601
|
Deaver JA, Eum SY, Toborek M. Circadian Disruption Changes Gut Microbiome Taxa and Functional Gene Composition. Front Microbiol 2018; 9:737. [PMID: 29706947 PMCID: PMC5909328 DOI: 10.3389/fmicb.2018.00737] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 03/29/2018] [Indexed: 12/22/2022] Open
Abstract
Disrupted circadian rhythms and alterations of the gut microbiome composition were proposed to affect host health. Therefore, the aim of this research was to identify whether these events are connected and if circadian rhythm disruption by abnormal light–dark (LD) cycles affects microbial community gene expression and host vulnerability to intestinal dysfunction. Mice were subjected to either a 4-week period of constant 24-h light or of normal 12-h LD cycles. Stool samples were collected at the beginning and after the circadian rhythm disruption. A metatranscriptomic analysis revealed an increase in Ruminococcus torques, a bacterial species known to decrease gut barrier integrity, and a decrease in Lactobacillus johnsonii, a bacterium that helps maintain the intestinal epithelial cell layer, after circadian rhythm disruption. In addition, genes involved in pathways promoting host beneficial immune responses were downregulated, while genes involved in the synthesis and transportation of the endotoxin lipopolysaccharide were upregulated in mice with disrupted circadian cycles. Importantly, these mice were also more prone to dysfunction of the intestinal barrier. These results further elucidate the impact of light-cycle disruption on the gut microbiome and its connection with increased incidence of disease in response to circadian rhythm disturbances.
Collapse
Affiliation(s)
- Jessica A Deaver
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sung Y Eum
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
602
|
Shkoporov AN, Ryan FJ, Draper LA, Forde A, Stockdale SR, Daly KM, McDonnell SA, Nolan JA, Sutton TD, Dalmasso M, McCann A, Ross RP, Hill C. Reproducible protocols for metagenomic analysis of human faecal phageomes. MICROBIOME 2018; 6:68. [PMID: 29631623 PMCID: PMC5892011 DOI: 10.1186/s40168-018-0446-z] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 03/13/2018] [Indexed: 05/07/2023]
Abstract
BACKGROUND Recent studies have demonstrated that the human gut is populated by complex, highly individual and stable communities of viruses, the majority of which are bacteriophages. While disease-specific alterations in the gut phageome have been observed in IBD, AIDS and acute malnutrition, the human gut phageome remains poorly characterised. One important obstacle in metagenomic studies of the human gut phageome is a high level of discrepancy between results obtained by different research groups. This is often due to the use of different protocols for enriching virus-like particles, nucleic acid purification and sequencing. The goal of the present study is to develop a relatively simple, reproducible and cost-efficient protocol for the extraction of viral nucleic acids from human faecal samples, suitable for high-throughput studies. We also analyse the effect of certain potential confounding factors, such as storage conditions, repeated freeze-thaw cycles, and operator bias on the resultant phageome profile. Additionally, spiking of faecal samples with an exogenous phage standard was employed to quantitatively analyse phageomes following metagenomic sequencing. Comparative analysis of phageome profiles to bacteriome profiles was also performed following 16S rRNA amplicon sequencing. RESULTS Faecal phageome profiles exhibit an overall greater individual specificity when compared to bacteriome profiles. The phageome and bacteriome both exhibited moderate change when stored at + 4 °C or room temperature. Phageome profiles were less impacted by multiple freeze-thaw cycles than bacteriome profiles, but there was a greater chance for operator effect in phageome processing. The successful spiking of faecal samples with exogenous bacteriophage demonstrated large variations in the total viral load between individual samples. CONCLUSIONS The faecal phageome sequencing protocol developed in this study provides a valuable additional view of the human gut microbiota that is complementary to 16S amplicon sequencing and/or metagenomic sequencing of total faecal DNA. The protocol was optimised for several confounding factors that are encountered while processing faecal samples, to reduce discrepancies observed within and between research groups studying the human gut phageome. Rapid storage, limited freeze-thaw cycling and spiking of faecal samples with an exogenous phage standard are recommended for optimum results.
Collapse
Affiliation(s)
| | - Feargal J. Ryan
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | | | - Amanda Forde
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Stephen R. Stockdale
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork Ireland
| | - Karen M. Daly
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | | | - James A. Nolan
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | | | - Marion Dalmasso
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Normandie Univ, UNICAEN, EA4651 ABTE, F-14032 Caen, France
| | - Angela McCann
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - R. Paul Ross
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Colin Hill
- APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|
603
|
Gautam A, Kumar R, Chakraborty N, Muhie S, Hoke A, Hammamieh R, Jett M. Altered fecal microbiota composition in all male aggressor-exposed rodent model simulating features of post-traumatic stress disorder. J Neurosci Res 2018; 96:1311-1323. [PMID: 29633335 DOI: 10.1002/jnr.24229] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 01/25/2018] [Accepted: 02/08/2018] [Indexed: 12/21/2022]
Abstract
The bidirectional role of gut-brain axis that integrates the gut and central nervous system activities has recently been investigated. We studied "cage-within-cage resident-intruder" all-male model, where subject male mice (C57BL/6J) are exposed to aggressor mice (SJL albino), and gut microbiota-derived metabolites were identified in plasma after 10 days of exposure. We assessed 16S ribosomal RNA gene from fecal samples collected daily from these mice during the 10-day study. Alpha diversity using Chao indices indicated no change in diversity in aggressor-exposed samples. The abundance profile showed the top phyla were Firmicutes and Bacteroidetes, Tenericutes, Verrucomicrobia, Actinobacteria and Proteobacteria, respectively. The phyla Firmicutes and Bacteroidetes are vulnerable to PTSD-eliciting stress and the Firmicutes/Bacteroidetes ratio increases with stress. Principal coordinate analysis showed the control and aggressor-exposed samples cluster separately where samples from early time points (day 1-3) clustered together and were distinct from late time points (day 4-9). The genus-based analysis revealed all control time points clustered together and aggressor-exposed samples had multiple clusters. The decrease in proportion of Firmicutes after aggressor exposure persisted throughout the study. The proportion of Verrucomicrobia immediately decreased and was significantly shifted at most of the later time points. The genus Oscillospira, Lactobacillus, Akkermansia and Anaeroplasma are the top four genera that differed between control and stressor-exposed mice. The data showed immediate effect on microbiome composition during a 10 day time period of stress exposure. Studying the longitudinal effects of a stressor is an important step toward an improved mechanistic understanding of the microbiome dynamics.
Collapse
Affiliation(s)
- Aarti Gautam
- US Army Center for Environmental Health Research, Fort Detrick, MD, USA
| | - Raina Kumar
- US Army Center for Environmental Health Research, Fort Detrick, MD, USA.,Advanced Biomedical Computing Center, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Nabarun Chakraborty
- US Army Center for Environmental Health Research, Fort Detrick, MD, USA.,The Geneva Foundation, Fort Detrick, MD, USA
| | - Seid Muhie
- US Army Center for Environmental Health Research, Fort Detrick, MD, USA.,The Geneva Foundation, Fort Detrick, MD, USA
| | - Allison Hoke
- US Army Center for Environmental Health Research, Fort Detrick, MD, USA.,The Oak Ridge Institute for Science and Education, Fort Detrick, MD, USA
| | - Rasha Hammamieh
- US Army Center for Environmental Health Research, Fort Detrick, MD, USA
| | - Marti Jett
- US Army Center for Environmental Health Research, Fort Detrick, MD, USA
| |
Collapse
|
604
|
Grosicki GJ, Fielding RA, Lustgarten MS. Gut Microbiota Contribute to Age-Related Changes in Skeletal Muscle Size, Composition, and Function: Biological Basis for a Gut-Muscle Axis. Calcif Tissue Int 2018; 102:433-442. [PMID: 29058056 PMCID: PMC5858871 DOI: 10.1007/s00223-017-0345-5] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/13/2017] [Indexed: 12/12/2022]
Abstract
Skeletal muscle is a highly plastic tissue that plays a central role in human health and disease. Aging is associated with a decrease in muscle mass and function (sarcopenia) that is associated with a loss of independence and reduced quality of life. Gut microbiota, the bacteria, archaea, viruses, and eukaryotic microbes residing in the gastrointestinal tract are emerging as a potential contributor to age-associated muscle decline. Specifically, advancing age is characterized by a dysbiosis of gut microbiota that is associated with increased intestinal permeability, facilitating the passage of endotoxin and other microbial products (e.g., indoxyl sulfate) into the circulation. Upon entering the circulation, LPS and other microbial factors promote inflammatory signaling and skeletal muscle changes that are hallmarks of the aging muscle phenotype. This review will summarize existing literature suggesting cross-talk between gut microbiota and skeletal muscle health, with emphasis on the significance of this axis for mediating changes in aging skeletal muscle size, composition, and function.
Collapse
Affiliation(s)
- Gregory J Grosicki
- Nutrition, Exercise Physiology, and Sarcopenia Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Roger A Fielding
- Nutrition, Exercise Physiology, and Sarcopenia Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Michael S Lustgarten
- Nutrition, Exercise Physiology, and Sarcopenia Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA.
| |
Collapse
|
605
|
Codella R, Luzi L, Terruzzi I. Exercise has the guts: How physical activity may positively modulate gut microbiota in chronic and immune-based diseases. Dig Liver Dis 2018; 50:331-341. [PMID: 29233686 DOI: 10.1016/j.dld.2017.11.016] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/14/2017] [Accepted: 11/18/2017] [Indexed: 12/11/2022]
Abstract
Limited animal and human research findings suggests that exercise might have a beneficial role for health gut. Cardiorespiratory fitness correlates with health-associated gut parameters such as taxonomic diversity and richness. Physical exercise may augment intestinal microbial diversity through several mechanisms including promotion of an anti-inflammatory state. Disease-associated microbial functions were linked to distinct taxa in previous studies of familial type 1 diabetes mellitus (T1D). An integrated multi-approach in the study of T1D, including physical exercise, is advocated. The present review explores how exercise might modulate gut microbiota and microbiome characteristics in chronic and immune-based diseases, given the demonstrated relationship between gut function and human health.
Collapse
Affiliation(s)
- Roberto Codella
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy; Metabolism Research Center, IRCCS Policlinico San Donato, San Donato Milanese, Italy.
| | - Livio Luzi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy; Metabolism Research Center, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Ileana Terruzzi
- Diabetes Research Institute, Metabolism, Nutrigenomics and Cellular Differentiation Unit, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
606
|
Wolff NS, Hugenholtz F, Wiersinga WJ. The emerging role of the microbiota in the ICU. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2018; 22:78. [PMID: 29559006 PMCID: PMC5861706 DOI: 10.1186/s13054-018-1999-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This article is one of ten reviews selected from the Annual Update in Intensive Care and Emergency Medicine 2018. Other selected articles can be found online at https://www.biomedcentral.com/collections/annualupdate2018. Further information about the Annual Update in Intensive Care and Emergency Medicine is available from http://www.springer.com/series/8901.
Collapse
Affiliation(s)
- Nora Suzanne Wolff
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, Netherlands
| | - Floor Hugenholtz
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, Netherlands
| | - Willem Joost Wiersinga
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, Netherlands. .,Department of Medicine, Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
607
|
San-Juan-Vergara H, Zurek E, Ajami NJ, Mogollon C, Peña M, Portnoy I, Vélez JI, Cadena-Cruz C, Diaz-Olmos Y, Hurtado-Gómez L, Sanchez-Sit S, Hernández D, Urruchurtu I, Di-Ruggiero P, Guardo-García E, Torres N, Vidal-Orjuela O, Viasus D, Petrosino JF, Cervantes-Acosta G. A Lachnospiraceae-dominated bacterial signature in the fecal microbiota of HIV-infected individuals from Colombia, South America. Sci Rep 2018; 8:4479. [PMID: 29540734 PMCID: PMC5852036 DOI: 10.1038/s41598-018-22629-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 02/05/2018] [Indexed: 02/08/2023] Open
Abstract
HIV infection has a tremendous impact on the immune system's proper functioning. The mucosa-associated lymphoid tissue (MALT) is significantly disarrayed during HIV infection. Compositional changes in the gut microbiota might contribute to the mucosal barrier disruption, and consequently to microbial translocation. We performed an observational, cross-sectional study aimed at evaluating changes in the fecal microbiota of HIV-infected individuals from Colombia. We analyzed the fecal microbiota of 37 individuals via 16S rRNA gene sequencing; 25 HIV-infected patients and 12 control (non-infected) individuals, which were similar in body mass index, age, gender balance and socioeconomic status. To the best of our knowledge, no such studies have been conducted in Latin American countries. Given its compositional nature, microbiota data were normalized and transformed using Aitchison's Centered Log-Ratio. Overall, a change in the network structure in HIV-infected patients was revealed by using the SPIEC-EASI MB tool. Genera such as Blautia, Dorea, Yersinia, Escherichia-Shigella complex, Staphylococcus, and Bacteroides were highly relevant in HIV-infected individuals. Differential abundance analysis by both sparse Partial Least Square-Discriminant Analysis and Random Forest identified a greater abundance of Lachnospiraceae-OTU69, Blautia, Dorea, Roseburia, and Erysipelotrichaceae in HIV-infected individuals. We show here, for the first time, a predominantly Lachnospiraceae-based signature in HIV-infected individuals.
Collapse
Affiliation(s)
| | - Eduardo Zurek
- División de Ingenierías, Fundación Universidad del Norte, Barranquilla, Colombia
| | - Nadim J Ajami
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | | | - Mario Peña
- División Ciencias de la Salud, Fundación Universidad del Norte, Barranquilla, Colombia
| | - Ivan Portnoy
- División de Ingenierías, Fundación Universidad del Norte, Barranquilla, Colombia
| | - Jorge I Vélez
- División de Ingenierías, Fundación Universidad del Norte, Barranquilla, Colombia
| | - Christian Cadena-Cruz
- División Ciencias de la Salud, Fundación Universidad del Norte, Barranquilla, Colombia
| | - Yirys Diaz-Olmos
- División Ciencias de la Salud, Fundación Universidad del Norte, Barranquilla, Colombia
| | - Leidy Hurtado-Gómez
- División Ciencias de la Salud, Fundación Universidad del Norte, Barranquilla, Colombia
| | - Silvana Sanchez-Sit
- Maestría en Estadística Aplicada, Universidad del Norte, Barranquilla, Colombia
| | | | | | | | | | | | - Oscar Vidal-Orjuela
- División Ciencias de la Salud, Fundación Universidad del Norte, Barranquilla, Colombia
| | - Diego Viasus
- División Ciencias de la Salud, Fundación Universidad del Norte, Barranquilla, Colombia
| | - Joseph F Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | | |
Collapse
|
608
|
Gomez-Arango LF, Barrett HL, Wilkinson SA, Callaway LK, McIntyre HD, Morrison M, Dekker Nitert M. Low dietary fiber intake increases Collinsella abundance in the gut microbiota of overweight and obese pregnant women. Gut Microbes 2018; 9:189-201. [PMID: 29144833 PMCID: PMC6219589 DOI: 10.1080/19490976.2017.1406584] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
UNLABELLED The gut microbiota contributes to the regulation of glucose metabolism in pregnancy. Abundance of the genus Collinsella is positively correlated with circulating insulin; however, it is unclear what determines Collinsella abundance. This study aims to validate the correlation between Collinsella and insulin and to elucidate if macronutrient intake alters Collinsella abundance and gut microbiota composition. Gut microbiota profiles were assessed by 16S rRNA sequencing in 57 overweight and 73 obese pregnant women from the SPRING (Study of PRobiotics IN Gestational diabetes) trial at 16 weeks gestation and correlated with metabolic hormone levels and macronutrient intake. Gut microbiota composition in the top and bottom 10% of dietary fiber intake was evaluated through network analysis. Collinsella abundance correlated positively with circulating insulin (rho = 0.30, p = 0.0006), independent of maternal BMI, but negatively with dietary fiber intake (rho = -0.20, p = 0.025) in this cohort. Low dietary fiber intake was associated with a gut microbiota favoring lactate fermentation while high fiber intake promotes short-chain fatty acid-producing bacteria. Low dietary fiber may enable overgrowth of Collinsella and alter the overall fermentation pattern in gut microbiota. This suggests that dietary choices during pregnancy can modify the nutritional ecology of the gut microbiota, with potential deleterious effects on the metabolic and inflammatory health of the host. TRIAL REGISTRATION ANZCTR 12611001208998, registered 23/11/2011.
Collapse
Affiliation(s)
- Luisa F. Gomez-Arango
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Australia,Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Helen L. Barrett
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Australia,Faculty of Medicine, The University of Queensland, Brisbane, Australia,Obstetric Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Shelley A. Wilkinson
- Mater Health Services, Nutrition and Dietetics, Mater Hospital, Brisbane, Australia,Mater Research Institute –University of Queensland, Brisbane, Australia
| | - Leonie K. Callaway
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Australia,Faculty of Medicine, The University of Queensland, Brisbane, Australia,Obstetric Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - H. David McIntyre
- Faculty of Medicine, The University of Queensland, Brisbane, Australia,Mater Research Institute –University of Queensland, Brisbane, Australia
| | - Mark Morrison
- Faculty of Medicine, The University of Queensland, Brisbane, Australia,Diamantina Institute, The University of Queensland, Brisbane, Australia,School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Marloes Dekker Nitert
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Australia,Diamantina Institute, The University of Queensland, Brisbane, Australia,CONTACT: Marloes Dekker Nitert School of Chemistry and Molecular Biosciences, Building 76–452. The University of Queensland Brisbane, QLD 4072, Australia
| |
Collapse
|
609
|
Calvani R, Picca A, Lo Monaco MR, Landi F, Bernabei R, Marzetti E. Of Microbes and Minds: A Narrative Review on the Second Brain Aging. Front Med (Lausanne) 2018; 5:53. [PMID: 29552561 PMCID: PMC5840854 DOI: 10.3389/fmed.2018.00053] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 02/15/2018] [Indexed: 12/21/2022] Open
Abstract
In recent years, an extensive body of literature focused on the gut-brain axis and the possible role played by the gut microbiota in modulating brain morphology and function from birth to old age. Gut microbiota has been proposed as a relevant player during the early phases of neurodevelopment, with possible long-standing effects in later life. The reduction in gut microbiota diversity has also become one of the hallmarks of aging, and disturbances in its composition are associated with several (age-related) neurological conditions, including depression, Alzheimer's disease, and Parkinson's disease. Several pathways have been evoked for gut microbiota-brain communication, including neural connections (vagus nerve), circulating mediators derived by host-bacteria cometabolism, as well as the influence exerted by gut microbiota on host gut function, metabolism, and immune system. Although the most provoking data emerged from animal studies and despite the huge debate around the possible epiphenomenal nature of those findings, the gut microbiota-brain axis still remains a fascinating target to be exploited to attenuate some of the most burdensome consequences of aging.
Collapse
Affiliation(s)
- Riccardo Calvani
- Department of Geriatrics, Neurosciences and Orthopedics, Agostino Gemelli University Polyclinic, Catholic University of the Sacred Heart, Rome, Italy
| | - Anna Picca
- Department of Geriatrics, Neurosciences and Orthopedics, Agostino Gemelli University Polyclinic, Catholic University of the Sacred Heart, Rome, Italy
| | - Maria Rita Lo Monaco
- Department of Geriatrics, Neurosciences and Orthopedics, Agostino Gemelli University Polyclinic, Catholic University of the Sacred Heart, Rome, Italy
| | - Francesco Landi
- Department of Geriatrics, Neurosciences and Orthopedics, Agostino Gemelli University Polyclinic, Catholic University of the Sacred Heart, Rome, Italy
| | - Roberto Bernabei
- Department of Geriatrics, Neurosciences and Orthopedics, Agostino Gemelli University Polyclinic, Catholic University of the Sacred Heart, Rome, Italy
| | - Emanuele Marzetti
- Department of Geriatrics, Neurosciences and Orthopedics, Agostino Gemelli University Polyclinic, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
610
|
Xiao L, Van't Land B, Engen PA, Naqib A, Green SJ, Nato A, Leusink-Muis T, Garssen J, Keshavarzian A, Stahl B, Folkerts G. Human milk oligosaccharides protect against the development of autoimmune diabetes in NOD-mice. Sci Rep 2018; 8:3829. [PMID: 29497108 PMCID: PMC5832804 DOI: 10.1038/s41598-018-22052-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 02/14/2018] [Indexed: 12/19/2022] Open
Abstract
Development of Type 1 diabetes (T1D) is influenced by non-genetic factors, such as optimal microbiome development during early life that "programs" the immune system. Exclusive and prolonged breastfeeding is an independent protective factor against the development of T1D, likely via bioactive components. Human Milk Oligosaccharides (HMOS) are microbiota modulators, known to regulate immune responses directly. Here we show that early life provision (only for a period of six weeks) of 1% authentic HMOS (consisting of both long-chain, as well as short-chain structures), delayed and suppressed T1D development in non-obese diabetic mice and reduced development of severe pancreatic insulitis in later life. These protective effects were associated with i) beneficial alterations in fecal microbiota composition, ii) anti-inflammatory microbiota-generating metabolite (i.e. short chain fatty acids (SCFAs)) changes in fecal, as well as cecum content, and iii) induction of anti-diabetogenic cytokine profiles. Moreover, in vitro HMOS combined with SCFAs induced development of tolerogenic dendritic cells (tDCs), priming of functional regulatory T cells, which support the protective effects detected in vivo. In conclusion, HMOS present in human milk are therefore thought to be vital in the protection of children at risk for T1D, supporting immune and gut microbiota development in early life.
Collapse
Affiliation(s)
- Ling Xiao
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, The Netherlands
| | - Belinda Van't Land
- Nutricia Research, Department of Immunology/Human milk research platform, Utrecht, The Netherlands.
- University Medical Center Utrecht, The Wilhelmina Children's Hospital, Laboratory of Translational Immunology, Utrecht, The Netherlands.
| | - Phillip A Engen
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, USA
| | - Ankur Naqib
- DNA Services Facility, Research Resources Center, Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Stefan J Green
- DNA Services Facility, Research Resources Center, Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Angie Nato
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, The Netherlands
| | - Thea Leusink-Muis
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, The Netherlands
| | - Johan Garssen
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, The Netherlands
- Nutricia Research, Department of Immunology/Human milk research platform, Utrecht, The Netherlands
| | - Ali Keshavarzian
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, The Netherlands
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, USA
- Department of Pharmacology, Department of Physiology, Rush University Medical Center, Chicago, IL, USA
| | - Bernd Stahl
- Nutricia Research, Department of Immunology/Human milk research platform, Utrecht, The Netherlands
| | - Gert Folkerts
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, The Netherlands
| |
Collapse
|
611
|
A Walnut-Enriched Diet Affects Gut Microbiome in Healthy Caucasian Subjects: A Randomized, Controlled Trial. Nutrients 2018; 10:nu10020244. [PMID: 29470389 PMCID: PMC5852820 DOI: 10.3390/nu10020244] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/19/2018] [Accepted: 02/20/2018] [Indexed: 02/06/2023] Open
Abstract
Regular walnut consumption is associated with better health. We have previously shown that eight weeks of walnut consumption (43 g/day) significantly improves lipids in healthy subjects. In the same study, gut microbiome was evaluated. We included 194 healthy subjects (134 females, 63 ± 7 years, BMI 25.1 ± 4.0 kg/m2) in a randomized, controlled, prospective, cross-over study. Following a nut-free run-in period, subjects were randomized to two diet phases (eight weeks each); 96 subjects first followed a walnut-enriched diet (43 g/day) and then switched to a nut-free diet, while 98 subjects followed the diets in reverse order. While consuming the walnut-enriched diet, subjects were advised to either reduce fat or carbohydrates or both to account for the additional calories. Fecal samples were collected from 135 subjects at the end of the walnut-diet and the control-diet period for microbiome analyses. The 16S rRNA gene sequencing data was clustered with a 97% similarity into Operational Taxonomic Units (OTUs). UniFrac distances were used to determine diversity between groups. Differential abundance was evaluated using the Kruskal–Wallis rank sum test. All analyses were performed using Rhea. Generalized UniFrac distance shows that walnut consumption significantly affects microbiome composition and diversity. Multidimensional scaling (metric and non-metric) indicates dissimilarities of approximately 5% between walnut and control (p = 0.02). The abundance of Ruminococcaceae and Bifidobacteria increased significantly (p < 0.02) while Clostridium sp. cluster XIVa species (Blautia; Anaerostipes) decreased significantly (p < 0.05) during walnut consumption. The effect of walnut consumption on the microbiome only marginally depended on whether subjects replaced fat, carbohydrates or both while on walnuts. Daily intake of 43 g walnuts over eight weeks significantly affects the gut microbiome by enhancing probiotic- and butyric acid-producing species in healthy individuals. Further evaluation is required to establish whether these changes are preserved during longer walnut consumption and how these are linked to the observed changes in lipid metabolism.
Collapse
|
612
|
Lamichhane S, Yde CC, Jensen HM, Morovic W, Hibberd AA, Ouwehand AC, Saarinen MT, Forssten SD, Wiebe L, Marcussen J, Bertelsen K, Meier S, Young JF, Bertram HC. Metabolic Fate of 13C-Labeled Polydextrose and Impact on the Gut Microbiome: A Triple-Phase Study in a Colon Simulator. J Proteome Res 2018; 17:1041-1053. [DOI: 10.1021/acs.jproteome.7b00683] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Santosh Lamichhane
- Department of Food Science, Aarhus University, Kirstinebjergvej 10, 5792 Aarslev and Blichers Allé 20, 8830 Tjele, Denmark
- Turku
Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku 20520, Finland
| | - Christian C. Yde
- Department of Food Science, Aarhus University, Kirstinebjergvej 10, 5792 Aarslev and Blichers Allé 20, 8830 Tjele, Denmark
- DuPont Nutrition Biosciences ApS, Edwin Rahrs Vej 38, 8220 Brabrand, Denmark
| | - Henrik Max Jensen
- DuPont Nutrition Biosciences ApS, Edwin Rahrs Vej 38, 8220 Brabrand, Denmark
| | - Wesley Morovic
- DuPont Nutrition and Health, 3329 Agriculture Drive, Madison, Wisconsin 53716, United States
| | - Ashley A. Hibberd
- DuPont Nutrition and Health, 3329 Agriculture Drive, Madison, Wisconsin 53716, United States
| | - Arthur C. Ouwehand
- Dupont Nutrition and Health, Sokeritehtaantie 20, 02460 Kantvik, Finland
| | - Markku T. Saarinen
- Dupont Nutrition and Health, Sokeritehtaantie 20, 02460 Kantvik, Finland
| | - Sofia D. Forssten
- Dupont Nutrition and Health, Sokeritehtaantie 20, 02460 Kantvik, Finland
| | - Lars Wiebe
- DuPont Nutrition Biosciences ApS, Tårnvej 25, 7200 Grindsted, Denmark
| | - Jørn Marcussen
- DuPont Nutrition Biosciences ApS, Edwin Rahrs Vej 38, 8220 Brabrand, Denmark
| | - Kresten Bertelsen
- DuPont Nutrition Biosciences ApS, Edwin Rahrs Vej 38, 8220 Brabrand, Denmark
- Vestas Wind Systems A/S, Hedeager
42, 8200 Aarhus
N, Denmark
| | - Sebastian Meier
- Department
of Chemistry, Technical University of Denmark, Kemitorvet, Building 207, 2800 Kgs. Lyngby, Denmark
| | - Jette F. Young
- Department of Food Science, Aarhus University, Kirstinebjergvej 10, 5792 Aarslev and Blichers Allé 20, 8830 Tjele, Denmark
| | - Hanne Christine Bertram
- Department of Food Science, Aarhus University, Kirstinebjergvej 10, 5792 Aarslev and Blichers Allé 20, 8830 Tjele, Denmark
| |
Collapse
|
613
|
How to isolate, identify and determine antimicrobial susceptibility of anaerobic bacteria in routine laboratories. Clin Microbiol Infect 2018; 24:1139-1148. [PMID: 29458156 DOI: 10.1016/j.cmi.2018.02.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/09/2018] [Accepted: 02/11/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND There has been increased interest in the study of anaerobic bacteria that cause human infection during the past decade. Many new genera and species have been described using 16S rRNA gene sequencing of clinical isolates obtained from different infection sites with commercially available special culture media to support the growth of anaerobes. Several systems, such as anaerobic pouches, boxes, jars and chambers provide suitable anaerobic culture conditions to isolate even strict anaerobic bacteria successfully from clinical specimens. Beside the classical, time-consuming identification methods and automated biochemical tests, the use of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry has revolutionized identification of even unusual and slow-growing anaerobes directly from culture plates, providing the possibility of providing timely information about anaerobic infections. AIMS The aim of this review article is to present methods for routine laboratories, which carry out anaerobic diagnostics on different levels. SOURCES Relevant data from the literature mostly published during the last 7 years are encompassed and discussed. CONTENT The review involves topics on the anaerobes that are members of the commensal microbiota and their role causing infection, the key requirements for collection and transport of specimens, processing of specimens in the laboratory, incubation techniques, identification and antimicrobial susceptibility testing of anaerobic bacteria. Advantages, drawbacks and specific benefits of the methods are highlighted. IMPLICATIONS The present review aims to update and improve anaerobic microbiology in laboratories with optimal conditions as well as encourage its routine implementation in laboratories with restricted resources.
Collapse
|
614
|
Bishehsari F, Engen PA, Preite NZ, Tuncil YE, Naqib A, Shaikh M, Rossi M, Wilber S, Green SJ, Hamaker BR, Khazaie K, Voigt RM, Forsyth CB, Keshavarzian A. Dietary Fiber Treatment Corrects the Composition of Gut Microbiota, Promotes SCFA Production, and Suppresses Colon Carcinogenesis. Genes (Basel) 2018; 9:genes9020102. [PMID: 29462896 PMCID: PMC5852598 DOI: 10.3390/genes9020102] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/29/2018] [Accepted: 02/13/2018] [Indexed: 12/26/2022] Open
Abstract
Epidemiological studies propose a protective role for dietary fiber in colon cancer (CRC). One possible mechanism of fiber is its fermentation property in the gut and ability to change microbiota composition and function. Here, we investigate the role of a dietary fiber mixture in polyposis and elucidate potential mechanisms using TS4Cre × cAPCl°x468 mice. Stool microbiota profiling was performed, while functional prediction was done using PICRUSt. Stool short-chain fatty acid (SCFA) metabolites were measured. Histone acetylation and expression of SCFA butyrate receptor were assessed. We found that SCFA-producing bacteria were lower in the polyposis mice, suggesting a decline in the fermentation product of dietary fibers with polyposis. Next, a high fiber diet was given to polyposis mice, which significantly increased SCFA-producing bacteria as well as SCFA levels. This was associated with an increase in SCFA butyrate receptor and a significant decrease in polyposis. In conclusion, we found polyposis to be associated with dysbiotic microbiota characterized by a decline in SCFA-producing bacteria, which was targetable by high fiber treatment, leading to an increase in SCFA levels and amelioration of polyposis. The prebiotic activity of fiber, promoting beneficial bacteria, could be the key mechanism for the protective effects of fiber on colon carcinogenesis. SCFA-promoting fermentable fibers are a promising dietary intervention to prevent CRC.
Collapse
Affiliation(s)
- Faraz Bishehsari
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL USA.
| | - Phillip A Engen
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL USA.
| | - Nailliw Z Preite
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL USA.
| | - Yunus E Tuncil
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN USA.
| | - Ankur Naqib
- DNA Services Facility, Research Resources Center, University of Illinois at Chicago, Chicago, IL USA.
| | - Maliha Shaikh
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL USA.
| | - Marco Rossi
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL USA.
| | - Sherry Wilber
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL USA.
| | - Stefan J Green
- DNA Services Facility, Research Resources Center, University of Illinois at Chicago, Chicago, IL USA.
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL USA.
| | - Bruce R Hamaker
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN USA.
| | - Khashayarsha Khazaie
- Department of Immunology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | - Robin M Voigt
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL USA.
| | - Christopher B Forsyth
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL USA.
| | - Ali Keshavarzian
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL USA.
- Department of Physiology, Rush University Medical Center, Chicago, IL USA.
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht Netherlands.
- Department of Pharmacology, Rush University Medical Center, Chicago, IL USA.
| |
Collapse
|
615
|
Chauhan NS, Pandey R, Mondal AK, Gupta S, Verma MK, Jain S, Ahmed V, Patil R, Agarwal D, Girase B, Shrivastava A, Mobeen F, Sharma V, Srivastava TP, Juvekar SK, Prasher B, Mukerji M, Dash D. Western Indian Rural Gut Microbial Diversity in Extreme Prakriti Endo-Phenotypes Reveals Signature Microbes. Front Microbiol 2018; 9:118. [PMID: 29487572 PMCID: PMC5816807 DOI: 10.3389/fmicb.2018.00118] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 01/18/2018] [Indexed: 12/12/2022] Open
Abstract
Heterogeneity amidst healthy individuals at genomic level is being widely acknowledged. This, in turn, is modulated by differential response to environmental cues and treatment regimens, necessitating the need for stratified/personalized therapy. We intend to understand the molecular determinants of Ayurvedic way (ancient Indian system of medicine) of endo-phenotyping individuals into distinct constitution types termed “Prakriti,” which forms the basis of personalized treatment. In this study, we explored and analyzed the healthy human gut microbiome structure within three predominant Prakriti groups from a genetically homogenous cohort to discover differentially abundant taxa, using 16S rRNA gene based microbial community profiling. We found Bacteroidetes and Firmicutes as major gut microbial components in varying composition, albeit with similar trend across Prakriti. Multiple species of the core microbiome showed differential abundance within Prakriti types, with gender specific signature taxons. Our study reveals that despite overall uniform composition of gut microbial community, healthy individuals belonging to different Prakriti groups have enrichment of specific bacteria. It highlights the importance of Prakriti based endo-phenotypes to explain the variability amongst healthy individuals in gut microbial flora that have important consequences for an individual's health, disease and treatment.
Collapse
Affiliation(s)
- Nar S Chauhan
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, India
| | - Rajesh Pandey
- CSIR Ayurgenomics Unit - TRISUTRA (Translational Research and Innovative Science ThRough Ayurgenomics), CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Anupam K Mondal
- G.N. Ramachandran Knowledge Centre for Genome Informatics, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India.,Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (IGIB), New Delhi, India
| | - Shashank Gupta
- CSIR Ayurgenomics Unit - TRISUTRA (Translational Research and Innovative Science ThRough Ayurgenomics), CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Manoj K Verma
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, India
| | - Sweta Jain
- CSIR Ayurgenomics Unit - TRISUTRA (Translational Research and Innovative Science ThRough Ayurgenomics), CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Vasim Ahmed
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, India
| | - Rutuja Patil
- Vadu Rural Health Program, KEM Hospital Research Centre, Pune, India
| | - Dhiraj Agarwal
- Vadu Rural Health Program, KEM Hospital Research Centre, Pune, India
| | - Bhushan Girase
- Vadu Rural Health Program, KEM Hospital Research Centre, Pune, India
| | | | - Fauzul Mobeen
- School of Basic Sciences, Indian Institute of Technology, Mandi, India
| | - Vikas Sharma
- School of Basic Sciences, Indian Institute of Technology, Mandi, India
| | | | - Sanjay K Juvekar
- Vadu Rural Health Program, KEM Hospital Research Centre, Pune, India
| | - Bhavana Prasher
- CSIR Ayurgenomics Unit - TRISUTRA (Translational Research and Innovative Science ThRough Ayurgenomics), CSIR-Institute of Genomics and Integrative Biology, New Delhi, India.,Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (IGIB), New Delhi, India.,Genomics and Molecular Medicine and CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Mitali Mukerji
- CSIR Ayurgenomics Unit - TRISUTRA (Translational Research and Innovative Science ThRough Ayurgenomics), CSIR-Institute of Genomics and Integrative Biology, New Delhi, India.,Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (IGIB), New Delhi, India.,Genomics and Molecular Medicine and CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Debasis Dash
- CSIR Ayurgenomics Unit - TRISUTRA (Translational Research and Innovative Science ThRough Ayurgenomics), CSIR-Institute of Genomics and Integrative Biology, New Delhi, India.,G.N. Ramachandran Knowledge Centre for Genome Informatics, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India.,Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (IGIB), New Delhi, India
| |
Collapse
|
616
|
Van den Abbeele P, Taminiau B, Pinheiro I, Duysburgh C, Jacobs H, Pijls L, Marzorati M. Arabinoxylo-Oligosaccharides and Inulin Impact Inter-Individual Variation on Microbial Metabolism and Composition, Which Immunomodulates Human Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:1121-1130. [PMID: 29363966 DOI: 10.1021/acs.jafc.7b04611] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Fecal batch fermentations coupled to cocultures of epithelial cells and macrophages were used to compare how arabinoxylo-oligosaccharides (AXOS) and inulin modulate gut microbial activity and composition of three different human donors and subsequently the epithelial permeability and immune response. Both inulin and AXOS decreased the pH during incubation (-1.5 pH units), leading to increased productions of acetate, propionate, and butyrate. Differences in terms of metabolites production could be linked to specific microbial alterations at genus level upon inulin/AXOS supplementation (i.e., Bifidobacterium, Bacteroides, Prevotella and unclassified Erysipelotrichaceae), as shown by 16S-targeted Illumina sequencing. Both products stimulated gut barrier and immune function with increases in TEER, NF-KB, IL-10, and IL-6. Ingredients with different structures selectively modulate the microbiota of a specific donor leading to differential changes at metabolic level. The extent of this effect is donor specific and is linked to a final specific modulation of the host's immune system.
Collapse
Affiliation(s)
| | - Bernard Taminiau
- Department of Food Science, University of Liège (ULG) , Quartier Vallée 2, Avenue de Cureghem 10, 4000 Liège, Belgium
| | - Iris Pinheiro
- ProDigest bvba , Technologiepark 3, 9052 Ghent, Belgium
| | | | - Heidi Jacobs
- Cosucra-Groupe Warcoing S.A. , Rue de la Sucrerie 1, 7740 Pecq, Belgium
| | - Loek Pijls
- Cosucra-Groupe Warcoing S.A. , Rue de la Sucrerie 1, 7740 Pecq, Belgium
| | - Massimo Marzorati
- Center of Microbial Ecology and Technology (CMET), Ghent University , Coupure Links 653, 9000 Ghent, Belgium
| |
Collapse
|
617
|
Rajpoot M, Sharma AK, Sharma A, Gupta GK. Understanding the microbiome: Emerging biomarkers for exploiting the microbiota for personalized medicine against cancer. Semin Cancer Biol 2018; 52:1-8. [PMID: 29425888 DOI: 10.1016/j.semcancer.2018.02.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 11/03/2017] [Accepted: 02/05/2018] [Indexed: 02/08/2023]
Abstract
The human body is a home to more than 1 trillion microbes with a diverse variety of commensal microbes that play a crucial role towards the health of the individual. These microbes occupy different habitats such as gut, skin, vagina, oral etc. Not only the types and abundance of microbes are different in different organs, but also these may differ in different individuals. The genome of these microbiota and their ecosystem constitute to form a microbiome. Factors such as diet, environment, host genetics etc. may be the reason behind the wide microbial diversity. A number of studies performed on human microbiome have revealed that microbiota present in healthy and diseased individuals are distinct. Altered microbiome is many a times the reason behind the overexpression of genes which may cause complex diseases including cancer. Manipulation of the human microbiome can be done by microbial supplements such as probiotics or synbiotics, diet or prebiotics and microbial suppression strategies using antibiotics. Recent advances in genome sequencing technologies and metagenomic analysis provide us the broader understanding of these commensal microbes and highlighting the distinctive features of microbiome during healthy and disease states. Molecular pathological epidemiology (MPE) studies have been very helpful in providing insights into the pathological process behind disease evolution and progression by determining the specific etiological factors. New emerging field of research targets the microbiome for therapeutic purposes by which personalized medicines can be made for treating various types of tumors. Screening programmes might be helpful in identifying patients who are at the verge of developing cancer and in delivering appropriate approaches according to individual risk modes so that disease could be prevented.
Collapse
Affiliation(s)
- Meenakshi Rajpoot
- Department of Biotechnology, Maharishi Markandeshwar University, Mullana, Ambala, Haryana, 133207, India
| | - Anil K Sharma
- Department of Biotechnology, Maharishi Markandeshwar University, Mullana, Ambala, Haryana, 133207, India.
| | - Anil Sharma
- Department of Biochemistry and Molecular Biology, Mayo School of Medicine, Rochester, MN, 55905, USA
| | - Girish Kumar Gupta
- Department of Pharmaceutical Chemistry, M. M. College of Pharmacy, Maharishi Markandeshwar University, Mullana, Ambala, Haryana, 133207, India
| |
Collapse
|
618
|
Parajuli A, Grönroos M, Siter N, Puhakka R, Vari HK, Roslund MI, Jumpponen A, Nurminen N, Laitinen OH, Hyöty H, Rajaniemi J, Sinkkonen A. Urbanization Reduces Transfer of Diverse Environmental Microbiota Indoors. Front Microbiol 2018; 9:84. [PMID: 29467728 PMCID: PMC5808279 DOI: 10.3389/fmicb.2018.00084] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 01/12/2018] [Indexed: 12/17/2022] Open
Abstract
Expanding urbanization is a major factor behind rapidly declining biodiversity. It has been proposed that in urbanized societies, the rarity of contact with diverse environmental microbiota negatively impacts immune function and ultimately increases the risk for allergies and other immune-mediated disorders. Surprisingly, the basic assumption that urbanization reduces exposure to environmental microbiota and its transfer indoors has rarely been examined. We investigated if the land use type around Finnish homes affects the diversity, richness, and abundance of bacterial communities indoors. Debris deposited on standardized doormats was collected in 30 rural and 26 urban households in and near the city of Lahti, Finland, in August 2015. Debris was weighed, bacterial community composition determined by high throughput sequencing of bacterial 16S ribosomal RNA (rRNA) gene on the Illumina MiSeq platform, and the percentage of four different land use types (i.e., built area, forest, transitional, and open area) within 200 m and 2000 m radiuses from each household was characterized. The quantity of doormat debris was inversely correlated with coverage of built area. The diversity of total bacterial, Proteobacterial, Actinobacterial, Bacteroidetes, and Firmicutes communities decreased as the percentage of built area increased. Their richness followed the same pattern except for Firmicutes for which no association was observed. The relative abundance of Proteobacteria and particularly Gammaproteobacteria increased, whereas that of Actinobacteria decreased with increasing built area. Neither Phylum Firmicutes nor Bacteroidetes varied with coverage of built area. Additionally, the relative abundance of potentially pathogenic bacterial families and genera increased as the percentage of built area increased. Interestingly, having domestic animals (including pets) only altered the association between the richness of Gammaproteobacteria and diversity of Firmicutes with the built area coverage suggesting that animal ownership minimally affects transfer of environmental microbiota indoors from the living environment. These results support the hypothesis that people living in densely built areas are less exposed to diverse environmental microbiota than people living in more sparsely built areas.
Collapse
Affiliation(s)
- Anirudra Parajuli
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Lahti, Finland
| | - Mira Grönroos
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Lahti, Finland
| | - Nathan Siter
- School of Artitechture, Tampere University of Technology, Tampere, Finland
| | - Riikka Puhakka
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Lahti, Finland
| | - Heli K. Vari
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Lahti, Finland
| | - Marja I. Roslund
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Lahti, Finland
| | - Ari Jumpponen
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Noora Nurminen
- Department of Virology, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Olli H. Laitinen
- Department of Virology, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Heikki Hyöty
- Department of Virology, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- Fimlab Laboratories, Pirkanmaa Hospital District, Tampere, Finland
| | - Juho Rajaniemi
- School of Artitechture, Tampere University of Technology, Tampere, Finland
| | - Aki Sinkkonen
- Ecosystems and Environment Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Lahti, Finland
| |
Collapse
|
619
|
Sharma S, Sinha VR. Current pharmaceutical strategies for efficient site specific delivery in inflamed distal intestinal mucosa. J Control Release 2018; 272:97-106. [DOI: 10.1016/j.jconrel.2018.01.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/03/2018] [Accepted: 01/05/2018] [Indexed: 02/07/2023]
|
620
|
Qin Y, Wade PA. Crosstalk between the microbiome and epigenome: messages from bugs. J Biochem 2018; 163:105-112. [PMID: 29161429 PMCID: PMC5892391 DOI: 10.1093/jb/mvx080] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 10/31/2017] [Indexed: 01/07/2023] Open
Abstract
Mammals exist in a complicated symbiotic relationship with their gut microbiome, which is postulated to have broad impacts on host health and disease. As omics-based technologies have matured, the potential mechanisms by which the microbiome affects host physiology are being addressed. The gut microbiome, which provides environmental cues, can modify host cell responses to stimuli through alterations in the host epigenome and, ultimately, gene expression. Increasing evidence highlights microbial generation of bioactive compounds that impact the transcriptional machinery in host cells. Here, we review current understanding of the crosstalk between gut microbiota and the host epigenome, including DNA methylation, histone modification and non-coding RNAs. These studies are providing insights into how the host responds to microbial signalling and are predicted to provide information for the application of precision medicine.
Collapse
Affiliation(s)
- Yufeng Qin
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Paul A Wade
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| |
Collapse
|
621
|
Reinoso Webb C, den Bakker H, Koboziev I, Jones-Hall Y, Rao Kottapalli K, Ostanin D, Furr KL, Mu Q, Luo XM, Grisham MB. Differential Susceptibility to T Cell-Induced Colitis in Mice: Role of the Intestinal Microbiota. Inflamm Bowel Dis 2018; 24:361-379. [PMID: 29361089 PMCID: PMC6176899 DOI: 10.1093/ibd/izx014] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Indexed: 12/12/2022]
Abstract
One of the best characterized mouse models of the inflammatory bowel diseases (IBD; Crohn's disease, ulcerative colitis) is the CD4+CD45RBhigh T cell transfer model of chronic colitis. Following our relocation to Texas Tech University Health Sciences Center (TTUHSC), we observed a dramatic reduction in the incidence of moderate-to-severe colitis from a 16-year historical average of 90% at Louisiana State University Health Sciences Center (LSUHSC) to <30% at TTUHSC. We hypothesized that differences in the commensal microbiota at the 2 institutions may account for the differences in susceptibility to T cell-induced colitis. Using bioinformatic analyses of 16S rRNA amplicon sequence data, we quantified and compared the major microbial populations in feces from healthy and colitic mice housed at the 2 institutions. We found that the bacterial composition differed greatly between mice housed at LSUHSC vs TTUHSC. We identified several genera strongly associated with, and signficantly overrepresented in high responding RAG-/- mice housed at LSUHSC. In addition, we found that colonization of healthy TTUHSC RAG-/- mice with feces obtained from healthy or colitic RAG-/- mice housed at LSUHSC transferred susceptibility to T cell-induced colitis such that the recipients developed chronic colitis with incidence and severity similar to mice generated at LSUHSC. Finally, we found that the treatment of mice with preexisting colitis with antibiotics remarkably attenuated disease. Taken together, our data demonstrate that specific microbial communities determine disease susceptibility and that manipulation of the intestinal microbiota alters the induction and/or perpetuation of chronic colitis.
Collapse
Affiliation(s)
- Cynthia Reinoso Webb
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX
| | | | - Iurii Koboziev
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Yava Jones-Hall
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, West Lafayette, IN
| | | | - Dmitry Ostanin
- Immunology Discovery, Translational Research and Development, Bristol Myers Squibb, Princeton, NJ
| | - Kathryn L Furr
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Qinghui Mu
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA
| | - Xin M Luo
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, VA
| | - Matthew B Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX,Correspondence address. Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, Texas 79430-6591. E-mail:
| |
Collapse
|
622
|
Ferreira RM, Pereira-Marques J, Pinto-Ribeiro I, Costa JL, Carneiro F, Machado JC, Figueiredo C. Gastric microbial community profiling reveals a dysbiotic cancer-associated microbiota. Gut 2018; 67:226-236. [PMID: 29102920 PMCID: PMC5868293 DOI: 10.1136/gutjnl-2017-314205] [Citation(s) in RCA: 441] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 10/25/2017] [Accepted: 10/26/2017] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Gastric carcinoma development is triggered by Helicobacter pylori. Chronic H. pylori infection leads to reduced acid secretion, which may allow the growth of a different gastric bacterial community. This change in the microbiome may increase aggression to the gastric mucosa and contribute to malignancy. Our aim was to evaluate the composition of the gastric microbiota in chronic gastritis and in gastric carcinoma. DESIGN The gastric microbiota was retrospectively investigated in 54 patients with gastric carcinoma and 81 patients with chronic gastritis by 16S rRNA gene profiling, using next-generation sequencing. Differences in microbial composition of the two patient groups were assessed using linear discriminant analysis effect size. Associations between the most relevant taxa and clinical diagnosis were validated by real-time quantitative PCR. Predictive functional profiling of microbial communities was obtained with PICRUSt. RESULTS The gastric carcinoma microbiota was characterised by reduced microbial diversity, by decreased abundance of Helicobacter and by the enrichment of other bacterial genera, mostly represented by intestinal commensals. The combination of these taxa into a microbial dysbiosis index revealed that dysbiosis has excellent capacity to discriminate between gastritis and gastric carcinoma. Analysis of the functional features of the microbiota was compatible with the presence of a nitrosating microbial community in carcinoma. The major observations were confirmed in validation cohorts from different geographic origins. CONCLUSIONS Detailed analysis of the gastric microbiota revealed for the first time that patients with gastric carcinoma exhibit a dysbiotic microbial community with genotoxic potential, which is distinct from that of patients with chronic gastritis.
Collapse
Affiliation(s)
- Rui M Ferreira
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Ipatimup – Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Joana Pereira-Marques
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Ipatimup – Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal,Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Ines Pinto-Ribeiro
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Ipatimup – Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal,Faculty of Medicine, University of Porto, Porto, Portugal
| | - Jose L Costa
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Ipatimup – Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal,Faculty of Medicine, University of Porto, Porto, Portugal
| | - Fatima Carneiro
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Ipatimup – Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal,Faculty of Medicine, University of Porto, Porto, Portugal,Department of Pathology, Centro Hospitalar São João, Porto, Portugal
| | - Jose C Machado
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Ipatimup – Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal,Faculty of Medicine, University of Porto, Porto, Portugal
| | - Ceu Figueiredo
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal,Ipatimup – Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal,Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
623
|
Kuo SM. Does Modification of the Large Intestinal Microbiome Contribute to the Anti-Inflammatory Activity of Fermentable Fiber? Curr Dev Nutr 2018; 2:nzx004. [PMID: 30377676 PMCID: PMC6201682 DOI: 10.3945/cdn.117.001180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 11/21/2017] [Indexed: 12/16/2022] Open
Abstract
Fiber is an inadequately understood and insufficiently consumed nutrient. This review examines the possible causal relation between fiber-induced microbiome changes and the anti-inflammatory activity of fiber. To demonstrate the dominant role of fermentable plant fiber in shaping the intestinal microbiome, animal and human fiber-feeding studies are reviewed. Using culture-, PCR-, and sequencing-based microbial analyses, a higher prevalence of Bifidobacterium and Lactobacillus genera was observed from the feeding of different types of fermentable fiber. This finding was reported in studies performed on several host species including human. Health conditions and medications that are linked to intestinal microbial alterations likely also change the nutrient environment of the large intestine. The unique gene clusters of Bifidobacterium and Lactobacillus that enable the catabolism of plant glycans and the ability of Bifidobacterium and Lactobacillus to reduce the colonization of proteobacteria probably contribute to their prevalence in a fiber-rich intestinal environment. The fiber-induced microbiome changes could contribute to the anti-inflammatory activity of fiber. Although most studies did not measure fecal microbial density or total daily fecal microbial output (colon microbial load), limited evidence suggests that the increase in intestinal commensal microbial load plays an important role in the anti-inflammatory activity of fiber. Various probiotic supplements, including Bifidobacterium and Lactobacillus, showed anti-inflammatory activity only in the presence of fiber, which promoted microbial growth as indicated by increasing plasma short-chain fatty acids. Probiotics alone or pure fiber administered under sterile conditions showed no anti-inflammatory activity. The potential mechanisms that could mediate the anti-inflammatory effect of common microbial metabolites are reviewed, but more in vivo trials are needed. Future studies including simultaneous microbial composition and load measurements are also important.
Collapse
Affiliation(s)
- Shiu-Ming Kuo
- Department of Exercise and Nutrition Sciences, University at Buffalo, SUNY, Buffalo, NY
| |
Collapse
|
624
|
Gut Microbiota as a Driver of Inflammation in Nonalcoholic Fatty Liver Disease. Mediators Inflamm 2018; 2018:9321643. [PMID: 29563854 PMCID: PMC5833468 DOI: 10.1155/2018/9321643] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 07/12/2017] [Accepted: 07/26/2017] [Indexed: 02/06/2023] Open
Abstract
The prevalence of nonalcoholic fatty liver disease and the consequent burden of metabolic syndrome have increased in recent years. Although the pathogenesis of nonalcoholic fatty liver disease is not completely understood, it is thought to be the hepatic manifestation of the dysregulation of insulin-dependent pathways leading to insulin resistance and adipose tissue accumulation in the liver. Recently, the gut-liver axis has been proposed as a key player in the pathogenesis of NAFLD, as the passage of bacteria-derived products into the portal circulation could lead to a trigger of innate immunity, which in turn leads to liver inflammation. Additionally, higher prevalence of intestinal dysbiosis, larger production of endogenous ethanol, and higher prevalence of increased intestinal permeability and bacterial translocation were found in patients with liver injury. In this review, we describe the role of intestinal dysbiosis in the activation of the inflammatory cascade in NAFLD.
Collapse
|
625
|
D'hoe K, Conterno L, Fava F, Falony G, Vieira-Silva S, Vermeiren J, Tuohy K, Raes J. Prebiotic Wheat Bran Fractions Induce Specific Microbiota Changes. Front Microbiol 2018; 9:31. [PMID: 29416529 PMCID: PMC5787670 DOI: 10.3389/fmicb.2018.00031] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/09/2018] [Indexed: 01/08/2023] Open
Abstract
Wheat bran fibers are considered beneficial to human health through their impact on gut microbiota composition and activity. Here, we assessed the prebiotic potential of selected bran fractions by performing a series of fecal slurry anaerobic fermentation experiments using aleurone as well as total, ultrafine, and soluble wheat bran (swb) as carbon sources. By combining amplicon-based community profiling with a fluorescent in situ hybridization (FISH) approach, we found that incubation conditions favor the growth of Proteobacteria such as Escherichia and Bilophila. These effects were countered in all but one [total wheat bran (twb)] fermentation experiments. Growth of Bifidobacterium species was stimulated after fermentation using ultrafine, soluble, and twb, in the latter two as part of a general increase in bacterial load. Both ultrafine and swb fermentation resulted in a trade-off between Bifidobacterium and Bilophila, as previously observed in human dietary supplementation studies looking at the effect of inulin-type fructans on the human gut microbiota. Aleurone selectively stimulated growth of Dorea and butyrate-producing Roseburia. All fermentation experiments induced enhanced gas production; increased butyrate concentrations were only observed following soluble bran incubation. Our results open perspectives for the development of aleurone as a complementary prebiotic selectively targeting colon butyrate producers.
Collapse
Affiliation(s)
- Kevin D'hoe
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium.,Jeroen Raes Lab, VIB KU Leuven Center for Microbiology, Leuven, Belgium.,Research Group of Microbiology, Department of Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lorenza Conterno
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, Trento, Italy.,Fermentation and Distillation, Laimburg Research Centre, Bolzano, Italy
| | - Francesca Fava
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, Trento, Italy
| | - Gwen Falony
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium.,Jeroen Raes Lab, VIB KU Leuven Center for Microbiology, Leuven, Belgium
| | - Sara Vieira-Silva
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium.,Jeroen Raes Lab, VIB KU Leuven Center for Microbiology, Leuven, Belgium
| | | | - Kieran Tuohy
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, Trento, Italy
| | - Jeroen Raes
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium.,Jeroen Raes Lab, VIB KU Leuven Center for Microbiology, Leuven, Belgium.,Research Group of Microbiology, Department of Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
626
|
Wang J, Qin C, He T, Qiu K, Sun W, Zhang X, Jiao N, Zhu W, Yin J. Alfalfa-containing diets alter luminal microbiota structure and short chain fatty acid sensing in the caecal mucosa of pigs. J Anim Sci Biotechnol 2018; 9:11. [PMID: 29372054 PMCID: PMC5769528 DOI: 10.1186/s40104-017-0216-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 12/13/2017] [Indexed: 01/08/2023] Open
Abstract
Background Pork produced by outdoor-reared pigs raised mostly on alfalfa pastures attracts increasing population of consumer from most of the world. In China, pigs were raised with alfalfa-containing diets to seek for good quality pork. However, the influence of dietary alfalfa involving high level of insoluble dietary fiber (IDF) on pig intestinal luminal microbiota composition remains unclear. The objective of this study was to investigate the effects of alfalfa on luminal microbiota and short chain fatty acids (SCFA) production, and gene expressions involved in SCFA sensing, transporting and absorbing in pig caecal mucosa. Results Twenty-four growing pigs were randomly allotted to four diets containing 0%, 5%, 10% and 15% alfalfa meal for a 28-d experiment. Ingestion of alfalfa meal-contained diets significantly increased the ratio of body weight gain to feed consumption. Illumina MiSeq sequencing of the V3 region of the 16S rRNA genes showed that alfalfa-containing diet significantly decreased the relative abundance of genera Turicibacter, Acidiphilium, Paracoccus, Propionibacterium, Corynebacterium, Pseudomonas, Acinetobacter, and Staphylococcus, and increased the relative abundance of genera Lachnospira, Marvinbryantia, and Desulfovibrio in the caecal digesta. Butyrate concentration was significantly increased in the hindgut by the supplementation of alfalfa meal in diets. The mRNA gene expressions of FFAR3, SMCT1, MCT1, PYY, and GCG were significantly increased in the caecal mucosa of pigs fed alfalfa meal. Conclusions Our results suggested that alfalfa-containing diet has exerted significant impacts on caecal microbiota composition, butyrate concentration and significantly upregulated mRNA expression of host caecal mucosal genes involved in SCFA sensing and absorption as well as regulation of satiety. Electronic supplementary material The online version of this article (10.1186/s40104-017-0216-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jiawei Wang
- 1State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193 China.,2Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang, Nanjing, 210095 China.,3College of Biotechnology and Food Science, Hebei Normal University for Nationalities, Chengde, 067000 China
| | - Chunfu Qin
- 1State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193 China
| | - Ting He
- 1State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193 China
| | - Kai Qiu
- 1State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193 China
| | - Wenjuan Sun
- 1State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193 China
| | - Xin Zhang
- 1State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193 China
| | - Ning Jiao
- 1State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193 China
| | - Weiyun Zhu
- 2Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang, Nanjing, 210095 China
| | - Jingdong Yin
- 1State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193 China
| |
Collapse
|
627
|
Hugenholtz F, de Vos WM. Mouse models for human intestinal microbiota research: a critical evaluation. Cell Mol Life Sci 2018; 75:149-160. [PMID: 29124307 PMCID: PMC5752736 DOI: 10.1007/s00018-017-2693-8] [Citation(s) in RCA: 368] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 09/29/2017] [Indexed: 02/06/2023]
Abstract
Since the early days of the intestinal microbiota research, mouse models have been used frequently to study the interaction of microbes with their host. However, to translate the knowledge gained from mouse studies to a human situation, the major spatio-temporal similarities and differences between intestinal microbiota in mice and humans need to be considered. This is done here with specific attention for the comparative physiology of the intestinal tract, the effect of dietary patterns and differences in genetics. Detailed phylogenetic and metagenomic analysis showed that while many common genera are found in the human and murine intestine, these differ strongly in abundance and in total only 4% of the bacterial genes are found to share considerable identity. Moreover, a large variety of murine strains is available yet most of the microbiota research is performed in wild-type, inbred strains and their transgenic derivatives. It has become increasingly clear that the providers, rearing facilities and the genetic background of these mice have a significant impact on the microbial composition and this is illustrated with recent experimental data. This may affect the reproducibility of mouse microbiota studies and their conclusions. Hence, future studies should take these into account to truly show the effect of diet, genotype or environmental factors on the microbial composition.
Collapse
Affiliation(s)
- Floor Hugenholtz
- Laboratory of Microbiology, Wageningen University, Stippeneng 4, Building 124, 6708 WE, Wageningen, The Netherlands
- Division of Infectious Diseases, Department of Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Willem M de Vos
- Laboratory of Microbiology, Wageningen University, Stippeneng 4, Building 124, 6708 WE, Wageningen, The Netherlands.
- Research Programme Unit Immunobiology, Department of Bacteriology and Immunology, Helsinki University, P.O. Box 21, 00014, Helsinki, Finland.
| |
Collapse
|
628
|
Alou M, Ndongo S, Frégère L, Labas N, Andrieu C, Richez M, Couderc C, Baudoin JP, Abrahão J, Brah S, Diallo A, Sokhna C, Cassir N, La Scola B, Cadoret F, Raoult D. Taxonogenomic description of four new Clostridium species isolated from human gut: 'Clostridium amazonitimonense', 'Clostridium merdae', 'Clostridium massilidielmoense' and 'Clostridium nigeriense'. New Microbes New Infect 2018; 21:128-139. [PMID: 29348922 PMCID: PMC5767839 DOI: 10.1016/j.nmni.2017.11.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/09/2017] [Accepted: 11/16/2017] [Indexed: 12/29/2022] Open
Abstract
Culturomics investigates microbial diversity of the human microbiome by combining diversified culture conditions, matrix-assisted laser desorption/ionization time-of-flight mass spectrometry and 16S rRNA gene identification. The present study allowed identification of four putative new Clostridium sensu stricto species: 'Clostridium amazonitimonense' strain LF2T, 'Clostridium massilidielmoense' strain MT26T, 'Clostridium nigeriense' strain Marseille-P2414T and 'Clostridium merdae' strain Marseille-P2953T, which we describe using the concept of taxonogenomics. We describe the main characteristics of each bacterium and present their complete genome sequence and annotation.
Collapse
Affiliation(s)
- M.T. Alou
- Aix-Marseille Université, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, INSERM 1095, Marseille, France
| | - S. Ndongo
- Aix-Marseille Université, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, INSERM 1095, Marseille, France
| | - L. Frégère
- Aix-Marseille Université, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, INSERM 1095, Marseille, France
| | - N. Labas
- Aix-Marseille Université, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, INSERM 1095, Marseille, France
| | - C. Andrieu
- Aix-Marseille Université, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, INSERM 1095, Marseille, France
| | - M. Richez
- Aix-Marseille Université, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, INSERM 1095, Marseille, France
| | - C. Couderc
- Aix-Marseille Université, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, INSERM 1095, Marseille, France
| | - J.-P. Baudoin
- Aix-Marseille Université, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, INSERM 1095, Marseille, France
| | - J. Abrahão
- Laboratório de Vírus, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - S. Brah
- Hopital National de Niamey, BP 247, Niamey, Niger
| | - A. Diallo
- Aix-Marseille Université, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, INSERM 1095, Marseille, France
- Campus Commun UCAD-IRD of Hann, Route des pères Maristes, Hann Maristes, BP 1386, CP 18524, Dakar, Senegal
| | - C. Sokhna
- Aix-Marseille Université, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, INSERM 1095, Marseille, France
- Campus Commun UCAD-IRD of Hann, Route des pères Maristes, Hann Maristes, BP 1386, CP 18524, Dakar, Senegal
| | - N. Cassir
- Aix-Marseille Université, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, INSERM 1095, Marseille, France
| | - B. La Scola
- Aix-Marseille Université, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, INSERM 1095, Marseille, France
| | - F. Cadoret
- Aix-Marseille Université, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, INSERM 1095, Marseille, France
| | - D. Raoult
- Aix-Marseille Université, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, INSERM 1095, Marseille, France
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
629
|
Li B, Evivie SE, Lu J, Jiao Y, Wang C, Li Z, Liu F, Huo G. Lactobacillus helveticus KLDS1.8701 alleviates d-galactose-induced aging by regulating Nrf-2 and gut microbiota in mice. Food Funct 2018; 9:6586-6598. [DOI: 10.1039/c8fo01768a] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We first revealed a close association between probiotic-manipulated gut microbiota and hepatic Nrf-2 dependent mechanisms to suppress d-galactose-induced aging.
Collapse
Affiliation(s)
- Bailiang Li
- Key Laboratory of Dairy Science
- Ministry of Education
- Northeast Agricultural University
- Harbin 150030
- China
| | - Smith Etareri Evivie
- Key Laboratory of Dairy Science
- Ministry of Education
- Northeast Agricultural University
- Harbin 150030
- China
| | - Jingjing Lu
- Key Laboratory of Dairy Science
- Ministry of Education
- Northeast Agricultural University
- Harbin 150030
- China
| | - Yuehua Jiao
- Center of Drug Safety Evaluation
- Heilongjiang University of Chinese Medicine
- Harbin 150040
- China
| | - Chengfeng Wang
- Key Laboratory of Dairy Science
- Ministry of Education
- Northeast Agricultural University
- Harbin 150030
- China
| | - Ziye Li
- Key Laboratory of Dairy Science
- Ministry of Education
- Northeast Agricultural University
- Harbin 150030
- China
| | - Fei Liu
- Key Laboratory of Dairy Science
- Ministry of Education
- Northeast Agricultural University
- Harbin 150030
- China
| | - Guicheng Huo
- Key Laboratory of Dairy Science
- Ministry of Education
- Northeast Agricultural University
- Harbin 150030
- China
| |
Collapse
|
630
|
Turroni F, Milani C, Duranti S, Ferrario C, Lugli GA, Mancabelli L, van Sinderen D, Ventura M. Bifidobacteria and the infant gut: an example of co-evolution and natural selection. Cell Mol Life Sci 2018; 75:103-118. [PMID: 28983638 PMCID: PMC11105234 DOI: 10.1007/s00018-017-2672-0] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 09/29/2017] [Indexed: 12/16/2022]
Abstract
Throughout the human life, the gut microbiota interacts with us in a number of different ways, thereby influencing our health status. The acquisition of such an interactive gut microbiota commences at birth. Medical and environmental factors including diet, antibiotic exposure and mode of delivery are major factors that shape the composition of the microbial communities in the infant gut. Among the most abundant members of the infant microbiota are species belonging to the Bifidobacterium genus, which are believed to confer beneficial effects upon their host. Bifidobacteria may be acquired directly from the mother by vertical transmission and their persistence in the infant gut is associated with their saccharolytic activity toward glycans that are abundant in the infant gut. Here, we discuss the establishment of the infant gut microbiota and the contribution of bifidobacteria to this early life microbial consortium.
Collapse
Affiliation(s)
- Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Sabrina Duranti
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Chiara Ferrario
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Leonardo Mancabelli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy.
- Microbiome Research Hub, University of Parma, Parma, Italy.
| |
Collapse
|
631
|
Santoro A, Ostan R, Candela M, Biagi E, Brigidi P, Capri M, Franceschi C. Gut microbiota changes in the extreme decades of human life: a focus on centenarians. Cell Mol Life Sci 2018; 75:129-148. [PMID: 29032502 PMCID: PMC5752746 DOI: 10.1007/s00018-017-2674-y] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 09/29/2017] [Indexed: 12/20/2022]
Abstract
The gut microbiota (GM) is a complex, evolutionarily molded ecological system, which contributes to a variety of physiological functions. The GM is highly dynamic, being sensitive to environmental stimuli, and its composition changes over the host's entire lifespan. However, the basic question of how much these changes may be ascribed to variables such as population, diet, genetics and gender, and/or to the aging process per se is still largely unanswered. We argue that comparison among studies on centenarians-the best model of healthy aging and longevity-recruited from different geographical areas/populations (different genetics and dietary habits) can help to disentangle the contribution of aging and non-aging-related variables to GM remodeling with age. The current review focuses on the role of population, gender and host genetics as possible drivers of GM modification along the human aging process. The feedback impact of age-associated GM variation on the GM-brain axis and GM metabolomics is also discussed. We likewise address the role of GM in neurodegenerative diseases such as Parkinson's and Alzheimer's, and its possible therapeutic use, taking advantage of the fact that centenarians are characterized by an extreme (healthy) phenotype versus patients suffering from age-related pathologies. Finally, it is argued that longitudinal studies combining metagenomics sequencing and in-depth phylogenetic analysis with a comprehensive phenotypic characterization of centenarians and patients using up-to-date omics (metabolomics, transcriptomics and meta-transcriptomics) are urgently needed.
Collapse
Affiliation(s)
- Aurelia Santoro
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum-University of Bologna, Via San Giacomo 12, 40126, Bologna, Italy.
- Interdepartmental Centre "L. Galvani" (CIG) Alma Mater Studiorum-University of Bologna, Via San Giacomo 12, 40126, Bologna, Italy.
| | - Rita Ostan
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum-University of Bologna, Via San Giacomo 12, 40126, Bologna, Italy
- Interdepartmental Centre "L. Galvani" (CIG) Alma Mater Studiorum-University of Bologna, Via San Giacomo 12, 40126, Bologna, Italy
| | - Marco Candela
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Elena Biagi
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Patrizia Brigidi
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Miriam Capri
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Alma Mater Studiorum-University of Bologna, Via San Giacomo 12, 40126, Bologna, Italy
- Interdepartmental Centre "L. Galvani" (CIG) Alma Mater Studiorum-University of Bologna, Via San Giacomo 12, 40126, Bologna, Italy
| | - Claudio Franceschi
- Institute of Neurological Sciences (IRCCS), Via Altura 3, 40139, Bologna, Italy
| |
Collapse
|
632
|
Rubio-Jurado B, Balderas-Peña LMA, García-Luna EE, Zavala-Cerna MG, Riebeling-Navarro C, Reyes PA, Nava-Zavala AH. Obesity, Thrombotic Risk, and Inflammation in Cancer. Adv Clin Chem 2018; 85:71-89. [PMID: 29655462 DOI: 10.1016/bs.acc.2018.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neoplasms exhibits a high incidence and mortality rates due to their complex and commonly overlapping clinical, biochemical, and morphologic profiles influenced by acquired or inherited molecular abnormalities, cell of origin, and level of differentiation. Obesity appears related to ~20% of cancers including endometrial, esophageal, colorectal, postmenopausal breast, prostate, and renal. Several factors other than obesity, i.e., insulin, insulin-like growth factor, sexual hormones, and adipokines may play a potential role in neoplasia. Cancer-associated hypercoagulable and thrombotic states are influenced by abnormalities in the vascular wall and susceptibility to invasion, interference in blood flow and increase in circulating tissue factor and thrombin, activation of cell growth factors, the presence of a central catheter, chemotherapies, neoplasm type, and surgery. In cancer, thromboembolic complications are the second most frequent cause of death with pulmonary thromboembolism in ~50% of cases postmortem. Thrombosis worsens prognosis as demonstrated with a survival rate as low as 12% per year vs 36% in nonthrombic patients. Deep vein thrombosis is the most frequent thromboembolic complication in cancer. It is usually detected at diagnosis and within the first 3 months of chemotherapy. The underlining mechanisms of this association should be further studied to identify patients at higher risk and develop adequate prevention, diagnostic, and treatment measures. The D-dimer test can be successfully used to assess the fibrinolytic phase of coagulation and as such is routinely used in suspected cases of deep vein thrombosis and pulmonary thromboembolism. In addition, significant advances have been made in understanding the composition and functional capabilities of the gut microbiota in the inflammatory process, obesity, and its roles in cancer; however, the intricate balance that exists within the microbiota may not only affect the host directly, it can also disrupt the entire microbial community. CONCLUSIONS Cancer is a prothrombotic and inflammatory state in which the activation of coagulation is related to tumor growth, angiogenesis, and metastasis. It is important to identify the relationship between body mass index with these processes and clarify their importance in cancer prognosis. Future research should answer the question if manipulation of resident microbial communities could potentially improve prognosis and treatment outcome.
Collapse
|
633
|
Jung JY, Ahn Y, Khare S, Gokulan K, Piñeiro SA, Cerniglia CE. An in vitro study to assess the impact of tetracycline on the human intestinal microbiome. Anaerobe 2017; 49:85-94. [PMID: 29294359 DOI: 10.1016/j.anaerobe.2017.12.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/22/2017] [Accepted: 12/27/2017] [Indexed: 01/24/2023]
Abstract
The human intestinal microbiome, a generally stable ecosystem, could be potentially altered by the ingestion of antimicrobial drug residues in foods derived from animals. Data and the scientific published literature on the effects of antimicrobial residues on the human intestinal microbiome are reviewed by national regulatory authorities as part of the human food safety evaluation of veterinary antimicrobial agents used in food-producing animals. In this study, we determined if tetracycline, at low residue concentrations, could impact the human intestinal microbiome structure and the resistance-gene profile, following acute and subchronic exposure. The effects of 0.15, 1.5, 15, and 150 μg/ml of tetracycline, after 24 h and 40 days of exposure, in 3% human fecal suspensions, collected from three individuals (A, B, and C) were investigated using in vitro batch cultures. Results were variable, with either no change or minor changes in total bacterial 16S rRNA gene copies after exposure of fecal samples to tetracycline, because of the inter-individual variation of human gastrointestinal tract microbiota. Bacterial community analysis using rRNA-based pyrosequencing revealed that Firmicutes and Bacteroidetes were the predominant phyla in the three fecal samples; the ratio of phylotypes varied among individuals. The evaluation of bacterial community changes at the genus level, from control to tetracycline-treated fecal samples, suggested that tetracycline under the conditions of this study could lead to slight differences in the composition of intestinal microbiota. The genus Bacteroides (of the Bacteroidetes) was consistently altered from 1.68 to 5.70% and 4.82-8.22% at tetracycline concentrations of 0.15 μg/ml or above at both time points for individual A, respectively, and increased 5.13-13.50% and 10.92-22.18% for individual B, respectively. Clostridium family XI increased 3.50-25.34% in the presence of tetracycline at 40 days for individual C. Principal Component Analysis (PCA) confirmed the pyrosequencing findings of inter-individual variability of the ratio of phylotypes and the effect of tetracycline. Among the 23 tetracycline resistance genes (TRGs) screened, four tet genes (tetO, Q, W, and X) were major TRGs in control and tetracycline-dosed fecal samples. A variable to slight increase of copy number of TRGs appeared to be related to tetracycline treatment, interindividual variability and duration of exposure. Despite, the inherent variability of the intestinal microbiota observed among or within individuals, this pilot study contributes to the knowledge base of the impact of low residue concentrations of tetracycline on the human intestinal microbiome on the potential for antimicrobial resistance.
Collapse
Affiliation(s)
- Ji Young Jung
- Division of Microbiology, National Center for Toxicological Research, U.S. FDA, Jefferson, AR, USA
| | - Youngbeom Ahn
- Division of Microbiology, National Center for Toxicological Research, U.S. FDA, Jefferson, AR, USA
| | - Sangeeta Khare
- Division of Microbiology, National Center for Toxicological Research, U.S. FDA, Jefferson, AR, USA
| | - Kuppan Gokulan
- Division of Microbiology, National Center for Toxicological Research, U.S. FDA, Jefferson, AR, USA
| | - Silvia A Piñeiro
- Division of Human Food Safety, Center for Veterinary Medicine, U.S. FDA, Rockville, MD, USA
| | - Carl E Cerniglia
- Division of Microbiology, National Center for Toxicological Research, U.S. FDA, Jefferson, AR, USA.
| |
Collapse
|
634
|
Hillman ET, Lu H, Yao T, Nakatsu CH. Microbial Ecology along the Gastrointestinal Tract. Microbes Environ 2017; 32:300-313. [PMID: 29129876 PMCID: PMC5745014 DOI: 10.1264/jsme2.me17017] [Citation(s) in RCA: 349] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 08/19/2017] [Indexed: 02/06/2023] Open
Abstract
The ecosystem of the human gastrointestinal (GI) tract traverses a number of environmental, chemical, and physical conditions because it runs from the oral cavity to the anus. These differences in conditions along with food or other ingested substrates affect the composition and density of the microbiota as well as their functional roles by selecting those that are the most suitable for that environment. Previous studies have mostly focused on Bacteria, with the number of studies conducted on Archaea, Eukarya, and Viruses being limited despite their important roles in this ecosystem. Furthermore, due to the challenges associated with collecting samples directly from the inside of humans, many studies are still exploratory, with a primary focus on the composition of microbiomes. Thus, mechanistic studies to investigate functions are conducted using animal models. However, differences in physiology and microbiomes need to be clarified in order to aid in the translation of animal model findings into the context of humans. This review will highlight Bacteria, Archaea, Fungi, and Viruses, discuss differences along the GI tract of healthy humans, and perform comparisons with three common animal models: rats, mice, and pigs.
Collapse
Affiliation(s)
- Ethan T. Hillman
- Department of Agricultural and Biological Engineering, Purdue UniversityWest Lafayette, Indiana 47907USA
| | - Hang Lu
- Department of Animal Science, Purdue UniversityWest Lafayette, Indiana 47907USA
| | - Tianming Yao
- Department of Food Science, Purdue UniversityWest Lafayette, Indiana 47907USA
| | - Cindy H. Nakatsu
- Department of Agronomy, Purdue UniversityWest Lafayette, Indiana 47907USA
| |
Collapse
|
635
|
Pinto F, Medina DA, Pérez-Correa JR, Garrido D. Modeling Metabolic Interactions in a Consortium of the Infant Gut Microbiome. Front Microbiol 2017; 8:2507. [PMID: 29312209 PMCID: PMC5735223 DOI: 10.3389/fmicb.2017.02507] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 12/01/2017] [Indexed: 12/21/2022] Open
Abstract
The gut microbiome is a complex microbial community that has a significant influence on the host. Microbial interactions in the gut are mediated by dietary substrates, especially complex polysaccharides. In this environment, breakdown products from larger carbohydrates and short chain fatty acids are commonly shared among gut microbes. Understanding the forces that guide microbiome development and composition is important to determine its role in health and in the intervention of the gut microbiome as a therapeutic tool. Recently, modeling approaches such as genome-scale models and time-series analyses have been useful to predict microbial interactions. In this study, a bottom-up approach was followed to develop a mathematical model based on microbial growth equations that incorporate metabolic sharing and inhibition. The model was developed using experimental in vitro data from a system comprising four microorganisms of the infant gut microbiome (Bifidobacterium longum subsp. infantis, Lactobacillus acidophilus, Escherichia coli, and Bacteroides vulgatus), one substrate (fructooligosaccharides, FOS), and evaluating two metabolic products (acetate and lactate). After parameter optimization, the model accurately predicted bacterial abundance in co-cultures from mono-culture data. In addition, a good correlation was observed between the experimental data with predicted FOS consumption and acid production. B. infantis and L. acidophilus were dominant under these conditions. Further model validation included cultures with the four-species in a bioreactor using FOS. The model was able to predict the predominance of the two aforementioned species, as well as depletion of acetate and lactate. Finally, the model was tested for parameter identifiability and sensitivity. These results suggest that variations in microbial abundance and activities in the infant gut were mainly explained by metabolic interactions, and could be properly modeled using Monod kinetics with metabolic interactions. The model could be scaled to include data from larger consortia, or be applied to microbial communities where sharing metabolic resources is important in shaping bacterial abundance. Moreover, the model could be useful in designing microbial consortia with desired properties such as higher acid production.
Collapse
Affiliation(s)
| | | | | | - Daniel Garrido
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
636
|
Iizumi T, Battaglia T, Ruiz V, Perez Perez GI. Gut Microbiome and Antibiotics. Arch Med Res 2017; 48:727-734. [PMID: 29221800 DOI: 10.1016/j.arcmed.2017.11.004] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/15/2017] [Indexed: 12/12/2022]
Abstract
Despite that the human gastrointestinal tract is the most populated ecological niche by bacteria in the human body, much is still unknown about its characteristics. This site is highly susceptible to the effects of many external factors that may affect in the quality and the quantity of the microbiome. Specific factors such as diet, personal hygiene, pharmacological drugs and the use of antibiotics can produce a significant impact on the gut microbiota. The effect of these factors is more relevant early in life, when the gut microbiota has not yet fully established. In this review, we discussed the effect of type and doses of the antibiotics on the gut microbiota and what the major consequences in the use and abuse of these antimicrobial agents.
Collapse
Affiliation(s)
- Tadasu Iizumi
- Department of Medicine, New York University School of Medicine NYU Langone Health, New York, NY, USA.
| | - Thomas Battaglia
- Department of Medicine, New York University School of Medicine NYU Langone Health, New York, NY, USA
| | - Victoria Ruiz
- Department of Medicine, New York University School of Medicine NYU Langone Health, New York, NY, USA
| | - Guillermo I Perez Perez
- Department of Medicine, New York University School of Medicine NYU Langone Health, New York, NY, USA
| |
Collapse
|
637
|
Konstantinov SR. Diet, microbiome, and colorectal cancer. Best Pract Res Clin Gastroenterol 2017; 31:675-681. [PMID: 29566911 DOI: 10.1016/j.bpg.2017.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 09/03/2017] [Indexed: 01/31/2023]
Abstract
The scientific interests in the colorectal cancer (CRC) associated microbiome have increased significantly in the past decade. Mechanistically, several members of the human microbiome and products thereof have been implicated as inductors of the pathogenic inflammation related to CRC. Conversely, the activities of the human intestinal microbial community influenced by specific diet might confer a protective effect against the CRC risks and progression. As the microbiome is both a key contributor and one of the tools to prevent CRC, the current review gives a summary of the CRC-associated microbiome and the dietary strategies relevant to CRC. As more evidences become available, new microbiome-based treatments and specific diets may emerge to reduce the CRC risk and improve CRC patients' quality of life.
Collapse
Affiliation(s)
- Sergey R Konstantinov
- Department of Gasteroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, 's Gravendijkwal 230, NL-3015, CE Rotterdam, The Netherlands.
| |
Collapse
|
638
|
Milani C, Duranti S, Bottacini F, Casey E, Turroni F, Mahony J, Belzer C, Delgado Palacio S, Arboleya Montes S, Mancabelli L, Lugli GA, Rodriguez JM, Bode L, de Vos W, Gueimonde M, Margolles A, van Sinderen D, Ventura M. The First Microbial Colonizers of the Human Gut: Composition, Activities, and Health Implications of the Infant Gut Microbiota. Microbiol Mol Biol Rev 2017; 81:e00036-17. [PMID: 29118049 PMCID: PMC5706746 DOI: 10.1128/mmbr.00036-17] [Citation(s) in RCA: 1108] [Impact Index Per Article: 138.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The human gut microbiota is engaged in multiple interactions affecting host health during the host's entire life span. Microbes colonize the neonatal gut immediately following birth. The establishment and interactive development of this early gut microbiota are believed to be (at least partially) driven and modulated by specific compounds present in human milk. It has been shown that certain genomes of infant gut commensals, in particular those of bifidobacterial species, are genetically adapted to utilize specific glycans of this human secretory fluid, thus representing a very intriguing example of host-microbe coevolution, where both partners are believed to benefit. In recent years, various metagenomic studies have tried to dissect the composition and functionality of the infant gut microbiome and to explore the distribution across the different ecological niches of the infant gut biogeography of the corresponding microbial consortia, including those corresponding to bacteria and viruses, in healthy and ill subjects. Such analyses have linked certain features of the microbiota/microbiome, such as reduced diversity or aberrant composition, to intestinal illnesses in infants or disease states that are manifested at later stages of life, including asthma, inflammatory bowel disease, and metabolic disorders. Thus, a growing number of studies have reported on how the early human gut microbiota composition/development may affect risk factors related to adult health conditions. This concept has fueled the development of strategies to shape the infant microbiota composition based on various functional food products. In this review, we describe the infant microbiota, the mechanisms that drive its establishment and composition, and how microbial consortia may be molded by natural or artificial interventions. Finally, we discuss the relevance of key microbial players of the infant gut microbiota, in particular bifidobacteria, with respect to their role in health and disease.
Collapse
Affiliation(s)
- Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Sabrina Duranti
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Francesca Bottacini
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Eoghan Casey
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Jennifer Mahony
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Susana Delgado Palacio
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Silvia Arboleya Montes
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Leonardo Mancabelli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Juan Miguel Rodriguez
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Lars Bode
- Department of Pediatrics and Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California-San Diego, La Jolla, California, USA
| | - Willem de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Department of Bacteriology & Immunology, RPU Immunobiology, University of Helsinki, Helsinki, Finland
| | - Miguel Gueimonde
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Abelardo Margolles
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA-CSIC, Villaviciosa, Asturias, Spain
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, National University of Ireland, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
639
|
Mallick H, Ma S, Franzosa EA, Vatanen T, Morgan XC, Huttenhower C. Experimental design and quantitative analysis of microbial community multiomics. Genome Biol 2017; 18:228. [PMID: 29187204 PMCID: PMC5708111 DOI: 10.1186/s13059-017-1359-z] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Studies of the microbiome have become increasingly sophisticated, and multiple sequence-based, molecular methods as well as culture-based methods exist for population-scale microbiome profiles. To link the resulting host and microbial data types to human health, several experimental design considerations, data analysis challenges, and statistical epidemiological approaches must be addressed. Here, we survey current best practices for experimental design in microbiome molecular epidemiology, including technologies for generating, analyzing, and integrating microbiome multiomics data. We highlight studies that have identified molecular bioactives that influence human health, and we suggest steps for scaling translational microbiome research to high-throughput target discovery across large populations.
Collapse
Affiliation(s)
- Himel Mallick
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Siyuan Ma
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Eric A Franzosa
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Tommi Vatanen
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Xochitl C Morgan
- Department of Microbiology and Immunology, The University of Otago, Dunedin, New Zealand
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
640
|
Ticinesi A, Lauretani F, Milani C, Nouvenne A, Tana C, Del Rio D, Maggio M, Ventura M, Meschi T. Aging Gut Microbiota at the Cross-Road between Nutrition, Physical Frailty, and Sarcopenia: Is There a Gut-Muscle Axis? Nutrients 2017; 9:nu9121303. [PMID: 29189738 PMCID: PMC5748753 DOI: 10.3390/nu9121303] [Citation(s) in RCA: 239] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/22/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023] Open
Abstract
Inadequate nutrition and physical inactivity are the mainstays of primary sarcopenia-physiopathology in older individuals. Gut microbiota composition is strongly dependent on both of these elements, and conversely, can also influence the host physiology by modulating systemic inflammation, anabolism, insulin sensitivity, and energy production. The bacterial metabolism of nutrients theoretically influences skeletal muscle cell functionality through producing mediators that drive all of these systemic effects. In this study, we review the scientific literature supporting the concept of the involvement of gut microbiota in primary sarcopenia physiopathology. First, we examine studies associating fecal microbiota alterations with physical frailty, i.e., the loss of muscle performance and normal muscle mass. Then, we consider studies exploring the effects of exercise on gut microbiota composition. Finally, we examine studies demonstrating the possible effects of mediators produced by gut microbiota on skeletal muscle, and intervention studies considering the effects of prebiotic or probiotic administration on muscle function. Even if there is no evidence of a distinct gut microbiota composition in older sarcopenic patients, we conclude that the literature supports the possible presence of a "gut-muscle axis", whereby gut microbiota may act as the mediator of the effects of nutrition on muscle cells.
Collapse
Affiliation(s)
- Andrea Ticinesi
- Microbiome Research Hub, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy.
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126 Parma, Italy.
- Dipartimento Medico-Geriatrico-Riabilitativo, Azienda Ospedaliero-Universitaria di Parma, Via Antonio Gramsci 14, 43126 Parma, Italy.
| | - Fulvio Lauretani
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126 Parma, Italy.
- Dipartimento Medico-Geriatrico-Riabilitativo, Azienda Ospedaliero-Universitaria di Parma, Via Antonio Gramsci 14, 43126 Parma, Italy.
| | - Christian Milani
- Microbiome Research Hub, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy.
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy.
| | - Antonio Nouvenne
- Microbiome Research Hub, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy.
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126 Parma, Italy.
- Dipartimento Medico-Geriatrico-Riabilitativo, Azienda Ospedaliero-Universitaria di Parma, Via Antonio Gramsci 14, 43126 Parma, Italy.
| | - Claudio Tana
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126 Parma, Italy.
- Dipartimento Medico-Geriatrico-Riabilitativo, Azienda Ospedaliero-Universitaria di Parma, Via Antonio Gramsci 14, 43126 Parma, Italy.
| | - Daniele Del Rio
- Microbiome Research Hub, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy.
- Laboratory of Phytochemicals in Physiology, Department of Food and Drug Science, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy.
| | - Marcello Maggio
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126 Parma, Italy.
- Dipartimento Medico-Geriatrico-Riabilitativo, Azienda Ospedaliero-Universitaria di Parma, Via Antonio Gramsci 14, 43126 Parma, Italy.
| | - Marco Ventura
- Microbiome Research Hub, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy.
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy.
| | - Tiziana Meschi
- Microbiome Research Hub, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy.
- Department of Medicine and Surgery, University of Parma, Via Antonio Gramsci 14, 43126 Parma, Italy.
- Dipartimento Medico-Geriatrico-Riabilitativo, Azienda Ospedaliero-Universitaria di Parma, Via Antonio Gramsci 14, 43126 Parma, Italy.
| |
Collapse
|
641
|
Complete Genome Sequence of Collinsella aerofaciens Isolated from the Gut of a Healthy Indian Subject. GENOME ANNOUNCEMENTS 2017; 5:5/47/e01361-17. [PMID: 29167267 PMCID: PMC5701492 DOI: 10.1128/genomea.01361-17] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Collinsella aerofaciens, a rod-shaped nonmotile obligate anaerobe, is the most abundant actinobacterium in the gastrointestinal tract of healthy humans. An altered abundance of C. aerofaciens may be linked with several health disorders, including irritable bowel syndrome. In the present study, we report the complete genome sequence of C. aerofaciens strain indica.
Collapse
|
642
|
Brown RL, Sequeira RP, Clarke TB. The microbiota protects against respiratory infection via GM-CSF signaling. Nat Commun 2017; 8:1512. [PMID: 29142211 PMCID: PMC5688119 DOI: 10.1038/s41467-017-01803-x] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 10/17/2017] [Indexed: 02/08/2023] Open
Abstract
The microbiota promotes resistance to respiratory infection, but the mechanistic basis for this is poorly defined. Here, we identify members of the microbiota that protect against respiratory infection by the major human pathogens Streptococcus pneumoniae and Klebsiella pneumoniae. We show that the microbiota enhances respiratory defenses via granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling, which stimulates pathogen killing and clearance by alveolar macrophages through extracellular signal-regulated kinase signaling. Increased pulmonary GM-CSF production in response to infection is primed by the microbiota through interleukin-17A. By combining models of commensal colonization in antibiotic-treated and germ-free mice, using cultured commensals from the Actinobacteria, Bacteroidetes, Firmicutes, and Proteobacteria phyla, we found that potent Nod-like receptor-stimulating bacteria in the upper airway (Staphylococcus aureus and Staphylococcus epidermidis) and intestinal microbiota (Lactobacillus reuteri, Enterococcus faecalis, Lactobacillus crispatus and Clostridium orbiscindens) promote resistance to lung infection through Nod2 and GM-CSF. Our data reveal the identity, location, and properties of bacteria within the microbiota that regulate lung immunity, and delineate the host signaling axis they activate to protect against respiratory infection.
Collapse
Affiliation(s)
- Rebecca L Brown
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, SW7 2AZ, UK
| | - Richard P Sequeira
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, SW7 2AZ, UK
| | - Thomas B Clarke
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
643
|
Roth-Walter F, Pacios LF, Bianchini R, Jensen-Jarolim E. Linking iron-deficiency with allergy: role of molecular allergens and the microbiome. Metallomics 2017; 9:1676-1692. [PMID: 29120476 DOI: 10.1039/c7mt00241f] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Atopic individuals tend to develop a Th2 dominant immune response, resulting in hyperresponsiveness to harmless antigens, termed allergens. In the last decade, epidemiological studies have emerged that connected allergy with a deficient iron-status. Immune activation under iron-deficient conditions results in the expansion of Th2-, but not Th1 cells, can induce class-switching in B-cells and hampers the proper activation of M2, but not M1 macrophages. Moreover, many allergens, in particular with the lipocalin and lipocalin-like folds, seem to be capable of binding iron indirectly via siderophores harboring catechol moieties. The resulting locally restricted iron-deficiency may then lead during immune activation to the generation of Th2-cells and thus prepare for allergic sensitization. Moreover, iron-chelators seem to also influence clinical reactivity: mast cells accumulate iron before degranulation and seem to respond differently depending on the type of the encountered siderophore. Whereas deferoxamine triggers degranulation of connective tissue-type mast cells, catechol-based siderophores reduce activation and degranulation and improve clinical symptoms. Considering the complex interplay of iron, siderophores and immune molecules, it remains to be determined whether iron-deficiencies are the cause or the result of allergy.
Collapse
Affiliation(s)
- Franziska Roth-Walter
- Department of Comparative Medicine, at the Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University of Vienna and University of Vienna, Vienna, Austria.
| | - Luis F Pacios
- Centro de Biotecnología y Genómica de Plantas (CBGP, UPM-INIA), Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Campus de Montegancedo-UPM, 28223 Pozuelo de Alarcón, Madrid, Spain
| | - Rodolfo Bianchini
- Department of Comparative Medicine, at the Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University of Vienna and University of Vienna, Vienna, Austria.
| | - Erika Jensen-Jarolim
- Department of Comparative Medicine, at the Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University of Vienna and University of Vienna, Vienna, Austria. and Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
644
|
Tian Y, Nichols RG, Cai J, Patterson AD, Cantorna MT. Vitamin A deficiency in mice alters host and gut microbial metabolism leading to altered energy homeostasis. J Nutr Biochem 2017; 54:28-34. [PMID: 29227833 DOI: 10.1016/j.jnutbio.2017.10.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/28/2017] [Accepted: 10/18/2017] [Indexed: 12/24/2022]
Abstract
Vitamin A deficiency (A-) is a worldwide public health problem. To better understand how vitamin A status influences gut microbiota and host metabolism, we systematically analyzed urine, cecum, serum and liver samples from vitamin A sufficient (A+) and deficient (A-) mice using 1H NMR-based metabolomics, quantitative (q)PCR and 16S rRNA gene sequencing coupled with multivariate data analysis. The microbiota in the cecum of A- mice showed compositional as well as functional shifts compared to the microbiota from A+ mice. Targeted 1H NMR analyses revealed significant changes in microbial metabolite concentrations including higher butyrate and hippurate and decreased acetate and 4-hydroxyphenylacetate in A+ relative to A- mice. Bacterial butyrate-producing genes including butyryl-CoA:acetate CoA-transferase and butyrate kinase were significantly higher in bacteria from A+ versus bacteria from A- mice. A- mice had disturbances in multiple metabolic pathways including alterations in energy (hyperglycemia, glycogenesis, TCA cycle and lipoprotein biosynthesis), amino acid and nucleic acid metabolism. A- mice had hyperglycemia, liver dysfunction, changes in bacterial metabolism and altered gut microbial communities. Moreover, integrative analyses indicated a strong correlation between gut microbiota and host energy metabolism pathways in the liver. Vitamin A regulates host and bacterial metabolism, and the result includes alterations in energy homeostasis.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, United States; Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, United States; CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, University of Chinese Academy of Sciences, Wuhan, 430071, China
| | - Robert G Nichols
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, United States
| | - Jingwei Cai
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, United States
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, United States; Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, United States
| | - Margherita T Cantorna
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, United States.
| |
Collapse
|
645
|
Patrone V, Puglisi E, Cardinali M, Schnitzler TS, Svegliati S, Festa A, Gabrielli A, Morelli L. Gut microbiota profile in systemic sclerosis patients with and without clinical evidence of gastrointestinal involvement. Sci Rep 2017; 7:14874. [PMID: 29093509 PMCID: PMC5665912 DOI: 10.1038/s41598-017-14889-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 10/18/2017] [Indexed: 02/08/2023] Open
Abstract
Recent evidence suggests that there is a link between the gut microbial community and immune-mediated disorders. Systemic sclerosis (SSc) is an autoimmune disease characterized by immunonological abnormalities, vascular lesions, and extensive fibrosis. Since the gastrointestinal tract is one of the organs most involved, the goal of this study was to explore the composition of the intestinal microbiota in SSc patients with (SSc/GI+) and without gastrointestinal involvement (SSc/GI-) in comparison to healthy controls (HC). The fecal bacterial composition was investigated by Illumina sequencing of 16 S rRNA gene amplicons. The fecal microbiota of SSc/GI+ subjects was characterized by higher levels of Lactobacillus, Eubacterium and Acinetobacter compared with healthy controls, and lower proportions of Roseburia, Clostridium, and Ruminococcus. The gut microbiota of SSc/GI- subjects was more similar to the microbiota of HC than to that of SSc/GI+ subjects albeit Streptococcus salivarius was over-represented in SSc/GI- fecal samples compared with both SSc/GI+ subjects and controls. Our study reveals microbial signatures of dysbiosis in the gut microbiota of SSc patients that are associated with clinical evidence of gastrointestinal disease. Further studies are needed to elucidate the potential role of these perturbations in the onset and progression of systemic sclerosis, and gastrointestinal involvement in particular.
Collapse
Affiliation(s)
- Vania Patrone
- Dipartimento di Scienze e Tecnologie Alimentari per una filiera agroalimentare sostenibile (DiSTAS), Facoltà di Scienze Agrarie, Alimentari ed Ambientali, Università Cattolica del Sacro Cuore, Via Emilia Parmense 84, 29122, Piacenza, Italy
| | - Edoardo Puglisi
- Dipartimento di Scienze e Tecnologie Alimentari per una filiera agroalimentare sostenibile (DiSTAS), Facoltà di Scienze Agrarie, Alimentari ed Ambientali, Università Cattolica del Sacro Cuore, Via Emilia Parmense 84, 29122, Piacenza, Italy.
| | - Marco Cardinali
- Clinica Medica, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Via Tronto 10/A, 60126, Ancona, Italy
| | - Tobias S Schnitzler
- Clinica Medica, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Via Tronto 10/A, 60126, Ancona, Italy
| | - Silvia Svegliati
- Clinica Medica, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Via Tronto 10/A, 60126, Ancona, Italy
| | - Antonella Festa
- Clinica Medica, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Via Tronto 10/A, 60126, Ancona, Italy
| | - Armando Gabrielli
- Clinica Medica, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Via Tronto 10/A, 60126, Ancona, Italy.
| | - Lorenzo Morelli
- Dipartimento di Scienze e Tecnologie Alimentari per una filiera agroalimentare sostenibile (DiSTAS), Facoltà di Scienze Agrarie, Alimentari ed Ambientali, Università Cattolica del Sacro Cuore, Via Emilia Parmense 84, 29122, Piacenza, Italy
| |
Collapse
|
646
|
Schwager E, Mallick H, Ventz S, Huttenhower C. A Bayesian method for detecting pairwise associations in compositional data. PLoS Comput Biol 2017; 13:e1005852. [PMID: 29140991 PMCID: PMC5706738 DOI: 10.1371/journal.pcbi.1005852] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 11/29/2017] [Accepted: 10/25/2017] [Indexed: 12/12/2022] Open
Abstract
Compositional data consist of vectors of proportions normalized to a constant sum from a basis of unobserved counts. The sum constraint makes inference on correlations between unconstrained features challenging due to the information loss from normalization. However, such correlations are of long-standing interest in fields including ecology. We propose a novel Bayesian framework (BAnOCC: Bayesian Analysis of Compositional Covariance) to estimate a sparse precision matrix through a LASSO prior. The resulting posterior, generated by MCMC sampling, allows uncertainty quantification of any function of the precision matrix, including the correlation matrix. We also use a first-order Taylor expansion to approximate the transformation from the unobserved counts to the composition in order to investigate what characteristics of the unobserved counts can make the correlations more or less difficult to infer. On simulated datasets, we show that BAnOCC infers the true network as well as previous methods while offering the advantage of posterior inference. Larger and more realistic simulated datasets further showed that BAnOCC performs well as measured by type I and type II error rates. Finally, we apply BAnOCC to a microbial ecology dataset from the Human Microbiome Project, which in addition to reproducing established ecological results revealed unique, competition-based roles for Proteobacteria in multiple distinct habitats.
Collapse
Affiliation(s)
- Emma Schwager
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Himel Mallick
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Steffen Ventz
- Department of Computer Science and Statistics, University of Rhode Island, Kingstown, Rhode Island, United States of America
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| |
Collapse
|
647
|
Dahl C, Stanislawski M, Iszatt N, Mandal S, Lozupone C, Clemente JC, Knight R, Stigum H, Eggesbø M. Gut microbiome of mothers delivering prematurely shows reduced diversity and lower relative abundance of Bifidobacterium and Streptococcus. PLoS One 2017; 12:e0184336. [PMID: 29069100 PMCID: PMC5656300 DOI: 10.1371/journal.pone.0184336] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 08/22/2017] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Preterm birth is the main reason for neonatal deaths worldwide. We investigate whether maternal gut microbiota may play a previously overlooked role. METHODS The Norwegian Microbiota Study (NoMIC) is a case control study on preterm birth (<259 days of gestation, calculated primarily based on the last menstrual period), including two consecutively born term infants per infant born prematurely. Eligible mothers were fluent in Norwegian and recruited from the maternity ward at a county hospital in Eastern Norway in the period 2002-2005. Fecal samples were collected at day 4 postpartum, and analyzed using 16S ribosomal RNA gene sequencing. We used samples from 121 mothers giving birth vaginally. Measures of alpha diversity (Shannon, Phylogenetic Diversity and Observed Operational Taxonomic Units) and microbiome composition were combined with information from the Medical Birth Registry, pregnancy journals, and questionnaires. RESULTS The association between maternal gut diversity and preterm delivery was examined using logistic regression. One IQR increase in Shannon diversity was significantly associated with 38% lower odds of spontaneous preterm birth, (95% confident interval (CI): 1%, 61%), and the association was stronger when adjusting for maternal age, marital status, ethnicity, parity, BMI, education, antibiotic use, pets in the household, income and smoking (48% lower odds, 95% CI: 4.2%, 72%). Mothers delivering prematurely also had lower abundance of OTUs belonging to Bifidobacterium and Streptococcus, and of the Clostridiales order. CONCLUSION Analysis of maternal gut microbiota using next-generation sequencing shows that low gut diversity, with a distinct microbial composition, is associated with spontaneous preterm delivery.
Collapse
Affiliation(s)
- Cecilie Dahl
- Department of Environmental Exposure and Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | - Maggie Stanislawski
- University of Colorado School of Medicine, Colorado, United States of America
| | - Nina Iszatt
- Department of Environmental Exposure and Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | | | - Catherine Lozupone
- University of Colorado School of Medicine, Colorado, United States of America
| | - Jose C. Clemente
- Department of Genetics and Genomic Sciences and Icahn Institute for Genomics & Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, United States of America
- Department of Medicine, Division of Clinical Immunology and Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Rob Knight
- Departments of Pediatrics and Computer Science & Engineering, University of California San Diego, San Diego, United States of America
| | - Hein Stigum
- Department of Non-Communicable Diseases, Norwegian Institute of Public Health, Oslo, Norway
| | - Merete Eggesbø
- Department of Environmental Exposure and Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
- * E-mail:
| |
Collapse
|
648
|
Vogt NM, Kerby RL, Dill-McFarland KA, Harding SJ, Merluzzi AP, Johnson SC, Carlsson CM, Asthana S, Zetterberg H, Blennow K, Bendlin BB, Rey FE. Gut microbiome alterations in Alzheimer's disease. Sci Rep 2017; 7:13537. [PMID: 29051531 PMCID: PMC5648830 DOI: 10.1038/s41598-017-13601-y] [Citation(s) in RCA: 1325] [Impact Index Per Article: 165.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/27/2017] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. However, the etiopathogenesis of this devastating disease is not fully understood. Recent studies in rodents suggest that alterations in the gut microbiome may contribute to amyloid deposition, yet the microbial communities associated with AD have not been characterized in humans. Towards this end, we characterized the bacterial taxonomic composition of fecal samples from participants with and without a diagnosis of dementia due to AD. Our analyses revealed that the gut microbiome of AD participants has decreased microbial diversity and is compositionally distinct from control age- and sex-matched individuals. We identified phylum- through genus-wide differences in bacterial abundance including decreased Firmicutes, increased Bacteroidetes, and decreased Bifidobacterium in the microbiome of AD participants. Furthermore, we observed correlations between levels of differentially abundant genera and cerebrospinal fluid (CSF) biomarkers of AD. These findings add AD to the growing list of diseases associated with gut microbial alterations, as well as suggest that gut bacterial communities may be a target for therapeutic intervention.
Collapse
Affiliation(s)
- Nicholas M Vogt
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue J5/1 Mezzanine, Madison, WI 53792, USA
| | - Robert L Kerby
- Department of Bacteriology, University of Wisconsin-Madison, 1550 Linden Drive, Madison, WI 53706, USA
| | - Kimberly A Dill-McFarland
- Department of Bacteriology, University of Wisconsin-Madison, 1550 Linden Drive, Madison, WI 53706, USA
| | - Sandra J Harding
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue J5/1 Mezzanine, Madison, WI 53792, USA
| | - Andrew P Merluzzi
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue J5/1 Mezzanine, Madison, WI 53792, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue J5/1 Mezzanine, Madison, WI 53792, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, WI 53705, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, WARF Building, 610 Walnut Street, 9th Floor, Suite 957, Madison, WI 53726, USA
| | - Cynthia M Carlsson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue J5/1 Mezzanine, Madison, WI 53792, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, WI 53705, USA
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, WARF Building, 610 Walnut Street, 9th Floor, Suite 957, Madison, WI 53726, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue J5/1 Mezzanine, Madison, WI 53792, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, WI 53705, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Molecular Neuroscience, University College London Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at University College London, London, United Kingdom
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue J5/1 Mezzanine, Madison, WI 53792, USA.
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, WARF Building, 610 Walnut Street, 9th Floor, Suite 957, Madison, WI 53726, USA.
| | - Federico E Rey
- Department of Bacteriology, University of Wisconsin-Madison, 1550 Linden Drive, Madison, WI 53706, USA.
| |
Collapse
|
649
|
Consumption of kiwifruit capsules increases Faecalibacterium prausnitzii abundance in functionally constipated individuals: a randomised controlled human trial. J Nutr Sci 2017; 6:e52. [PMID: 29152256 PMCID: PMC5672330 DOI: 10.1017/jns.2017.52] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/19/2017] [Accepted: 08/01/2017] [Indexed: 12/12/2022] Open
Abstract
This study investigated the impact of ACTAZIN™ green (2400 and 600 mg) and Livaux™ (2400 mg) gold kiwifruit supplements on faecal microbial composition and metabolites in healthy and functionally constipated (FC) participants. The participants were recruited into the healthy group (n 20; one of whom did not complete the study) and the FC group (n 9), each of whom consumed all the treatments and a placebo (isomalt) for 4 weeks in a randomised cross-over design interspersed with 2-week washout periods. Modification of faecal microbiota composition and metabolism was determined by 16S rRNA gene sequencing and GC, and colonic pH was calculated using SmartPill® wireless motility capsules. A total of thirty-two taxa were measured at greater than 1 % abundance in at least one sample, ten of which differed significantly between the baseline healthy and FC groups. Specifically, Bacteroidales and Roseburia spp. were significantly more abundant (P < 0·05) in the healthy group and taxa including Ruminococcaceae, Dorea spp. and Akkermansia spp. were significantly more abundant (P < 0·05) in the FC group. In the FC group, Faecalibacterium prausnitzii abundance significantly increased (P = 0·024) from 3·4 to 7·0 % following Livaux™ supplementation, with eight of the nine participants showing a net increase. Lower proportions of F. prausnitzii are often associated with gastrointestinal disorders. The discovery that Livaux™ supplementation increased F. prausnitzii abundance offers a potential strategy for improving gut microbiota composition, as F. prausnitzii is a butyrate producer and has also been shown to exert anti-inflammatory effects in many studies.
Collapse
|
650
|
Vinderola G, Gueimonde M, Gomez-Gallego C, Delfederico L, Salminen S. Correlation between in vitro and in vivo assays in selection of probiotics from traditional species of bacteria. Trends Food Sci Technol 2017. [DOI: 10.1016/j.tifs.2017.08.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|