subset of the Early Childhood Antibiotics and the Microbiome (ECAM) study, which tracked the microbiome composition and development of 43 infants in the United States from birth to 2 years of age, identifying microbiome associations with antibiotic exposure, delivery mode, and diet. For more information about QIIME 2, see https://qiime2.org. To troubleshoot or ask questions about QIIME 2 and microbiome analysis, join the active community at https://forum.qiime2.org. © 2020 The Authors. Basic Protocol: Using QIIME 2 with microbiome data Support Protocol: Further microbiome analyses
Collapse
Affiliation(s)
- Mehrbod Estaki
- Department of Pediatrics, University of California San Diego, La Jolla, California
| | - Lingjing Jiang
- Division of Biostatistics, University of California San Diego, La Jolla, California
| | - Nicholas A Bokulich
- Center for Applied Microbiome Science, Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona.,Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona
| | - Daniel McDonald
- Department of Pediatrics, University of California San Diego, La Jolla, California
| | - Antonio González
- Department of Pediatrics, University of California San Diego, La Jolla, California
| | - Tomasz Kosciolek
- Department of Pediatrics, University of California San Diego, La Jolla, California.,Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Cameron Martino
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California.,Center for Microbiome Innovation, University of California San Diego, La Jolla, California
| | - Qiyun Zhu
- Department of Pediatrics, University of California San Diego, La Jolla, California
| | - Amanda Birmingham
- Center for Computational Biology and Bioinformatics, University of California San Diego, La Jolla, California
| | - Yoshiki Vázquez-Baeza
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California.,Jacobs School of Engineering, University of California San Diego, La Jolla, California
| | - Matthew R Dillon
- Center for Applied Microbiome Science, Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona
| | - Evan Bolyen
- Center for Applied Microbiome Science, Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona
| | - J Gregory Caporaso
- Center for Applied Microbiome Science, Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona.,Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, California.,Center for Microbiome Innovation, University of California San Diego, La Jolla, California.,Department of Computer Science and Engineering, University of California San Diego, La Jolla, California.,Department of Bioengineering, University of California San Diego, La Jolla, California
| |
Collapse
|
122
|
Allaband C, Lingaraju A, Martino C, Russell B, Tripathi A, Poulsen O, Dantas Machado AC, Zhou D, Xue J, Elijah E, Malhotra A, Dorrestein PC, Knight R, Haddad GG, Zarrinpar A. Intermittent Hypoxia and Hypercapnia Alter Diurnal Rhythms of Luminal Gut Microbiome and Metabolome. mSystems 2021; 6:e0011621. [PMID: 34184915 PMCID: PMC8269208 DOI: 10.1128/msystems.00116-21] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/07/2021] [Indexed: 12/12/2022] Open
Abstract
Obstructive sleep apnea (OSA), characterized by intermittent hypoxia and hypercapnia (IHC), affects the composition of the gut microbiome and metabolome. The gut microbiome has diurnal oscillations that play a crucial role in regulating circadian and overall metabolic homeostasis. Thus, we hypothesized that IHC adversely alters the gut luminal dynamics of key microbial families and metabolites. The objective of this study was to determine the diurnal dynamics of the fecal microbiome and metabolome of Apoe-/- mice after a week of IHC exposure. Individually housed, 10-week-old Apoe-/- mice on an atherogenic diet were split into two groups. One group was exposed to daily IHC conditions for 10 h (Zeitgeber time 2 [ZT2] to ZT12), while the other was maintained in room air. Six days after the initiation of the IHC conditions, fecal samples were collected every 4 h for 24 h (6 time points). We performed 16S rRNA gene amplicon sequencing and untargeted liquid chromatography-mass spectrometry (LC-MS) to assess changes in the microbiome and metabolome. IHC induced global changes in the cyclical dynamics of the gut microbiome and metabolome. Ruminococcaceae, Lachnospiraceae, S24-7, and Verrucomicrobiaceae had the greatest shifts in their diurnal oscillations. In the metabolome, bile acids, glycerolipids (phosphocholines and phosphoethanolamines), and acylcarnitines were greatly affected. Multi-omic analysis of these results demonstrated that Ruminococcaceae and tauro-β-muricholic acid (TβMCA) cooccur and are associated with IHC conditions and that Coriobacteriaceae and chenodeoxycholic acid (CDCA) cooccur and are associated with control conditions. IHC significantly change the diurnal dynamics of the fecal microbiome and metabolome, increasing members and metabolites that are proinflammatory and proatherogenic while decreasing protective ones. IMPORTANCE People with obstructive sleep apnea are at a higher risk of high blood pressure, type 2 diabetes, cardiac arrhythmias, stroke, and sudden cardiac death. We wanted to understand whether the gut microbiome changes induced by obstructive sleep apnea could potentially explain some of these medical problems. By collecting stool from a mouse model of this disease at multiple time points during the day, we studied how obstructive sleep apnea changed the day-night patterns of microbes and metabolites of the gut. Since the oscillations of the gut microbiome play a crucial role in regulating metabolism, changes in these oscillations can explain why these patients can develop so many metabolic problems. We found changes in microbial families and metabolites that regulate many metabolic pathways contributing to the increased risk for heart disease seen in patients with obstructive sleep apnea.
Collapse
Affiliation(s)
- Celeste Allaband
- Division of Gastroenterology, University of California, San Diego, La Jolla, California, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Amulya Lingaraju
- Division of Gastroenterology, University of California, San Diego, La Jolla, California, USA
| | - Cameron Martino
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
| | - Baylee Russell
- Division of Gastroenterology, University of California, San Diego, La Jolla, California, USA
| | - Anupriya Tripathi
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy, University of California, San Diego, La Jolla, California, USA
| | - Orit Poulsen
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | | | - Dan Zhou
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Jin Xue
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Emmanuel Elijah
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy, University of California, San Diego, La Jolla, California, USA
| | - Atul Malhotra
- Center for Circadian Biology, University of California, San Diego, La Jolla, California, USA
| | - Pieter C. Dorrestein
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy, University of California, San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
| | - Rob Knight
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, California, USA
| | - Gabriel G. Haddad
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Department of Neuroscience, University of California, San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
| | - Amir Zarrinpar
- Division of Gastroenterology, University of California, San Diego, La Jolla, California, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, California, USA
- Institute of Diabetes and Metabolic Health, University of California, San Diego, La Jolla, California, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, California, USA
- VA Health Sciences San Diego, La Jolla, California, USA
| |
Collapse
|
123
|
Losol P, Choi JP, Kim SH, Chang YS. The Role of Upper Airway Microbiome in the Development of Adult Asthma. Immune Netw 2021; 21:e19. [PMID: 34277109 PMCID: PMC8263217 DOI: 10.4110/in.2021.21.e19] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
Clinical and molecular phenotypes of asthma are complex. The main phenotypes of adult asthma are characterized by eosinophil and/or neutrophil cell dominant airway inflammation that represent distinct clinical features. Upper and lower airways constitute a unique system and their interaction shows functional complementarity. Although human upper airway contains various indigenous commensals and opportunistic pathogenic microbiome, imbalance of this interactions lead to pathogen overgrowth and increased inflammation and airway remodeling. Competition for epithelial cell attachment, different susceptibilities to host defense molecules and antimicrobial peptides, and the production of proinflammatory cytokine and pattern recognition receptors possibly determine the pattern of this inflammation. Exposure to environmental factors, including infection, air pollution, smoking is commonly associated with asthma comorbidity, severity, exacerbation and resistance to anti-microbial and steroid treatment, and these effects may also be modulated by host and microbial genetics. Administration of probiotic, antibiotic and corticosteroid treatment for asthma may modify the composition of resident microbiota and clinical features. This review summarizes the effect of some environmental factors on the upper respiratory microbiome, the interaction between host-microbiome, and potential impact of asthma treatment on the composition of the upper airway microbiome.
Collapse
Affiliation(s)
- Purevsuren Losol
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Council, Seoul, Korea
| | - Jun-Pyo Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sae-Hoon Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Council, Seoul, Korea
| | - Yoon-Seok Chang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Council, Seoul, Korea
| |
Collapse
|
124
|
Dietary Habits and Gut Microbiota in Healthy Adults: Focusing on the Right Diet. A Systematic Review. Int J Mol Sci 2021; 22:ijms22136728. [PMID: 34201611 PMCID: PMC8269086 DOI: 10.3390/ijms22136728] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/13/2021] [Accepted: 06/15/2021] [Indexed: 12/17/2022] Open
Abstract
Diet is the first to affect our intestinal microbiota and therefore the state of eubiosis. Several studies are highlighting the potential benefits of taking certain nutritional supplements, but a dietary regime that can ensure the health of the intestinal microbiota, and the many pathways it governs, is not yet clearly defined. We performed a systematic review of the main studies concerning the impact of an omnivorous diet on the composition of the microbiota and the production of short-chain fatty acids (SCFAs). Some genera and phyla of interest emerged significantly and about half of the studies evaluated consider them to have an equally significant impact on the production of SCFAs, to be a source of nutrition for our colon cells, and many other processes. Although numerous randomized trials are still needed, the Mediterranean diet could play a valuable role in ensuring our health through direct interaction with our microbiota.
Collapse
|
125
|
Microbial colonization of the gastrointestinal tract of dairy calves - a review of its importance and relationship to health and performance. Anim Health Res Rev 2021; 22:97-108. [PMID: 34132191 DOI: 10.1017/s1466252321000062] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This review aims to explain how microbial colonization of the gastrointestinal tract (GIT) in young dairy calves is related to health and, consequently, to the performance of these animals. The review addresses everything from the fundamental aspects of microbial colonization to the current understanding about the microbiota manipulation to improve performance in adult animals. The ruminal microbiota is the most studied, mainly due to the high interest in the fermentative aspects, the production of short-chain fatty acids, and microbial proteins, and its effects on animal production. However, in recent years, the intestinal microbiota has gained space between studies, mainly due to the relationship to the host health and how it affects performance. Understanding how the GIT's microbiota looks like and how it is colonized may allow future studies to predict the best timing for dietary interventions as a way to manipulate it and, consequently, improve the health and performance of young ruminants.
Collapse
|
126
|
Deng J, Angulo MT, Saavedra S. Generalizing game-changing species across microbial communities. ISME COMMUNICATIONS 2021; 1:22. [PMID: 36737668 PMCID: PMC9723773 DOI: 10.1038/s43705-021-00022-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/10/2021] [Accepted: 05/18/2021] [Indexed: 02/07/2023]
Abstract
Microbes form multispecies communities that play essential roles in our environment and health. Not surprisingly, there is an increasing need for understanding if certain invader species will modify a given microbial community, producing either a desired or undesired change in the observed collection of resident species. However, the complex interactions that species can establish between each other and the diverse external factors underlying their dynamics have made constructing such understanding context-specific. Here we integrate tractable theoretical systems with tractable experimental systems to find general conditions under which non-resident species can change the collection of resident communities-game-changing species. We show that non-resident colonizers are more likely to be game-changers than transients, whereas game-changers are more likely to suppress than to promote resident species. Importantly, we find general heuristic rules for game-changers under controlled environments by integrating mutual invasibility theory with in vitro experimental systems, and general heuristic rules under changing environments by integrating structuralist theory with in vivo experimental systems. Despite the strong context-dependency of microbial communities, our work shows that under an appropriate integration of tractable theoretical and experimental systems, it is possible to unveil regularities that can then be potentially extended to understand the behavior of complex natural communities.
Collapse
Affiliation(s)
- Jie Deng
- Department of Civil and Environmental Engineering, MIT, Cambridge, MA, USA
| | - Marco Tulio Angulo
- CONACyT - Institute of Mathematics, Universidad Nacional Autónoma de México, Juriquilla, México.
| | - Serguei Saavedra
- Department of Civil and Environmental Engineering, MIT, Cambridge, MA, USA.
| |
Collapse
|
127
|
Weissman JL, Dogra S, Javadi K, Bolten S, Flint R, Davati C, Beattie J, Dixit K, Peesay T, Awan S, Thielen P, Breitwieser F, Johnson PLF, Karig D, Fagan WF, Bewick S. Exploring the functional composition of the human microbiome using a hand-curated microbial trait database. BMC Bioinformatics 2021; 22:306. [PMID: 34098872 PMCID: PMC8186035 DOI: 10.1186/s12859-021-04216-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 05/25/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Even when microbial communities vary wildly in their taxonomic composition, their functional composition is often surprisingly stable. This suggests that a functional perspective could provide much deeper insight into the principles governing microbiome assembly. Much work to date analyzing the functional composition of microbial communities, however, relies heavily on inference from genomic features. Unfortunately, output from these methods can be hard to interpret and often suffers from relatively high error rates. RESULTS We built and analyzed a domain-specific microbial trait database from known microbe-trait pairs recorded in the literature to better understand the functional composition of the human microbiome. Using a combination of phylogentically conscious machine learning tools and a network science approach, we were able to link particular traits to areas of the human body, discover traits that determine the range of body areas a microbe can inhabit, and uncover drivers of metabolic breadth. CONCLUSIONS Domain-specific trait databases are an effective compromise between noisy methods to infer complex traits from genomic data and exhaustive, expensive attempts at database curation from the literature that do not focus on any one subset of taxa. They provide an accurate account of microbial traits and, by limiting the number of taxa considered, are feasible to build within a reasonable time-frame. We present a database specific for the human microbiome, in the hopes that this will prove useful for research into the functional composition of human-associated microbial communities.
Collapse
Affiliation(s)
- J L Weissman
- Department of Biology, University of Maryland - College Park, College Park, MD, USA
| | - Sonia Dogra
- Department of Biology, University of Maryland - College Park, College Park, MD, USA
| | - Keyan Javadi
- Department of Biology, University of Maryland - College Park, College Park, MD, USA
| | - Samantha Bolten
- Department of Biology, University of Maryland - College Park, College Park, MD, USA
| | - Rachel Flint
- Department of Biology, University of Maryland - College Park, College Park, MD, USA
| | - Cyrus Davati
- Department of Biology, University of Maryland - College Park, College Park, MD, USA
| | - Jess Beattie
- Department of Biology, University of Maryland - College Park, College Park, MD, USA
| | - Keshav Dixit
- Department of Biology, University of Maryland - College Park, College Park, MD, USA
| | - Tejasvi Peesay
- Department of Biology, University of Maryland - College Park, College Park, MD, USA
| | - Shehar Awan
- Department of Biology, University of Maryland - College Park, College Park, MD, USA
| | - Peter Thielen
- Research and Exploratory Development Department, Johns Hopkins Applied Physics Laboratory, Laurel, MD, USA
| | - Florian Breitwieser
- Research and Exploratory Development Department, Johns Hopkins Applied Physics Laboratory, Laurel, MD, USA
| | - Philip L F Johnson
- Department of Biology, University of Maryland - College Park, College Park, MD, USA
| | - David Karig
- Bioengineering Department, Clemson University, Clemson, SC, USA
| | - William F Fagan
- Department of Biology, University of Maryland - College Park, College Park, MD, USA
| | - Sharon Bewick
- Biological Sciences Department, Clemson University, Clemson, SC, USA.
| |
Collapse
|
128
|
Meaddough E, Abenavoli L, Sarasua S, Boccuto L. Genetic and environmental factors influencing the interaction between the gut microbiota and the human host: implications for gastrointestinal disorders and treatment approaches. Minerva Gastroenterol (Torino) 2021; 67:369-376. [PMID: 34057334 DOI: 10.23736/s2724-5985.21.02927-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The growing knowledge on the microbiota hosted in the human intestine has allowed researchers and clinicians to discover a critical role for these microorganisms in the pathogenesis of several human disorders. In particular, perturbation in the microbiotic strains populating the gastrointestinal tract has been associated with several conditions affecting the digestive system. The composition of the human gut microbiota is influenced by both genetic factors, like the human and the microbiotic genomes, and environmental ones, such as diet or drugs. Alteration of the interaction between the gut microbiota and the human host has been proven to be associated with several gastrointestinal disorders as well as potential effects on pharmacological therapies.
Collapse
Affiliation(s)
- Erika Meaddough
- School of Nursing, Healthcare Genetics Program, College of Behavioral, Social and Health Sciences, Clemson University, Clemson, SC, USA
| | - Ludovico Abenavoli
- Department of Health Sciences, University Magna Graecia, Catanzaro, Italy
| | - Sara Sarasua
- School of Nursing, Healthcare Genetics Program, College of Behavioral, Social and Health Sciences, Clemson University, Clemson, SC, USA
| | - Luigi Boccuto
- School of Nursing, Healthcare Genetics Program, College of Behavioral, Social and Health Sciences, Clemson University, Clemson, SC, USA -
| |
Collapse
|
129
|
Pjevac P, Hausmann B, Schwarz J, Kohl G, Herbold CW, Loy A, Berry D. An Economical and Flexible Dual Barcoding, Two-Step PCR Approach for Highly Multiplexed Amplicon Sequencing. Front Microbiol 2021; 12:669776. [PMID: 34093488 PMCID: PMC8173057 DOI: 10.3389/fmicb.2021.669776] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/12/2021] [Indexed: 12/26/2022] Open
Abstract
In microbiome research, phylogenetic and functional marker gene amplicon sequencing is the most commonly-used community profiling approach. Consequently, a plethora of protocols for the preparation and multiplexing of samples for amplicon sequencing have been developed. Here, we present two economical high-throughput gene amplification and sequencing workflows that are implemented as standard operating procedures at the Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna. These workflows are based on a previously-published two-step PCR approach, but have been updated to either increase the accuracy of results, or alternatively to achieve orders of magnitude higher numbers of samples to be multiplexed in a single sequencing run. The high-accuracy workflow relies on unique dual sample barcoding. It allows the same level of sample multiplexing as the previously-published two-step PCR approach, but effectively eliminates residual read missasignments between samples (crosstalk) which are inherent to single barcoding approaches. The high-multiplexing workflow is based on combinatorial dual sample barcoding, which theoretically allows for multiplexing up to 299,756 amplicon libraries of the same target gene in a single massively-parallelized amplicon sequencing run. Both workflows presented here are highly economical, easy to implement, and can, without significant modifications or cost, be applied to any target gene of interest.
Collapse
Affiliation(s)
- Petra Pjevac
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Bela Hausmann
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Jasmin Schwarz
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Gudrun Kohl
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Craig W. Herbold
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Alexander Loy
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - David Berry
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Vienna, Austria
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| |
Collapse
|
130
|
Effect of arabinogalactan on the gut microbiome: A randomized, double-blind, placebo-controlled, crossover trial in healthy adults. Nutrition 2021; 90:111273. [PMID: 34004416 DOI: 10.1016/j.nut.2021.111273] [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] [Received: 11/01/2020] [Revised: 02/22/2021] [Accepted: 04/03/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Promising evidence suggests beneficial health effects of arabinogalactan, but little is known about the effect of this non-digestible carbohydrate on the gut microbiota, a crucial mediator of human health. The objective of this study was to investigate the effect of an arabinogalactan product (ResistAid) on the fecal microbiome and short-chain fatty acids and gastrointestinal tolerance in healthy adults in a randomized, double-blind, crossover trial. METHODS Thirty adults were randomly assigned to consume 15 g/d maltodextrin (control) or ResistAid for 6 wk. RESULTS At week 6, compared to placebo, ResistAid supplementation led to a significant decrease in the ratio of fecal Firmicutes to Bacteroidetes, driven by an increase in Bacteroidetes and a decrease in Firmicutes. Moreover, the relative abundance of Bifidobacterium tended to increase with ResistAid supplementation. Additionally, ResistAid significantly decreased the α-diversity of the fecal microbiome. Predicted functional abundances based on 16S rRNA sequences showed that ResistAid supplementation increased the gene abundance of the gut microbiome for α-l-rhamnosidase, β-fructosidase, and levanase, as well as tricarboxylic acid and vitamin B6 biosynthesis pathways. Fecal isovaleric, valeric, and hexanoic acids were significantly lower after ResistAid consumption. There were no statistically significant changes in bowel habit, stool consistency, gastrointestinal tolerance symptoms, chemistry profile, metabolic panel, or vitals, suggesting that consumption of 15 g daily ResistAid over 6 wk is safe. CONCLUSION These results demonstrate that the gut microbiome composition and predicted functions can be modulated by ResistAid consumption, perhaps suggesting a mechanistic explanation on its reported benefits in metabolic parameters and the immune system.
Collapse
|
131
|
Jarett JK, Kingsbury DD, Dahlhausen KE, Ganz HH. Best Practices for Microbiome Study Design in Companion Animal Research. Front Vet Sci 2021; 8:644836. [PMID: 33898544 PMCID: PMC8062777 DOI: 10.3389/fvets.2021.644836] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/09/2021] [Indexed: 12/31/2022] Open
Abstract
The gut microbiome is a community of microorganisms that inhabits an animal host's gastrointestinal tract, with important effects on animal health that are shaped by multiple environmental, dietary, and host-associated factors. Clinical and dietary trials in companion animals are increasingly including assessment of the microbiome, but interpretation of these results is often hampered by suboptimal choices in study design. Here, we review best practices for conducting feeding trials or clinical trials that intend to study the effects of an intervention on the microbiota. Choices for experimental design, including a review of basic designs, controls, and comparison groups, are discussed in the context of special considerations necessary for microbiome studies. Diet is one of the strongest influences on the composition of gut microbiota, so applications specific to nutritional interventions are discussed in detail. Lastly, we provide specific advice for successful recruitment of colony animals and household pets into an intervention study. This review is intended to serve as a resource to academic and industry researchers, clinicians, and veterinarians alike, for studies that test many different types of interventions.
Collapse
|
132
|
Davis ML, Huang Y, Wang K. Rank normalization empowers a t-test for microbiome differential abundance analysis while controlling for false discoveries. Brief Bioinform 2021; 22:6210069. [PMID: 33822893 DOI: 10.1093/bib/bbab059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/27/2021] [Accepted: 02/05/2021] [Indexed: 11/12/2022] Open
Abstract
A major task in the analysis of microbiome data is to identify microbes associated with differing biological conditions. Before conducting analysis, raw data must first be adjusted so that counts from different samples are comparable. A typical approach is to estimate normalization factors by which all counts in a sample are multiplied or divided. However, the inherent variation associated with estimation of normalization factors are often not accounted for in subsequent analysis, leading to a loss of precision. Rank normalization is a nonparametric alternative to the estimation of normalization factors in which each count for a microbial feature is replaced by its intrasample rank. Although rank normalization has been successfully applied to microarray analysis in the past, it has yet to be explored for microbiome data, which is characterized by high frequencies of 0s, strongly correlated features and compositionality. We propose to use rank normalization as an alternative to the estimation of normalization factors and examine its performance when paired with a two-sample t-test. On a rigorous 3rd-party benchmarking simulation, it is shown to offer strong control over the false discovery rate, and at sample sizes greater than 50 per treatment group, to offer an improvement in performance over commonly used normalization factors paired with t-tests, Wilcoxon rank-sum tests and methodologies implemented by R packages. On two real datasets, it yielded valid and reproducible results that were strongly in agreement with the original findings and the existing literature, further demonstrating its robustness and future potential. Availability: The data underlying this article are available online along with R code and supplementary materials at https://github.com/matthewlouisdavisBioStat/Rank-Normalization-Empowers-a-T-Test.
Collapse
Affiliation(s)
- Matthew L Davis
- Department of Biostatistics, University of Iowa College of Public Health, 145 N Riverside Dr, 52242, IA, USA
| | - Yuan Huang
- Department of Biostatistics, Yale School of Public Health, 60 College St, 06510, CT, USA
| | - Kai Wang
- Department of Biostatistics, University of Iowa College of Public Health, 145 N Riverside Dr, 52242, IA, USA
| |
Collapse
|
133
|
Zhao Y, Federico A, Faits T, Manimaran S, Segrè D, Monti S, Johnson WE. animalcules: interactive microbiome analytics and visualization in R. MICROBIOME 2021; 9:76. [PMID: 33775256 PMCID: PMC8006385 DOI: 10.1186/s40168-021-01013-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 02/02/2021] [Indexed: 05/27/2023]
Abstract
BACKGROUND Microbial communities that live in and on the human body play a vital role in health and disease. Recent advances in sequencing technologies have enabled the study of microbial communities at unprecedented resolution. However, these advances in data generation have presented novel challenges to researchers attempting to analyze and visualize these data. RESULTS To address some of these challenges, we have developed animalcules, an easy-to-use interactive microbiome analysis toolkit for 16S rRNA sequencing data, shotgun DNA metagenomics data, and RNA-based metatranscriptomics profiling data. This toolkit combines novel and existing analytics, visualization methods, and machine learning models. For example, the toolkit features traditional microbiome analyses such as alpha/beta diversity and differential abundance analysis, combined with new methods for biomarker identification are. In addition, animalcules provides interactive and dynamic figures that enable users to understand their data and discover new insights. animalcules can be used as a standalone command-line R package or users can explore their data with the accompanying interactive R Shiny interface. CONCLUSIONS We present animalcules, an R package for interactive microbiome analysis through either an interactive interface facilitated by R Shiny or various command-line functions. It is the first microbiome analysis toolkit that supports the analysis of all 16S rRNA, DNA-based shotgun metagenomics, and RNA-sequencing based metatranscriptomics datasets. animalcules can be freely downloaded from GitHub at https://github.com/compbiomed/animalcules or installed through Bioconductor at https://www.bioconductor.org/packages/release/bioc/html/animalcules.html . Video abstract.
Collapse
Affiliation(s)
- Yue Zhao
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, MA USA
- Bioinformatics Program, College of Engineering, Boston University, Boston, MA USA
| | - Anthony Federico
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, MA USA
- Bioinformatics Program, College of Engineering, Boston University, Boston, MA USA
| | - Tyler Faits
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, MA USA
- Bioinformatics Program, College of Engineering, Boston University, Boston, MA USA
| | - Solaiappan Manimaran
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, MA USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
| | - Daniel Segrè
- Bioinformatics Program, College of Engineering, Boston University, Boston, MA USA
- Departments of Biology, Biomedical Engineering, and Physics, Boston University, Boston, MA USA
| | - Stefano Monti
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, MA USA
- Bioinformatics Program, College of Engineering, Boston University, Boston, MA USA
| | - W. Evan Johnson
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, MA USA
- Bioinformatics Program, College of Engineering, Boston University, Boston, MA USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
| |
Collapse
|
134
|
Hou Q, Kolodkin-Gal I. Harvesting the complex pathways of antibiotic production and resistance of soil bacilli for optimizing plant microbiome. FEMS Microbiol Ecol 2021; 96:5872479. [PMID: 32672816 DOI: 10.1093/femsec/fiaa142] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/13/2020] [Indexed: 01/04/2023] Open
Abstract
A sustainable future increasing depends on our capacity to utilize beneficial plant microbiomes to meet our growing needs. Plant microbiome symbiosis is a hallmark of the beneficial interactions between bacteria and their host. Specifically, colonization of plant roots by biocontrol agents and plant growth-promoting bacteria can play an important role in maintaining the optimal rhizosphere environment, supporting plant growth and promoting its fitness. Rhizosphere communities confer immunity against a wide range of foliar diseases by secreting antibiotics and activating plant defences. At the same time, the rhizosphere is a highly competitive niche, with multiple microbial species competing for space and resources, engaged in an arms race involving the production of a vast array of antibiotics and utilization of a variety of antibiotic resistance mechanisms. Therefore, elucidating the mechanisms that govern antibiotic production and resistance in the rhizosphere is of great significance for designing beneficial communities with enhanced biocontrol properties. In this review, we used Bacillus subtilis and B. amyloliquefaciens as models to investigate the genetics of antibiosis and the potential for its translation of into improved plant microbiome performance.
Collapse
Affiliation(s)
- Qihui Hou
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ilana Kolodkin-Gal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
135
|
Thavamani A, Salem I, Sferra TJ, Sankararaman S. Impact of Altered Gut Microbiota and Its Metabolites in Cystic Fibrosis. Metabolites 2021; 11:123. [PMID: 33671639 PMCID: PMC7926988 DOI: 10.3390/metabo11020123] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/15/2021] [Accepted: 02/20/2021] [Indexed: 12/14/2022] Open
Abstract
Cystic fibrosis (CF) is the most common lethal, multisystemic genetic disorder in Caucasians. Mutations in the gene encoding the cystic fibrosis transmembrane regulator (CFTR) protein are responsible for impairment of epithelial anionic transport, leading to impaired fluid regulation and pH imbalance across multiple organs. Gastrointestinal (GI) manifestations in CF may begin in utero and continue throughout the life, resulting in a chronic state of an altered intestinal milieu. Inherent dysfunction of CFTR leads to dysbiosis of the gut. This state of dysbiosis is further perpetuated by acquired factors such as use of antibiotics for recurrent pulmonary exacerbations. Since the gastrointestinal microbiome and their metabolites play a vital role in nutrition, metabolic, inflammatory, and immune functions, the gut dysbiosis will in turn impact various manifestations of CF-both GI and extra-GI. This review focuses on the consequences of gut dysbiosis and its metabolic implications on CF disease and possible ways to restore homeostasis.
Collapse
Affiliation(s)
- Aravind Thavamani
- Department of Pediatrics, Division of Pediatric Gastroenterology, UH Rainbow Babies & Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (A.T.); (T.J.S.)
| | - Iman Salem
- Center for Medial Mycology, Case Western Reserve University School of Medicine, UH Cleveland Medical Center, Cleveland, OH 44106, USA;
| | - Thomas J. Sferra
- Department of Pediatrics, Division of Pediatric Gastroenterology, UH Rainbow Babies & Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (A.T.); (T.J.S.)
| | - Senthilkumar Sankararaman
- Department of Pediatrics, Division of Pediatric Gastroenterology, UH Rainbow Babies & Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (A.T.); (T.J.S.)
| |
Collapse
|
136
|
Abstract
The etiology of polycystic ovary syndrome (PCOS) remains unclear, although studies indicate that both genetic and environmental factors contribute to the syndrome. In 2012, Tremellen and Pearce proposed the idea that dysbiosis of the intestinal (gut) microbiome is a causative factor of metabolic and reproductive manifestations of PCOS. In the past 5 years, studies in both humans and rodent models have demonstrated that changes in the taxonomic composition of gut bacteria are associated with PCOS. Studies have also clearly shown that these changes in gut microbiota are associated with PCOS as opposed to obesity, since these changes are observed in women with PCOS that are both of a normal weight or obese, as well as in adolescent girls with PCOS and obesity compared with body mass index- and age-matched females without the disorder. Additionally, studies in both women with PCOS and rodent models of PCOS demonstrated that hyperandrogenism is associated with gut microbial dysbiosis, indicating that androgens may modulate the gut microbial community in females. One study reported that the fecal microbiome transplantation of stool from women with PCOS or exposure to certain bacteria resulted in a PCOS-like phenotype in mice, while other studies showed that exposure to a healthy gut microbiome, pre/probiotics, or specific gut metabolites resulted in protection from developing PCOS-like traits in mice. Altogether, these results suggest that dysbiosis of the gut microbiome may be sufficient to develop PCOS-like symptoms and that modulation of the gut microbiome may be a potential therapeutic target for PCOS.
Collapse
Affiliation(s)
- Maryan G Rizk
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, California, USA
| | - Varykina G Thackray
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, California, USA
| |
Collapse
|
137
|
Gut-Brain Axis Cross-Talk and Limbic Disorders as Biological Basis of Secondary TMAU. J Pers Med 2021; 11:jpm11020087. [PMID: 33572540 PMCID: PMC7912098 DOI: 10.3390/jpm11020087] [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: 01/09/2021] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 02/07/2023] Open
Abstract
Background: Trimethylaminuria (TMAU) is a rare metabolic syndrome characterized by the accumulation and the excretion of trimethylamine (TMA), a volatile diet compound produced by gut microbiota. Gut microbiota alterations are mainly involved in the secondary TMAU, whose patients show also different psychiatric conditions. We hypothesized that the biological activity of several molecules acting as intermediate in TMA metabolic reaction might be at the basis of TMAU psychiatric comorbidities. Methods: To corroborate this hypothesis, we performed the analysis of microbiota of both psychiatric suffering secondary TMAU patients and TMAU "mentally ill" controls, comparing the alteration of metabolites produced by their gut bacteria possibly involved in neurotransmission and, in the same time, belonging to biochemical pathways leading to TMA accumulation. Results: Microbiota analyses showed that Clostridiaceae, Lachnospiraceae and Coriobacteriaceae alterations represented the bacterial families with highest variations. This results in an excessive release of serotonin and an hyperactivation of the vagus nerve that might determine the widest spectrum of psychiatric disorders shown by affected patients. These metabolites, as short chain fatty acids, lactate and neurotransmitter precursors, are also related to TMA accumulation. Conclusions: Knowledge of microbiota-gut-brain axis may become a potential new strategy for improving metabolic diseases and to treat linked psychiatric disorders.
Collapse
|
138
|
Leeming ER, Louca P, Gibson R, Menni C, Spector TD, Le Roy CI. The complexities of the diet-microbiome relationship: advances and perspectives. Genome Med 2021; 13:10. [PMID: 33472701 PMCID: PMC7819159 DOI: 10.1186/s13073-020-00813-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023] Open
Abstract
Personalised dietary modulation of the gut microbiota may be key to disease management. Current investigations provide a broad understanding of the impact of diet on the composition and activity of the gut microbiota, yet detailed knowledge in applying diet as an actionable tool remains limited. Further to the relative novelty of the field, approaches are yet to be standardised and extremely heterogeneous research outcomes have ensued. This may be related to confounders associated with complexities in capturing an accurate representation of both diet and the gut microbiota. This review discusses the intricacies and current methodologies of diet-microbial relations, the implications and limitations of these investigative approaches, and future considerations that may assist in accelerating applications. New investigations should consider improved collection of dietary data, further characterisation of mechanistic interactions, and an increased focus on -omic technologies such as metabolomics to describe the bacterial and metabolic activity of food degradation, together with its crosstalk with the host. Furthermore, clinical evidence with health outcomes is required before therapeutic dietary strategies for microbial amelioration can be made. The potential to reach detailed understanding of diet-microbiota relations may depend on re-evaluation, progression, and unification of research methodologies, which consider the complexities of these interactions.
Collapse
Affiliation(s)
- Emily R Leeming
- The Department of Twin Research, St Thomas' Hospital, King's College London, 3-4th Floor South Wing Block D, Westminster Bridge Road, London, SE1 7EH, UK
| | - Panayiotis Louca
- The Department of Twin Research, St Thomas' Hospital, King's College London, 3-4th Floor South Wing Block D, Westminster Bridge Road, London, SE1 7EH, UK
| | - Rachel Gibson
- Department of Nutritional Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Cristina Menni
- The Department of Twin Research, St Thomas' Hospital, King's College London, 3-4th Floor South Wing Block D, Westminster Bridge Road, London, SE1 7EH, UK
| | - Tim D Spector
- The Department of Twin Research, St Thomas' Hospital, King's College London, 3-4th Floor South Wing Block D, Westminster Bridge Road, London, SE1 7EH, UK.
| | - Caroline I Le Roy
- The Department of Twin Research, St Thomas' Hospital, King's College London, 3-4th Floor South Wing Block D, Westminster Bridge Road, London, SE1 7EH, UK.
| |
Collapse
|
139
|
Lukiw WJ, Arceneaux L, Li W, Bond T, Zhao Y. Gastrointestinal (GI)-Tract Microbiome Derived Neurotoxins and their Potential Contribution to Inflammatory Neurodegeneration in Alzheimer's Disease (AD). JOURNAL OF ALZHEIMER'S DISEASE & PARKINSONISM 2021; 11:525. [PMID: 34457996 PMCID: PMC8395586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The human gastrointestinal (GI)-tract microbiome is a rich, complex and dynamic source of microorganisms that possess a staggering diversity and complexity. Importantly there is a significant variability in microbial complexity even amongst healthy individuals-this has made it difficult to link specific microbial abundance patterns with age-related neurological disease. GI-tract commensal microorganisms are generally beneficial to human metabolism and immunity, however enterotoxigenic forms of microbes possess significant potential to secrete what are amongst the most neurotoxic and pro-inflammatory biopolymers known. These include toxic glycolipids such as lipopolysaccharide (LPS), enterotoxins, microbial-derived amyloids and small non-coding RNA. One major microbial species of the GI-tract microbiome, about ~100-fold more abundant than Escherichia coli in deep GI-tract regions is Bacteroides fragilis, an anaerobic, rod-shaped Gram-negative bacterium. B. fragilis can secrete: (i) a particularly potent, pro-inflammatory and unique LPS subtype (BF-LPS); and (ii) a zinc-metalloproteinase known as B. fragilis-toxin (BFT) or fragilysin. Ongoing studies indicate that BF-LPS and/or BFT disrupt paracellular-and transcellular-barriers by cleavage of intercellular-proteins resulting in 'leaky' barriers. These barriers: (i) become defective and more penetrable with aging and disease; and (ii) permit entry of microbiome-derived neurotoxins into the systemic-circulation from which they next transit the blood-brain barrier and gain access to the CNS. Here LPS accumulates and significantly alters homeostatic patterns of gene expression. The affinity of LPS for neuronal nuclei is significantly enhanced in the presence of amyloid beta 42 (Aβ42) peptides. Recent research on the appearance of the brain thanatomicrobiome at the time of death and the increasing likelihood of a complex brain microbiome are reviewed and discussed. This paper will also highlight some recent advances in this extraordinary research area that links the pro-inflammatory exudates of the GI-tract microbiome with innate-immune disturbances and inflammatory-signaling within the CNS with reference to Alzheimer's disease (AD) wherever possible.
Collapse
Affiliation(s)
- Walter J. Lukiw
- LSU Neuroscience Center, Louisiana State University Health
Sciences Center, New Orleans, LA, United States,Department of Ophthalmology, LSU Health Sciences Center,
New Orleans, LA, United States,Department of Neurology, Louisiana State University Health
Sciences Center, New Orleans, LA, United States,Corresponding author: Dr. Walter J. Lukiw, LSU
Neuroscience Center, Louisiana State University Health Sciences Center, New
Orleans, LA, United States,
| | - Lisa Arceneaux
- LSU Neuroscience Center, Louisiana State University Health
Sciences Center, New Orleans, LA, United States
| | - Wenhong Li
- LSU Neuroscience Center, Louisiana State University Health
Sciences Center, New Orleans, LA, United States,Department of Pharmacology, School of Pharmacy, Jiangxi
University of Traditional Chinese Medicine (TCM), Nanchang, China
| | - Taylor Bond
- LSU Neuroscience Center, Louisiana State University Health
Sciences Center, New Orleans, LA, United States
| | - Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health
Sciences Center, New Orleans, LA, United States,Department of Anatomy and Cell Biology, Louisiana State
University, New Orleans, LA, United States
| |
Collapse
|
140
|
Scott EM, Lewin AC, Leis ML. Current ocular microbiome investigations limit reproducibility and reliability: Critical review and opportunities. Vet Ophthalmol 2020; 24:4-11. [PMID: 33382917 DOI: 10.1111/vop.12854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/13/2020] [Accepted: 12/11/2020] [Indexed: 12/14/2022]
Abstract
Enthusiasm for research describing microbial communities using next-generation sequencing (NGS) has outpaced efforts to standardize methodology. Without consistency in the way research is carried out in this field, the comparison of data between studies is near impossible and the utility of results remains limited. This holds true for bacterial microbiome research of the ocular surface, and other sites, in both humans and animals. In addition, the ocular surface remains under-explored when compared to other mucosal sites. Low bacterial biomass samples from the ocular surface lead to further technical challenges. Taken together, two major problems were identified: (1) Normalization of the workflow in studies utilizing NGS to investigate the ocular surface bacteriome is necessary in order to propel the field forward and improve research impact through cross-study comparisons. (2) Current microbiome profiling technology was developed for high bacterial biomass samples (such as feces or soil), posing a challenge for analyses of samples with low bacterial load such as the ocular surface. This article reviews the challenges and limitations currently facing ocular microbiome research and provides recommendations for minimum reporting standards for veterinary ophthalmologists and clinician scientists to limit inter-study variation, improve reproducibility, and ultimately render results from these studies more impactful. The move toward normalization of methodology will expedite and maximize the potential for microbiome research to translate into meaningful discovery and tangible clinical applications.
Collapse
Affiliation(s)
- Erin M Scott
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Andrew C Lewin
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Marina L Leis
- Department of Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
141
|
Mamic P, Chaikijurajai T, Tang WHW. Gut microbiome - A potential mediator of pathogenesis in heart failure and its comorbidities: State-of-the-art review. J Mol Cell Cardiol 2020; 152:105-117. [PMID: 33307092 DOI: 10.1016/j.yjmcc.2020.12.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/22/2020] [Accepted: 12/02/2020] [Indexed: 12/12/2022]
Abstract
Gut microbiome (GMB) has been increasingly recognized as a contributor to development and progression of heart failure (HF), immune-mediated subtypes of cardiomyopathy (myocarditis and anthracycline-induced cardiotoxicity), response to certain cardiovascular drugs, and HF-related comorbidities, such as chronic kidney disease, cardiorenal syndrome, insulin resistance, malnutrition, and cardiac cachexia. Gut microbiome is also responsible for the "gut hypothesis" of HF, which explains the adverse effects of gut barrier dysfunction and translocation of GMB on the progression of HF. Furthermore, accumulating evidence has suggested that gut microbial metabolites, including short chain fatty acids, trimethylamine N-oxide (TMAO), amino acid metabolites, and bile acids, are mechanistically linked to pathogenesis of HF, and could, therefore, serve as potential therapeutic targets for HF. Even though there are a variety of proposed therapeutic approaches, such as dietary modifications, prebiotics, probiotics, TMAO synthesis inhibitors, and fecal microbial transplant, targeting GMB in HF is still in its infancy and, indeed, requires further preclinical and clinical evidence. In this review, we aim to highlight the role gut microbiome plays in HF pathophysiology and its potential as a novel therapeutic target in HF.
Collapse
Affiliation(s)
- Petra Mamic
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University Medical Center, Stanford, CA, United States of America
| | - Thanat Chaikijurajai
- Kaufman Center for Heart Failure Treatment and Recovery, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, United States of America; Department of Internal Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - W H Wilson Tang
- Kaufman Center for Heart Failure Treatment and Recovery, Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, United States of America.
| |
Collapse
|
142
|
Smith PE, Waters SM, Gómez Expósito R, Smidt H, Carberry CA, McCabe MS. Synthetic Sequencing Standards: A Guide to Database Choice for Rumen Microbiota Amplicon Sequencing Analysis. Front Microbiol 2020; 11:606825. [PMID: 33363527 PMCID: PMC7752867 DOI: 10.3389/fmicb.2020.606825] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/05/2020] [Indexed: 01/04/2023] Open
Abstract
Our understanding of complex microbial communities, such as those residing in the rumen, has drastically advanced through the use of high throughput sequencing (HTS) technologies. Indeed, with the use of barcoded amplicon sequencing, it is now cost effective and computationally feasible to identify individual rumen microbial genera associated with ruminant livestock nutrition, genetics, performance and greenhouse gas production. However, across all disciplines of microbial ecology, there is currently little reporting of the use of internal controls for validating HTS results. Furthermore, there is little consensus of the most appropriate reference database for analyzing rumen microbiota amplicon sequencing data. Therefore, in this study, a synthetic rumen-specific sequencing standard was used to assess the effects of database choice on results obtained from rumen microbial amplicon sequencing. Four DADA2 reference training sets (RDP, SILVA, GTDB, and RefSeq + RDP) were compared to assess their ability to correctly classify sequences included in the rumen-specific sequencing standard. In addition, two thresholds of phylogenetic bootstrapping, 50 and 80, were applied to investigate the effect of increasing stringency. Sequence classification differences were apparent amongst the databases. For example the classification of Clostridium differed between all databases, thus highlighting the need for a consistent approach to nomenclature amongst different reference databases. It is hoped the effect of database on taxonomic classification observed in this study, will encourage research groups across various microbial disciplines to develop and routinely use their own microbiome-specific reference standard to validate analysis pipelines and database choice.
Collapse
Affiliation(s)
- Paul E Smith
- Teagasc Animal and Bioscience Research Department, Teagasc Grange, Meath, Ireland.,UCD School of Agricultural and Food Science, University College Dublin, Dublin, Ireland
| | - Sinead M Waters
- Teagasc Animal and Bioscience Research Department, Teagasc Grange, Meath, Ireland
| | - Ruth Gómez Expósito
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, Netherlands
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, Netherlands
| | - Ciara A Carberry
- Teagasc Animal and Bioscience Research Department, Teagasc Grange, Meath, Ireland
| | - Matthew S McCabe
- Teagasc Animal and Bioscience Research Department, Teagasc Grange, Meath, Ireland
| |
Collapse
|
143
|
Chen YA, Park J, Natsume-Kitatani Y, Kawashima H, Mohsen A, Hosomi K, Tanisawa K, Ohno H, Konishi K, Murakami H, Miyachi M, Kunisawa J, Mizuguchi K. MANTA, an integrative database and analysis platform that relates microbiome and phenotypic data. PLoS One 2020; 15:e0243609. [PMID: 33275647 PMCID: PMC7717536 DOI: 10.1371/journal.pone.0243609] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 11/24/2020] [Indexed: 11/18/2022] Open
Abstract
With an ever-increasing interest in understanding the relationships between the microbiota and the host, more tools to map, analyze and interpret these relationships have been developed. Most of these tools, however, focus on taxonomic profiling and comparative analysis among groups, with very few analytical tools designed to correlate microbiota and the host phenotypic data. We have developed a software program for creating a web-based integrative database and analysis platform called MANTA (Microbiota And pheNoType correlation Analysis platform). In addition to storing the data, MANTA is equipped with an intuitive user interface that can be used to correlate the microbial composition with phenotypic parameters. Using a case study, we demonstrated that MANTA was able to quickly identify the significant correlations between microbial abundances and phenotypes that are supported by previous studies. Moreover, MANTA enabled the users to quick access locally stored data that can help interpret microbiota-phenotype relations. MANTA is available at https://mizuguchilab.org/manta/ for download and the source code can be found at https://github.com/chenyian-nibio/manta.
Collapse
Affiliation(s)
- Yi-An Chen
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
- * E-mail: (YC); (KM)
| | - Jonguk Park
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Yayoi Natsume-Kitatani
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Hitoshi Kawashima
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Attayeb Mohsen
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Kumpei Tanisawa
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Shinjuku, Tokyo, Japan
| | - Harumi Ohno
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Shinjuku, Tokyo, Japan
| | - Kana Konishi
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Shinjuku, Tokyo, Japan
| | - Haruka Murakami
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Shinjuku, Tokyo, Japan
| | - Motohiko Miyachi
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Shinjuku, Tokyo, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Kenji Mizuguchi
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
- * E-mail: (YC); (KM)
| |
Collapse
|
144
|
Bokulich NA, Ziemski M, Robeson MS, Kaehler BD. Measuring the microbiome: Best practices for developing and benchmarking microbiomics methods. Comput Struct Biotechnol J 2020; 18:4048-4062. [PMID: 33363701 PMCID: PMC7744638 DOI: 10.1016/j.csbj.2020.11.049] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 12/12/2022] Open
Abstract
Microbiomes are integral components of diverse ecosystems, and increasingly recognized for their roles in the health of humans, animals, plants, and other hosts. Given their complexity (both in composition and function), the effective study of microbiomes (microbiomics) relies on the development, optimization, and validation of computational methods for analyzing microbial datasets, such as from marker-gene (e.g., 16S rRNA gene) and metagenome data. This review describes best practices for benchmarking and implementing computational methods (and software) for studying microbiomes, with particular focus on unique characteristics of microbiomes and microbiomics data that should be taken into account when designing and testing microbiomics methods.
Collapse
Affiliation(s)
- Nicholas A. Bokulich
- Laboratory of Food Systems Biotechnology, Institute of Food, Nutrition, and Health, ETH Zurich, Switzerland
| | - Michal Ziemski
- Laboratory of Food Systems Biotechnology, Institute of Food, Nutrition, and Health, ETH Zurich, Switzerland
| | - Michael S. Robeson
- University of Arkansas for Medical Sciences, Department of Biomedical Informatics, Little Rock, AR, USA
| | | |
Collapse
|
145
|
Keskey R, Cone JT, DeFazio JR, Alverdy JC. The use of fecal microbiota transplant in sepsis. Transl Res 2020; 226:12-25. [PMID: 32649987 PMCID: PMC7572598 DOI: 10.1016/j.trsl.2020.07.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/30/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022]
Abstract
Sepsis is defined as a dysregulated inflammatory response, which ultimately results from a perturbed interaction of both an altered immune system and the biomass and virulence of involved pathogens. This response has been tied to the intestinal microbiota, as the microbiota and its associated metabolites play an essential role in regulating the host immune response to infection. In turn, critical illness as well as necessary health care treatments result in a collapse of the intestinal microbiota diversity and a subsequent loss of health-promoting short chain fatty acids, such as butyrate, leading to the development of a maladaptive pathobiome. These perturbations of the microbiota contribute to the dysregulated immune response and organ failure associated with sepsis. Several case series have reported the ability of fecal microbiota transplant (FMT) to restore the host immune response and aid in recovery of septic patients. Additionally, animal studies have revealed the mechanism of FMT rescue in sepsis is likely related to the ability of FMT to restore butyrate producing bacteria and alter the innate immune response aiding in pathogen clearance. However, several studies have reported lethal complications associated with FMT, including bacteremia. Therefore, FMT in the treatment of sepsis is and should remain investigational until a more detailed mechanism of how FMT restores the host immune response in sepsis is determined, allowing for the development of more fine-tuned microbiota therapies.
Collapse
Affiliation(s)
- Robert Keskey
- Section of General Surgery, Department of Surgery, University of Chicago, Chicago, Illinois
| | - Jennifer T Cone
- Section of Trauma and Acute Care Surgery, Department of Surgery, University of Chicago, Chicago, Illinois
| | - Jennifer R DeFazio
- Division of Pediatric Surgery, New York-Presbyterian Morgan Stanley Children's Hospital, Columbia University Medical Center, New York, New York
| | - John C Alverdy
- Section of General Surgery, Department of Surgery, University of Chicago, Chicago, Illinois.
| |
Collapse
|
146
|
Ruck CE, Odumade OA, Smolen KK. Vaccine Interactions With the Infant Microbiome: Do They Define Health and Disease? Front Pediatr 2020; 8:565368. [PMID: 33324590 PMCID: PMC7725791 DOI: 10.3389/fped.2020.565368] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022] Open
Abstract
Over the past decade, there has been a growing awareness of the vital role of the microbiome in the function of the immune system. Recently, several studies have demonstrated a relationship between the composition of the microbiome and the vaccine-specific immune response. As a result of these findings, the administration of probiotics has been proposed as a means of boosting vaccine-specific immunity. Early results have so far been highly inconsistent, with little evidence of sustained benefit. To date, a precise determination of the aspects of the microbiome that impact immunity is still lacking, and the mechanisms of action are also unknown. Further investigations into these questions are necessary to effectively manipulate the microbiome for the purpose of boosting immunity and enhancing vaccine-specific responses in infants. In this review, we summarize recent studies aimed at altering the neonatal gut microbiome to enhance vaccine responses and highlight gaps in knowledge and understanding. We also discuss research strategies aimed at filling these gaps and developing potential therapeutic interventions.
Collapse
Affiliation(s)
- Candice E. Ruck
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Oludare A. Odumade
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Division of Medicine Critical Care, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Kinga K. Smolen
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Institute for Medical Immunology, Université libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
147
|
Ballini A, Scacco S, Boccellino M, Santacroce L, Arrigoni R. Microbiota and Obesity: Where Are We Now? BIOLOGY 2020; 9:biology9120415. [PMID: 33255588 PMCID: PMC7761345 DOI: 10.3390/biology9120415] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/14/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023]
Abstract
Simple Summary Emerging new data reported in the international scientific literature show that specific alterations in the human gut microbiota are characteristic in obesity and obesity-related metabolic diseases. Obesity is conditioned by a multitude of factors, and the microbiota is certainly an important player. The analysis of the data obtained from experimental studies allow us to hypothesize that changes in the composition of the microbiota may be the cause, and not simply the consequence, of alterations in human metabolism. Clinical trials on wide samples that investigate the role of diet-induced modulation of the gut microbiota on the host metabolism are needed to understand the interactions at the molecular level for the observed correlations between metabolism and microbiota changes. Abstract Genetic and environmental factors are underlying causes of obesity and other metabolic diseases, so it is therefore difficult to find suitable and effective medical treatments. However, without a doubt, the gut microbiota—and also the bacteria present in the oral cavity—act as key factors in the development of these pathologies, yet the mechanisms have not been fully described. Certainly, a more detailed knowledge of the structure of the microbiota—composition, intra- and inter-species relationships, metabolic functions—could be of great help in counteracting the onset of obesity. Identifying key bacterial species will allow us to create a database of “healthy” bacteria, making it possible to manipulate the bacterial community according to metabolic and clinical needs. Targeting gut microbiota in clinical care as treatment for obesity and health-related complications—even just for weight loss has become a real possibility. In this topical review we provide an overview of the role of the microbiota on host energy homeostasis and obesity-related metabolic diseases, therefore addressing the therapeutic potential of novel and existing strategies (impact of nutrition/dietary modulation, and fecal microbiota transplantation) in the treatment of metabolic disease.
Collapse
Affiliation(s)
- Andrea Ballini
- Department of Biosciences, Biotechnologies and Biopharmaceutics, Campus Universitario, University of Bari “Aldo Moro”, 70125 Bari, Italy;
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70121 Bari, Italy
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Salvatore Scacco
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70121 Bari, Italy
- Correspondence: (S.S.); (M.B.); (R.A.)
| | - Mariarosaria Boccellino
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Correspondence: (S.S.); (M.B.); (R.A.)
| | - Luigi Santacroce
- Microbiology and Virology Laboratory, Ionian Department, Policlinico University Hospital, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Roberto Arrigoni
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), 70124 Bari, Italy
- Correspondence: (S.S.); (M.B.); (R.A.)
| |
Collapse
|
148
|
Ramirez J, Guarner F, Bustos Fernandez L, Maruy A, Sdepanian VL, Cohen H. Antibiotics as Major Disruptors of Gut Microbiota. Front Cell Infect Microbiol 2020; 10:572912. [PMID: 33330122 PMCID: PMC7732679 DOI: 10.3389/fcimb.2020.572912] [Citation(s) in RCA: 417] [Impact Index Per Article: 83.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022] Open
Abstract
Advances in culture-independent research techniques have led to an increased understanding of the gut microbiota and the role it plays in health and disease. The intestine is populated by a complex microbial community that is organized around a network of metabolic interdependencies. It is now understood that the gut microbiota is vital for normal development and functioning of the human body, especially for the priming and maturation of the adaptive immune system. Antibiotic use can have several negative effects on the gut microbiota, including reduced species diversity, altered metabolic activity, and the selection of antibiotic-resistant organisms, which in turn can lead to antibiotic-associated diarrhea and recurrent Clostridioides difficile infections. There is also evidence that early childhood exposure to antibiotics can lead to several gastrointestinal, immunologic, and neurocognitive conditions. The increase in the use of antibiotics in recent years suggests that these problems are likely to become more acute or more prevalent in the future. Continued research into the structure and function of the gut microbiota is required to address this challenge.
Collapse
Affiliation(s)
- Jaime Ramirez
- Gastroenterology and Nutrition Department, Instituto Nacional de Pediatria, Mexico City, Mexico.,Facultad Nacional de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Francisco Guarner
- Digestive System Research Unit, Vall d'Hebron Institute of Research (VHIR), Barcelona, Spain
| | - Luis Bustos Fernandez
- Instituto de Gastroenterologia, Centro Medico Bustos Fernandez (CMBF), Buenos Aires, Argentina
| | - Aldo Maruy
- Catedra de Pediatria, Hospital Cayetano Heredia, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Vera Lucia Sdepanian
- Division of Pediatric Gastroenterology, Pediatric Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Henry Cohen
- Gastroenterology, National School of Medicine, Montevideo, Uruguay
| |
Collapse
|
149
|
Insects' potential: Understanding the functional role of their gut microbiome. J Pharm Biomed Anal 2020; 194:113787. [PMID: 33272789 DOI: 10.1016/j.jpba.2020.113787] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 12/17/2022]
Abstract
The study of insect-associated microbial communities is a field of great importance in agriculture, principally because of the role insects play as pests. In addition, there is a recent focus on the potential of the insect gut microbiome in areas such as biotechnology, given some microorganisms produce molecules with biotechnological and industrial applications, and also in biomedicine, since some bacteria and fungi are a reservoir of antibiotic resistance genes (ARGs). To date, most studies aiming to characterize the role of the gut microbiome of insects have been based on high-throughput sequencing of the 16S rRNA gene and/or metagenomics. However, recently functional approaches such as metatranscriptomics, metaproteomics and metabolomics have also been employed. Besides providing knowledge about the taxonomic distribution of microbial populations, these techniques also reveal their functional and metabolic capabilities. This information is essential to gain a better understanding of the role played by microbes comprising the microbial communities in their hosts, as well as to indicate their possible exploitation. This review provides an overview of how far we have come in characterizing insect gut functionality through omics, as well as the challenges and future perspectives in this field.
Collapse
|
150
|
Araujo DV, Watson GA, Oliva M, Heirali A, Coburn B, Spreafico A, Siu LL. Bugs as drugs: The role of microbiome in cancer focusing on immunotherapeutics. Cancer Treat Rev 2020; 92:102125. [PMID: 33227623 DOI: 10.1016/j.ctrv.2020.102125] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 11/01/2020] [Accepted: 11/02/2020] [Indexed: 12/16/2022]
Abstract
The human microbiome comprising microorganisms, their collective genomes and metabolic products has gained tremendous research interest in oncology, as multiple cohorts and case studies have demonstrated discernible interpatient differences in this ecosystem based on clinical variables including disease type, stage, diet, antibiotic usage, cancer treatments, therapeutic responses and toxicities. The modulation of the gut microbiome is the subject of many ongoing preclinical and clinical investigations, through the manipulation of diet, as well as the use of prebiotics, probiotics, specific antibiotics, fecal microbial transplantation, microbial consortia and stool substitutes. Standardization and quality control are needed to maximize the information being generated in this growing field, ranging from technical assays to measure microbiome composition, to methodological aspects in the analysis and reporting of results. Proof-of-mechanism and proof-of-concept clinical trials with appropriate controls are needed to confirm or refute the feasibility, safety and ultimately the clinical utility of human microbiome modulation in cancer patients.
Collapse
Affiliation(s)
- Daniel V Araujo
- Princess Margaret Cancer Centre, Division of Medical Oncology and Hematology, University Health Network, Toronto, Canada
| | - Geoffrey A Watson
- Princess Margaret Cancer Centre, Division of Medical Oncology and Hematology, University Health Network, Toronto, Canada
| | - Marc Oliva
- Catalan Institute of Oncology (Hospital Duran i Reynals), Department of Medical Oncology, IDIBELL, Barcelona, Spain
| | - Alya Heirali
- Toronto General Research Institute, Division of Infectious Diseases, University Health Network, Toronto, Canada
| | - Bryan Coburn
- Toronto General Research Institute, Division of Infectious Diseases, University Health Network, Toronto, Canada
| | - Anna Spreafico
- Princess Margaret Cancer Centre, Division of Medical Oncology and Hematology, University Health Network, Toronto, Canada
| | - Lillian L Siu
- Princess Margaret Cancer Centre, Division of Medical Oncology and Hematology, University Health Network, Toronto, Canada.
| |
Collapse
|