1
|
Chakravarty K, Gaur S, Kumar R, Jha NK, Gupta PK. Exploring the Multifaceted Therapeutic Potential of Probiotics: A Review of Current Insights and Applications. Probiotics Antimicrob Proteins 2025; 17:341-363. [PMID: 39069588 DOI: 10.1007/s12602-024-10328-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2024] [Indexed: 07/30/2024]
Abstract
The interplay between human health and the microbiome has gained extensive attention, with probiotics emerging as pivotal therapeutic agents due to their vast potential in treating various health issues. As significant modulators of the gut microbiota, probiotics are crucial in maintaining intestinal homeostasis and enhancing the synthesis of short-chain fatty acids. Despite extensive research over the past decades, there remains an urgent need for a comprehensive and detailed review that encapsulates probiotics' latest insights and applications. This review focusses on the multifaceted roles of probiotics in promoting health and preventing disease, highlighting the complex mechanisms through which these beneficial bacteria influence both gut flora and the human body at large. This paper also explores probiotics' neurological and gastrointestinal applications, focussing on their significant impact on the gut-brain axis and their therapeutic potential in a broad spectrum of pathological conditions. Current innovations in probiotic formulations, mainly focusing on integrating genomics and biotechnological advancements, have also been comprehensively discussed herein. This paper also critically examines the regulatory landscape that governs probiotic use, ensuring safety and efficacy in clinical and dietary settings. By presenting a comprehensive overview of recent studies and emerging trends, this review aims to illuminate probiotics' extensive therapeutic capabilities, leading to future research and clinical applications. However, besides extensive research, further advanced explorations into probiotic interactions and mechanisms will be essential for developing more targeted and effective therapeutic strategies, potentially revolutionizing health care practices for consumers.
Collapse
Affiliation(s)
- Kashyapi Chakravarty
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh, 201309, India
| | - Smriti Gaur
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh, 201309, India.
| | - Rohit Kumar
- Centre for Development of Biomaterials and Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Niraj Kumar Jha
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, 602105, India
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, 140401, India
| | - Piyush Kumar Gupta
- Centre for Development of Biomaterials and Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, 201310, India.
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand, 248002, India.
| |
Collapse
|
2
|
Pan Z, Luo H, He F, Du Y, Wang J, Zeng H, Xu Z, Sun Y, Li M. Guava polysaccharides attenuate high fat and STZ-induced hyperglycemia by regulating gut microbiota and arachidonic acid metabolism. Int J Biol Macromol 2024; 276:133725. [PMID: 38986994 DOI: 10.1016/j.ijbiomac.2024.133725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/18/2024] [Accepted: 07/05/2024] [Indexed: 07/12/2024]
Abstract
This study investigated the hypoglycemic mechanism of guava polysaccharides (GP) through the gut microbiota (GM) and related metabolites. Our findings demonstrated that GP significantly mitigated high-fat diet- and streptozotocin-induced hyperglycemia, insulin resistance, hyperlipidemia, elevated alanine aminotransferase, high hepatic inflammation levels, and prevented pancreatic atrophy and hepatomegaly. Interestingly, the benefits of GP were attributed to alterations in the GM. GP decreased the ratio of Firmicutes to Bacteroidetes, significantly inhibiting deleterious bacteria, including Uncultured_f_Desulfovibrionaceae, Bilophila, and Desulfovibrio, while promoting the proliferation of probiotic Bifidobacterium and Bacteroides. In addition, GP promoted the generation of short-chain fatty acids. Notably, the arachidonic acid (AA) metabolism pathway was enriched in liver metabolites. GP significantly elevated hepatic AA and 15-hydroxyeicosatetraenoic acid, while reducing prostaglandin E2 and 5- and 12-hydroxyeicosatetraenoic acid. This modulation is accompanied by the downregulation of hepatic cyclooxygenase-1, 12-lipoxygenase, P38, and c-Jun N-terminal kinase mRNA expression, and the upregulation of cytochrome P4502J5 and insulin receptor substrate 1/2 mRNA expression. However, GP antibiotic treatment did not induce significant alterations in FBG and AA levels or gene expression. Overall, our findings suggest that the hypoglycemic effect of GP may be intricately linked to alterations in AA metabolism, which depends on the GM.
Collapse
Affiliation(s)
- Zhuangguang Pan
- Guangdong Provincial Key Lab of Food Safety and Quality, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; College of Food Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Haolin Luo
- Guangdong Provincial Key Lab of Food Safety and Quality, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; College of Food Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Fangqing He
- Guangdong Provincial Key Lab of Food Safety and Quality, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; College of Food Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Yixuan Du
- Guangdong Provincial Key Lab of Food Safety and Quality, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; College of Food Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Junyi Wang
- Guangdong Provincial Key Lab of Food Safety and Quality, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; College of Food Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Huize Zeng
- Guangdong Provincial Key Lab of Food Safety and Quality, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; College of Food Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Zhenlin Xu
- Guangdong Provincial Key Lab of Food Safety and Quality, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; College of Food Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Yuanming Sun
- Guangdong Provincial Key Lab of Food Safety and Quality, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; College of Food Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Meiying Li
- Guangdong Provincial Key Lab of Food Safety and Quality, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; College of Food Science, South China Agricultural University, Guangzhou 510642, PR China.
| |
Collapse
|
3
|
Xia T, Zhang Z, Zhao Y, Kang C, Zhang X, Tian Y, Yu J, Cao H, Wang M. The anti-diabetic activity of polyphenols-rich vinegar extract in mice via regulating gut microbiota and liver inflammation. Food Chem 2022; 393:133443. [PMID: 35751216 DOI: 10.1016/j.foodchem.2022.133443] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 01/11/2023]
Abstract
Polyphenols in vinegar are benefit to human health. The purpose of this research was to identify the polyphenols-rich vinegar extract (VE) and evaluate the anti-diabetic mechanisms in vivo. The results showed that 29 polyphenols were identified by UPLC-Q/Trap-MS/MS analysis. 4-Hydroxybenzoic acid, ferulic acid, and ethyl ferulate were the main polyphenols. In addition, VE relieved the symptoms of type 2 diabetes mellitus (T2DM) by down-regulating blood glucose and lipemia. VE reduced inflammation by inhibiting TLR4/NF-κB signaling pathway. Furthermore, VE treatment restored gut microbiota dysbiosis (upregulating Bacteroidetes, Lactobacillus, Bifidobacterium, and Bacteroides and downregulating Firmicutes, Proteobacteria, and Enterorhabdus abundances), and increased short chain fatty acids contents in diabetic mice, which participated in anti-diabetic effect of VE by correlation analysis. These findings suggest that VE may be a candidate for T2DM intervention by regulating gut microbiota and inflammation.
Collapse
Affiliation(s)
- Ting Xia
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China.
| | - Zhujun Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China.
| | - Yuxuan Zhao
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China.
| | - Chaoyan Kang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China.
| | - Xianglong Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China.
| | - Yinglei Tian
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China.
| | - Jiaqi Yu
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China.
| | - Hui Cao
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088, China.
| | - Min Wang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China.
| |
Collapse
|
4
|
Michel C, Blottière HM. Neonatal Programming of Microbiota Composition: A Plausible Idea That Is Not Supported by the Evidence. Front Microbiol 2022; 13:825942. [PMID: 35783422 PMCID: PMC9247513 DOI: 10.3389/fmicb.2022.825942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
Underpinning the theory "developmental origins of health and disease" (DOHaD), evidence is accumulating to suggest that the risks of adult disease are in part programmed by exposure to environmental factors during the highly plastic "first 1,000 days of life" period. An elucidation of the mechanisms involved in this programming is challenging as it would help developing new strategies to promote adult health. The intestinal microbiome is proposed as a long-lasting memory of the neonatal environment. This proposal is supported by indisputable findings such as the concomitance of microbiota assembly and the first 1,000-day period, the influence of perinatal conditions on microbiota composition, and the impact of microbiota composition on host physiology, and is based on the widely held but unconfirmed view that the microbiota is long-lastingly shaped early in life. In this review, we examine the plausibility of the gut microbiota being programmed by the neonatal environment and evaluate the evidence for its validity. We highlight that the capacity of the pioneer bacteria to control the implantation of subsequent bacteria is supported by both theoretical principles and statistical associations, but remains to be demonstrated experimentally. In addition, our critical review of the literature on the long-term repercussions of selected neonatal modulations of the gut microbiota indicates that sustained programming of the microbiota composition by neonatal events is unlikely. This does not exclude the microbiota having a role in DOHaD due to a possible interaction with tissue and organ development during the critical windows of neonatal life.
Collapse
Affiliation(s)
| | - Hervé M. Blottière
- Nantes Université, INRAE, UMR 1280, PhAN, Nantes, France
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, France
| |
Collapse
|
5
|
Meliț LE, Mărginean CO, Săsăran MO. The Yin-Yang Concept of Pediatric Obesity and Gut Microbiota. Biomedicines 2022; 10:biomedicines10030645. [PMID: 35327446 PMCID: PMC8945275 DOI: 10.3390/biomedicines10030645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 02/04/2023] Open
Abstract
The era of pediatric obesity is no longer a myth. Unfortunately, pediatric obesity has reached alarming incidence levels worldwide and the factors that contribute to its development have been intensely studied in multiple recent and emerging studies. Gut microbiota was recently included in the wide spectrum of factors implicated in the determination of obesity, but its role in pediatric obese patients is far from being fully understood. In terms of the infant gut microbiome, multiple factors have been demonstrated to shape its content, including maternal diet and health, type of delivery, feeding patterns, weaning and dietary habits. Nevertheless, the role of the intrauterine environment, such as the placental microbial community, cannot be completely excluded. Most studies have identified Firmicutes and Bacteroidetes as the most important players related to obesity risk in gut microbiota reflecting an increase of Firmicutes and a decrease in Bacteroidetes in the context of obesity; however, multiple inconsistencies between studies were recently reported, especially in pediatric populations, and there is a scarcity of studies performed in this age group.
Collapse
Affiliation(s)
- Lorena Elena Meliț
- Department of Pediatrics I, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania;
| | - Cristina Oana Mărginean
- Department of Pediatrics I, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania;
- Correspondence:
| | - Maria Oana Săsăran
- Department of Pediatrics III, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania;
| |
Collapse
|
6
|
Lun YZ, Qiu W, Zhao W, Lin H, Zhong M, Sun J. Characteristics of Intestinal Flora in Pregnant Women with Mild Thalassemia Revealed by Metagenomics. Jundishapur J Microbiol 2021; 14. [DOI: 10.5812/jjm.119925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/26/2021] [Accepted: 11/26/2021] [Indexed: 09/02/2023] Open
Abstract
Background: At present, there is no report that the intestinal flora of pregnant women with mild thalassemia is different from that of healthy pregnant women. Objectives: This study compared the composition and changes of the intestinal flora of pregnant women with mild thalassemia to those of healthy pregnant women using metagenomic sequencing technology and evaluated the potential microecological risk for pregnant women and the fetus. Methods: The present study was carried out on 14 mild thalassemia pregnant women with similar backgrounds in the Affiliated Hospital of Putian University, Fujian, China. In the same period, 6 healthy pregnant women were selected as the control group. The genomic deoxyribonucleic acid was extracted from the sable stool samples of pregnant women. Illumina HiSeq sequencing technology was adopted after library preparation. Prodigal software (ver 2.6.3), Salmon software (ver 1.6.0), and Kraken software (ver 2) were used to analyze the sequence data. Moreover, analysis of variance and Duncan’s multiple-comparison test or Wilcoxon rank-sum test were used as statistical methods. Results: The characteristics of the intestinal flora of pregnant women with mild thalassemia differed significantly from those of healthy pregnant women, showing an increase in some conditionally pathogenic bacteria (e.g., Prevotella stercorea rose and Escherichia coli) and a decrease in some probiotic bacteria, which might affect pregnant women and cause physiological function damage to their offspring by changing metabolic pathways; however, further validation is needed. Conclusions: The diversity and composition of intestinal flora in pregnant women with mild thalassemia vary significantly from those in healthy pregnant women, especially at the genus and species levels, representing more profound alterations in intestinal microecology.
Collapse
|
7
|
Double-Barrel Shotgun: Probiotic Lactic Acid Bacteria with Antiviral Properties Modified to Serve as Vaccines. Microorganisms 2021; 9:microorganisms9081565. [PMID: 34442644 PMCID: PMC8401918 DOI: 10.3390/microorganisms9081565] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/17/2022] Open
Abstract
Contrary to the general belief that the sole function of probiotics is to keep intestinal microbiota in a balanced state and stimulate the host’s immune response, several studies have shown that certain strains of lactic acid bacteria (LAB) have direct and/or indirect antiviral properties. LAB can stimulate the innate antiviral immune defence system in their host, produce antiviral peptides, and release metabolites that prevent either viral replication or adhesion to cell surfaces. The SARS-CoV (COVID-19) pandemic shifted the world’s interest towards the development of vaccines against viral infections. It is hypothesised that the adherence of SARS-CoV spike proteins to the surface of Bifidobacterium breve could elicit an immune response in its host and trigger the production of antibodies. The question now remains as to whether probiotic LAB could be genetically modified to synthesize viral antigens and serve as vaccines—this concept and the role that LAB play in viral infection are explored in this review.
Collapse
|
8
|
Tabouy L, Getselter D, Ziv O, Karpuj M, Tabouy T, Lukic I, Maayouf R, Werbner N, Ben-Amram H, Nuriel-Ohayon M, Koren O, Elliott E. Dysbiosis of microbiome and probiotic treatment in a genetic model of autism spectrum disorders. Brain Behav Immun 2018; 73:310-319. [PMID: 29787855 DOI: 10.1016/j.bbi.2018.05.015] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 05/08/2018] [Accepted: 05/18/2018] [Indexed: 02/06/2023] Open
Abstract
Recent studies have determined that the microbiome has direct effects on behavior, and may be dysregulated in neurodevelopmental conditions. Considering that neurodevelopmental conditions, such as autism, have a strong genetic etiology, it is necessary to understand if genes associated with neurodevelopmental disorders, such as Shank3, can influence the gut microbiome, and if probiotics can be a therapeutic tool. In this study, we have identified dysregulation of several genera and species of bacteria in the gut and colon of both male and female Shank3 KO mice. L. reuteri, a species with decreased relative abundance in the Shank3 KO mice, positively correlated with the expression of gamma-Aminobutyric acid (GABA) receptor subunits in the brain. Treatment of Shank3 KO mice with L. reuteri induced an attenuation of unsocial behavior specifically in male Shank3 mice, and a decrease in repetitive behaviors in both male and female Shank3 KO mice. In addition, L. reuteri treatment affected GABA receptor gene expression and protein levels in multiple brain regions. This study identifies bacterial species that are sensitive to an autism-related mutation, and further suggests a therapeutic potential for probiotic treatment.
Collapse
Affiliation(s)
- Laure Tabouy
- Molecular and Behavioral Neurosciences Lab, Faculty of Medicine in the Galilee, Bar-Ilan University, 1311502 Safed, Israel
| | - Dimitry Getselter
- Molecular and Behavioral Neurosciences Lab, Faculty of Medicine in the Galilee, Bar-Ilan University, 1311502 Safed, Israel
| | - Oren Ziv
- Microbiome Research Lab, Faculty of Medicine in the Galilee, Bar-Ilan University, 1311502 Safed, Israel
| | - Marcela Karpuj
- Genomic Center Faculty of Medicine in the Galilee, Bar-Ilan University, 1311502 Safed, Israel; Biotechnology Engineering Department, Orte Braude, 21616 Karmiel, Israel
| | - Timothée Tabouy
- UMR 518 Applied Mathematics and Informatics (MIA)-Paris, French National Institute for Agricultural Research INRA/AgroParisTech, Paris-Saclay University, 75005 Paris, France
| | - Iva Lukic
- Molecular and Behavioral Neurosciences Lab, Faculty of Medicine in the Galilee, Bar-Ilan University, 1311502 Safed, Israel
| | - Rasha Maayouf
- Molecular and Behavioral Neurosciences Lab, Faculty of Medicine in the Galilee, Bar-Ilan University, 1311502 Safed, Israel
| | - Nir Werbner
- Microbiome Research Lab, Faculty of Medicine in the Galilee, Bar-Ilan University, 1311502 Safed, Israel
| | - Hila Ben-Amram
- Microbiome Research Lab, Faculty of Medicine in the Galilee, Bar-Ilan University, 1311502 Safed, Israel
| | - Meital Nuriel-Ohayon
- Microbiome Research Lab, Faculty of Medicine in the Galilee, Bar-Ilan University, 1311502 Safed, Israel
| | - Omry Koren
- Microbiome Research Lab, Faculty of Medicine in the Galilee, Bar-Ilan University, 1311502 Safed, Israel
| | - Evan Elliott
- Molecular and Behavioral Neurosciences Lab, Faculty of Medicine in the Galilee, Bar-Ilan University, 1311502 Safed, Israel.
| |
Collapse
|
9
|
Rutanga JP, Van Puyvelde S, Heroes AS, Muvunyi CM, Jacobs J, Deborggraeve S. 16S metagenomics for diagnosis of bloodstream infections: opportunities and pitfalls. Expert Rev Mol Diagn 2018; 18:749-759. [PMID: 29985081 DOI: 10.1080/14737159.2018.1498786] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Bacterial bloodstream infections (BSI) form a large public health threat worldwide. Current routine diagnosis is based on blood culture (BC) but this technique suffers from limited sensitivity. Molecular diagnostic tools have been developed for identification of bacteria in the blood of BSI patients. 16S metagenomics is an open-ended technique that can detect simultaneously all bacteria in a given sample based on PCR amplification of the 16S ribosomal RNA gene (rDNA) followed by sequencing of the PCR amplicons and taxonomic labeling of the sequence reads at genus or species level. Areas covered: Here we review the studies that have used 16S metagenomics for the identification of bacteria in human blood samples. We also discuss the potential added value of 16S metagenomics in the diagnosis of BSI, challenges as well as future directions for implementation in clinical settings. Expert commentary: 16S metagenomics has the potential to complement conventional BC; however, the technique currently suffers from several technical limitations jeopardizing implementation in routine clinical microbiology laboratories. Further studies are required to assess the cost-efficiency and clinical impact of 16S metagenomics in comparison to BC which remains the gold standard diagnostic method for BSI.
Collapse
Affiliation(s)
- Jean Pierre Rutanga
- a College of Science and Technology , University of Rwanda , Kigali , Rwanda.,b Department of Biomedical Sciences , Institute of Tropical Medicine , Antwerp , Belgium.,d Department of Microbiology and Immunology , KU Leuven , Leuven , Belgium
| | - Sandra Van Puyvelde
- b Department of Biomedical Sciences , Institute of Tropical Medicine , Antwerp , Belgium.,c Wellcome Trust Sanger Institute , Hinxton , United Kingdom
| | - Anne-Sophie Heroes
- d Department of Microbiology and Immunology , KU Leuven , Leuven , Belgium.,e Department of Clinical Sciences , Institute of Tropical Medicine , Antwerp , Belgium
| | - Claude Mambo Muvunyi
- f College of Medicine and Health Sciences , University of Rwanda , Kigali , Rwanda
| | - Jan Jacobs
- d Department of Microbiology and Immunology , KU Leuven , Leuven , Belgium.,e Department of Clinical Sciences , Institute of Tropical Medicine , Antwerp , Belgium
| | - Stijn Deborggraeve
- b Department of Biomedical Sciences , Institute of Tropical Medicine , Antwerp , Belgium
| |
Collapse
|
10
|
Zununi Vahed S, Moghaddas Sani H, Rahbar Saadat Y, Barzegari A, Omidi Y. Type 1 diabetes: Through the lens of human genome and metagenome interplay. Biomed Pharmacother 2018; 104:332-342. [PMID: 29775902 DOI: 10.1016/j.biopha.2018.05.052] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/09/2018] [Accepted: 05/09/2018] [Indexed: 02/07/2023] Open
Abstract
Diabetes is a genetic- and epigenetic-related disease from which a large population worldwide suffers. Some genetic factors along with various mutations related to the immune system for disease mechanism(s) have contrastively been determined. However, sometimes mechanisms have not been fully managed for the clarification of the initiation and/or progression of diseases to help patients. In the recent years, due to familiarity with the role of gut microbiota in the health, it has been found that the changes of the microbial balance in the industrialized societies can cause a battery of modern diseases, for which we have no specific definition of how they emerge. This work aims to explore the relationship between the human gut microbiota and the immune system along with their possible role in avoiding/emerging of type 1 diabetes (T1D) accompanied with the relation between genome and metagenome and their imbalance in causing T1D. Moreover, it provides novel view on how to balance the intestinal microbiota by lifestyle to hinder the mechanisms leading to T1D.
Collapse
Affiliation(s)
| | | | - Yalda Rahbar Saadat
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Barzegari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Mortzfeld BM, Taubenheim J, Fraune S, Klimovich AV, Bosch TCG. Stem Cell Transcription Factor FoxO Controls Microbiome Resilience in Hydra. Front Microbiol 2018; 9:629. [PMID: 29666616 PMCID: PMC5891625 DOI: 10.3389/fmicb.2018.00629] [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: 01/11/2018] [Accepted: 03/19/2018] [Indexed: 01/18/2023] Open
Abstract
The aging process is considered to be the result of accumulating cellular deterioration in an individual organism over time. It can be affected by the combined influence of genetic, epigenetic, and environmental factors including life-style-associated events. In the non-senescent freshwater polyp Hydra, one of the classical model systems for evolutionary developmental biology and regeneration, transcription factor FoxO modulates both stem cell proliferation and innate immunity. This provides strong support for the role of FoxO as a critical rate-of-aging regulator. However, how environmental factors interact with FoxO remains unknown. Here, we find that deficiency in FoxO signaling in Hydra leads to dysregulation of antimicrobial peptide expression and that FoxO loss-of-function polyps are impaired in selection for bacteria resembling the native microbiome and more susceptible to colonization of foreign bacteria. These findings reveal a key role of FoxO signaling in the communication between host and microbiota and embed the evolutionary conserved longevity factor FoxO into the holobiont concept.
Collapse
Affiliation(s)
| | - Jan Taubenheim
- Zoological Institute, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Sebastian Fraune
- Zoological Institute, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | | | - Thomas C G Bosch
- Zoological Institute, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| |
Collapse
|
12
|
Ma N, Guo P, Zhang J, He T, Kim SW, Zhang G, Ma X. Nutrients Mediate Intestinal Bacteria-Mucosal Immune Crosstalk. Front Immunol 2018; 9:5. [PMID: 29416535 PMCID: PMC5787545 DOI: 10.3389/fimmu.2018.00005] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 01/03/2018] [Indexed: 12/20/2022] Open
Abstract
The intestine is the shared site of nutrient digestion, microbiota colonization and immune cell location and this geographic proximity contributes to a large extent to their interaction. The onset and development of a great many diseases, such as inflammatory bowel disease and metabolic syndrome, will be caused due to the imbalance of body immune. As competent assistants, the intestinal bacteria are also critical in disease prevention and control. Moreover, the gut commensal bacteria are essential for development and normal operation of immune system and the pathogens are also closely bound up with physiological disorders and diseases mediated by immune imbalance. Understanding how our diet and nutrient affect bacterial composition and dynamic function, and the innate and adaptive status of our immune system, represents not only a research need but also an opportunity or challenge to improve health. Herein, this review focuses on the recent discoveries about intestinal bacteria–immune crosstalk and nutritional regulation on their interplay, with an aim to provide novel insights that can aid in understanding their interactions.
Collapse
Affiliation(s)
- Ning Ma
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Pingting Guo
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Jie Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China.,Animal Husbandry and Veterinary Department, Beijing Vocational College of Agriculture, Beijing, China
| | - Ting He
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC, United States
| | - Guolong Zhang
- Department of Animal Science, Oklahoma State University, Stillwater, OK, United States
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
13
|
Taylor-Mulneix DL, Hamidou Soumana I, Linz B, Harvill ET. Evolution of Bordetellae from Environmental Microbes to Human Respiratory Pathogens: Amoebae as a Missing Link. Front Cell Infect Microbiol 2017; 7:510. [PMID: 29322035 PMCID: PMC5732149 DOI: 10.3389/fcimb.2017.00510] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 11/27/2017] [Indexed: 12/16/2022] Open
Abstract
The genus Bordetella comprises several bacterial species that colonize the respiratory tract of mammals. It includes B. pertussis, a human-restricted pathogen that is the causative agent of Whooping Cough. In contrast, the closely related species B. bronchiseptica colonizes a broad range of animals as well as immunocompromised humans. Recent metagenomic studies have identified known and novel bordetellae isolated from different environmental sources, providing a new perspective on their natural history. Using phylogenetic analysis, we have shown that human and animal pathogenic bordetellae have most likely evolved from ancestors that originated from soil and water. Our recent study found that B. bronchiseptica can evade amoebic predation and utilize Dictyostelium discoideum as an expansion and transmission vector, which suggests that the evolutionary pressure to evade the amoebic predator enabled the rise of bordetellae as respiratory pathogens. Interactions with amoeba may represent the starting point for bacterial adaptation to eukaryotic cells. However, as bacteria evolve and adapt to a novel host, they can become specialized and restricted to a specific host. B. pertussis is known to colonize and cause infection only in humans, and this specialization to a closed human-to-human lifecycle has involved genome reduction and the loss of ability to utilize amoeba as an environmental reservoir. The discoveries from studying the interaction of Bordetella species with amoeba will elicit a better understanding of the evolutionary history of these and other important human pathogens.
Collapse
Affiliation(s)
- Dawn L Taylor-Mulneix
- Department of Infectious Diseases, Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Illiassou Hamidou Soumana
- Department of Infectious Diseases, Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Bodo Linz
- Department of Infectious Diseases, Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Eric T Harvill
- Department of Infectious Diseases, Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| |
Collapse
|
14
|
Hureaux J, Lacoeuille F, Lagarce F, Rousselet MC, Contini A, Saulnier P, Benoit JP, Urban T. Absence of lung fibrosis after a single pulmonary delivery of lipid nanocapsules in rats. Int J Nanomedicine 2017; 12:8159-8170. [PMID: 29184405 PMCID: PMC5687496 DOI: 10.2147/ijn.s146740] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lipid nanocapsules (LNCs) are potential drug carriers for pulmonary delivery since they can be nebulized without any structural or functional changes, and the aerosols produced are highly compatible with pulmonary drug delivery in human beings. The alveolar surface tension, in vitro cytotoxicity, biodistribution and pulmonary toxicity in rats of a single endotracheal spray of LNCs or paclitaxel-loaded LNCs were studied. In vitro cytotoxicity of LNCs after a spray remained unchanged. Biodistribution study showed a homogeneous repartition in the lungs in rats with an improvement in lung retention of the radiolabeled tracer loaded in LNCs compared to the absence of LNCs with a lung half-time of 8.8±0.7 hours. Bronchoalveolar fluid analysis revealed transient 7-day alveolar inflammation, reaching a maximum between days 2 and 4, characterized by a peak of granulocytes at day 1 followed by a peak of lymphocytes at day 3. Alveolar protein levels were increased at days 3 and 7. Acute inflammation was increased with paclitaxel-loaded LNCs in comparison with blank LNCs but dropped out at day 7. No histological pulmonary lesion was observed at day 60. LNCs lowered surface tension to a greater degree than Curosurf® in a physicochemical model of the pulmonary alveolus. A single pulmonary delivery of LNCs induces a short-term alveolar inflammation with no residual lesions in rats at day 60. These data permit to start the study of LNCs in surfactant replacement therapy.
Collapse
Affiliation(s)
- José Hureaux
- Unité Micro et Nanomédecines Biomimétiques (MINT), Université d'Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire.,Université d'Angers, CHU, Pôle Hippocrate, Service de Pneumologie
| | - Franck Lacoeuille
- Université d'Angers, CHU, Pôle Signal Image Stérilisation, Service de Médecine nucléaire.,CRCINA, Université Nantes Université Angers
| | - Frédéric Lagarce
- Unité Micro et Nanomédecines Biomimétiques (MINT), Université d'Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire.,Université d'Angers, CHU, Pôle Hippocrate, Pharmacie
| | - Marie-Christine Rousselet
- Université d'Angers, CHU, Pôle de Biologie-Pathologie, Département de Cytologie et d'Histologie Pathologique
| | - Aurélien Contini
- Université d'Angers, CHU, Pôle Signal Image Stérilisation, Service de Médecine nucléaire.,CRCINA, Université Nantes Université Angers
| | - Patrick Saulnier
- Unité Micro et Nanomédecines Biomimétiques (MINT), Université d'Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire.,Université d'Angers, CHU, Service la Recherche Clinique et Innovation, Angers, France
| | - Jean-Pierre Benoit
- Unité Micro et Nanomédecines Biomimétiques (MINT), Université d'Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire.,Université d'Angers, CHU, Pôle Hippocrate, Pharmacie
| | - Thierry Urban
- Unité Micro et Nanomédecines Biomimétiques (MINT), Université d'Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire.,Université d'Angers, CHU, Pôle Hippocrate, Service de Pneumologie
| |
Collapse
|
15
|
Mortzfeld BM, Bosch TCG. Eco-Aging: stem cells and microbes are controlled by aging antagonist FoxO. Curr Opin Microbiol 2017; 38:181-187. [DOI: 10.1016/j.mib.2017.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 01/10/2023]
|
16
|
Galloway-Peña JR, Jenq RR, Shelburne SA. Can Consideration of the Microbiome Improve Antimicrobial Utilization and Treatment Outcomes in the Oncology Patient? Clin Cancer Res 2017; 23:3263-3268. [PMID: 28298544 PMCID: PMC5496798 DOI: 10.1158/1078-0432.ccr-16-3173] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/25/2017] [Accepted: 03/10/2017] [Indexed: 12/24/2022]
Abstract
The need to provide effective and timely antimicrobial treatment to cancer patients with infections is well recognized but tempered by preliminary, but accumulating, evidence that antibiotic-induced microbiome dysbiosis affects cancer therapy response, noninfectious toxicities, and infectious complications. Given only a minority of empirically treated cancer patients are proven to have a true bacterial infection, it is important to consider the potential negative consequences of extensive broad-spectrum antimicrobial use on the commensal microbiota. Herein, we review the literature substantiating the dilemma oncologists face when treating suspected or documented infections with respect to the interaction between the host microbiome, antibiotics, and cancer-related clinical outcomes. We propose microbiome-based explorations that could assist oncologists in optimizing treatment strategies for cancer-related infections as well as the cancer itself. In addition, we discuss knowledge gaps and challenges in this nascent field that must be addressed to deliver medically relevant, translational applications. We anticipate that the emerging knowledge regarding the role of the microbiota in the health of cancer patients may cause a reappraisal of the manner in which antibiotics are used in the oncologic setting and how microorganisms are viewed by oncologists. Clin Cancer Res; 23(13); 3263-8. ©2017 AACRSee related commentary by Fessler and Gajewski, p. 3229.
Collapse
Affiliation(s)
- Jessica R Galloway-Peña
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Robert R Jenq
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samuel A Shelburne
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
17
|
Rodríguez-Díaz J, García-Mantrana I, Vila-Vicent S, Gozalbo-Rovira R, Buesa J, Monedero V, Collado MC. Relevance of secretor status genotype and microbiota composition in susceptibility to rotavirus and norovirus infections in humans. Sci Rep 2017; 7:45559. [PMID: 28358023 PMCID: PMC5372083 DOI: 10.1038/srep45559] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/28/2017] [Indexed: 02/07/2023] Open
Abstract
Host genetic factors, such as histo-blood group antigens (HBGAs), are associated with susceptibility to norovirus (NoV) and rotavirus (RV) infections. Recent advances point to the gut microbiome as a key player necessary for a viral pathogen to cause infection. In vitro NoV attachment to host cells and resulting infections have been linked to interactions with certain bacterial types in the gut microbiota. We investigated the relationship between host genotype, gut microbiota, and viral infections. Saliva and fecal samples from 35 adult volunteers were analysed for secretor status genotype, the gut microbiota composition by 16S rRNA gene sequencing, and salivary IgA titers to NoV and RV. Higher levels of IgA against NoV and RV were related to secretor-positive status. No significant differences were found between the FUT2 genotype groups, although the multivariate analysis showed a significant impact of host genotype on specific viral susceptibilities in the microbiome composition. A specific link was found between the abundance of certain bacterial groups, such as Faecalibacterium and Ruminococcus spp., and lower IgA titers against NoV and RV. As a conclusion, we can state that there is a link between host genetics, gut microbiota, and susceptibility to viral infections in humans.
Collapse
Affiliation(s)
- Jesús Rodríguez-Díaz
- Department of Microbiology, School of Medicine, University of Valencia, Valencia, Spain
| | - Izaskun García-Mantrana
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Susana Vila-Vicent
- Department of Microbiology, School of Medicine, University of Valencia, Valencia, Spain
| | | | - Javier Buesa
- Department of Microbiology, School of Medicine, University of Valencia, Valencia, Spain
| | - Vicente Monedero
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| |
Collapse
|
18
|
Wallace TC, Marzorati M, Spence L, Weaver CM, Williamson PS. New Frontiers in Fibers: Innovative and Emerging Research on the Gut Microbiome and Bone Health. J Am Coll Nutr 2017; 36:218-222. [DOI: 10.1080/07315724.2016.1257961] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Taylor C. Wallace
- Department of Nutrition and Food Studies, George Mason University, National Osteoporosis Foundation, Think Healthy Group, LLC, Fairfax, Virginia
| | | | | | - Connie M. Weaver
- Department of Nutrition Science, Women's Global Health Institute, Purdue University, West Lafayette, Indiana
| | | |
Collapse
|
19
|
Commensal gut bacteria modulate phosphorylation-dependent PPARγ transcriptional activity in human intestinal epithelial cells. Sci Rep 2017; 7:43199. [PMID: 28266623 PMCID: PMC5339702 DOI: 10.1038/srep43199] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 01/20/2017] [Indexed: 02/08/2023] Open
Abstract
In healthy subjects, the intestinal microbiota interacts with the host’s epithelium, regulating gene expression to the benefit of both, host and microbiota. The underlying mechanisms remain poorly understood, however. Although many gut bacteria are not yet cultured, constantly growing culture collections have been established. We selected 57 representative commensal bacterial strains to study bacteria-host interactions, focusing on PPARγ, a key nuclear receptor in colonocytes linking metabolism and inflammation to the microbiota. Conditioned media (CM) were harvested from anaerobic cultures and assessed for their ability to modulate PPARγ using a reporter cell line. Activation of PPARγ transcriptional activity was linked to the presence of butyrate and propionate, two of the main metabolites of intestinal bacteria. Interestingly, some stimulatory CMs were devoid of these metabolites. A Prevotella and an Atopobium strain were chosen for further study, and shown to up-regulate two PPARγ-target genes, ANGPTL4 and ADRP. The molecular mechanisms of these activations involved the phosphorylation of PPARγ through ERK1/2. The responsible metabolites were shown to be heat sensitive but markedly diverged in size, emphasizing the diversity of bioactive compounds found in the intestine. Here we describe different mechanisms by which single intestinal bacteria can directly impact their host’s health through transcriptional regulation.
Collapse
|
20
|
Romieu I, Dossus L, Barquera S, Blottière HM, Franks PW, Gunter M, Hwalla N, Hursting SD, Leitzmann M, Margetts B, Nishida C, Potischman N, Seidell J, Stepien M, Wang Y, Westerterp K, Winichagoon P, Wiseman M, Willett WC. Energy balance and obesity: what are the main drivers? Cancer Causes Control 2017; 28:247-258. [PMID: 28210884 PMCID: PMC5325830 DOI: 10.1007/s10552-017-0869-z] [Citation(s) in RCA: 423] [Impact Index Per Article: 52.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 02/06/2017] [Indexed: 12/19/2022]
Abstract
PURPOSE The aim of this paper is to review the evidence of the association between energy balance and obesity. METHODS In December 2015, the International Agency for Research on Cancer (IARC), Lyon, France convened a Working Group of international experts to review the evidence regarding energy balance and obesity, with a focus on Low and Middle Income Countries (LMIC). RESULTS The global epidemic of obesity and the double burden, in LMICs, of malnutrition (coexistence of undernutrition and overnutrition) are both related to poor quality diet and unbalanced energy intake. Dietary patterns consistent with a traditional Mediterranean diet and other measures of diet quality can contribute to long-term weight control. Limiting consumption of sugar-sweetened beverages has a particularly important role in weight control. Genetic factors alone cannot explain the global epidemic of obesity. However, genetic, epigenetic factors and the microbiota could influence individual responses to diet and physical activity. CONCLUSION Energy intake that exceeds energy expenditure is the main driver of weight gain. The quality of the diet may exert its effect on energy balance through complex hormonal and neurological pathways that influence satiety and possibly through other mechanisms. The food environment, marketing of unhealthy foods and urbanization, and reduction in sedentary behaviors and physical activity play important roles. Most of the evidence comes from High Income Countries and more research is needed in LMICs.
Collapse
Affiliation(s)
- Isabelle Romieu
- Nutrition and Metabolism Section, International Agency for Research on Cancer, 150 cours Albert Thomas, 69372, Lyon Cedex 08, France.
| | - Laure Dossus
- Nutrition and Metabolism Section, International Agency for Research on Cancer, 150 cours Albert Thomas, 69372, Lyon Cedex 08, France
| | - Simón Barquera
- Centro de Investigación en Nutrición y Salud, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Hervé M Blottière
- Micalis Institute, MGP MetagenoPolis, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Paul W Franks
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, CRC, University hospital Malmö, Malmö, Sweden
| | - Marc Gunter
- Nutrition and Metabolism Section, International Agency for Research on Cancer, 150 cours Albert Thomas, 69372, Lyon Cedex 08, France
| | - Nahla Hwalla
- Faculty of Agricultural and Food Science, American University of Beirut, Beirut, Lebanon
| | - Stephen D Hursting
- Department of Nutrition and the Nutrition Research Institute, The University of North Carolina, Chapel Hill, USA
| | - Michael Leitzmann
- Department of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Barrie Margetts
- Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, UK
| | - Chizuru Nishida
- Nutrition Policy and Scientific Advice (NPU), Department of Nutrition for Health and Development (NHD), World Health Organization (WHO), Geneva, Switzerland
| | - Nancy Potischman
- Office of the Associate Director, Applied Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, USA
| | - Jacob Seidell
- Faculty of Earth and Life Sciences, Department of Health Sciences, University Amsterdam, Amsterdam, The Netherlands
| | - Magdalena Stepien
- Nutrition and Metabolism Section, International Agency for Research on Cancer, 150 cours Albert Thomas, 69372, Lyon Cedex 08, France
| | - Youfa Wang
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, Joint Appointments, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, USA
| | - Klaas Westerterp
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | | | | | - Walter C Willett
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, USA
| |
Collapse
|
21
|
Pandin C, Le Coq D, Canette A, Aymerich S, Briandet R. Should the biofilm mode of life be taken into consideration for microbial biocontrol agents? Microb Biotechnol 2017; 10:719-734. [PMID: 28205337 PMCID: PMC5481536 DOI: 10.1111/1751-7915.12693] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 01/19/2017] [Accepted: 01/20/2017] [Indexed: 11/26/2022] Open
Abstract
Almost one‐third of crop yields are lost every year due to microbial alterations and diseases. The main control strategy to limit these losses is the use of an array of chemicals active against spoilage and unwanted pathogenic microorganisms. Their massive use has led to extensive environmental pollution, human poisoning and a variety of diseases. An emerging alternative to this chemical approach is the use of microbial biocontrol agents. Biopesticides have been used with success in several fields, but a better understanding of their mode of action is necessary to better control their activity and increase their use. Very few studies have considered that biofilms are the preferred mode of life of microorganisms in the target agricultural biotopes. Increasing evidence shows that the spatial organization of microbial communities on crop surfaces may drive important bioprotection mechanisms. The aim of this review is to summarize the evidence of biofilm formation by biocontrol agents on crops and discuss how this surface‐associated mode of life may influence their biology and interactions with other microorganisms and the host and, finally, their overall beneficial activity.
Collapse
Affiliation(s)
- Caroline Pandin
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Dominique Le Coq
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France.,Micalis Institute, INRA, AgroParisTech, CNRS, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Alexis Canette
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Stéphane Aymerich
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Romain Briandet
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| |
Collapse
|
22
|
Blottière HM, Doré J. Impact des nouveaux outils de métagénomique sur notre connaissance du microbiote intestinal et de son rôle en santé humaine. Med Sci (Paris) 2016; 32:944-951. [DOI: 10.1051/medsci/20163211009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
23
|
Tilg H, Cani PD, Mayer EA. Gut microbiome and liver diseases. Gut 2016; 65:2035-2044. [PMID: 27802157 DOI: 10.1136/gutjnl-2016-312729] [Citation(s) in RCA: 345] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/13/2016] [Accepted: 09/16/2016] [Indexed: 12/20/2022]
Abstract
The gut microbiota has recently evolved as a new important player in the pathophysiology of many intestinal and extraintestinal diseases. The liver is the organ which is in closest contact with the intestinal tract, and is exposed to a substantial amount of bacterial components and metabolites. Various liver disorders such as alcoholic liver disease, non-alcoholic liver disease and primary sclerosing cholangitis have been associated with an altered microbiome. This dysbiosis may influence the degree of hepatic steatosis, inflammation and fibrosis through multiple interactions with the host's immune system and other cell types. Whereas few results from clinical metagenomic studies in liver disease are available, evidence is accumulating that in liver cirrhosis an oral microbiome is overrepresented in the lower intestinal tract, potentially contributing to disease process and severity. A major role for the gut microbiota in liver disorders is also supported by the accumulating evidence that several complications of severe liver disease such as hepatic encephalopathy are efficiently treated by various prebiotics, probiotics and antibiotics. A better understanding of the gut microbiota and its components in liver diseases might provide a more complete picture of these complex disorders and also form the basis for novel therapies.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology & Endocrinology, Medical University Innsbruck, Innsbruck, Austria
| | - Patrice D Cani
- WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Université catholique de Louvain, Brussels, Belgium
| | - Emeran A Mayer
- Division of Digestive Diseases, G. Oppenheimer Center for Neurobiology of Stress and Resilience, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
24
|
Őrfi E, Szebeni J. The immune system of the gut and potential adverse effects of oral nanocarriers on its function. Adv Drug Deliv Rev 2016; 106:402-409. [PMID: 27693367 DOI: 10.1016/j.addr.2016.09.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is substantial effort in modern pharmacotherapy to use nanoparticle-based drug delivery systems (nDDS) for improving the oral absorption of drugs. An often neglected circumstance regarding this approach is that the gut is a major part of the immune system that may be vulnerable for immune-cell toxicity, or mediate humoral immune response against various components of nDDS, recognized as foreign. This review recapitulates the structure and function of gut-associated lymphoid tissue (GALT), i.e., the enteral section of mucosa-associated lymphoid tissue (MALT) and reminds how virus-like nDDS may potentially induce immunogenicity just as attenuated or killed viruses do in oral vaccines. Furthermore, we present examples for immune toxicities of emulsifiers and polymer-containing micelles, manifested in complement activation-related pseudoallergy (CARPA). A major message of the review is that early testing of immunogenicity or other adverse immune effects of nDDS in appropriate test systems or models may be prudent to recognize the risk of rare immune problems that may surface in late-stage clinical trials or after marketing of nDDS.
Collapse
Affiliation(s)
- Erik Őrfi
- Nanomedicine Research and Education Center, Department of Pathophysiology, Semmelweis University, and SeroScience Ltd, Budapest, Hungary
| | - János Szebeni
- Nanomedicine Research and Education Center, Department of Pathophysiology, Semmelweis University, and SeroScience Ltd, Budapest, Hungary; Department of Nanobiotechnology and Regenerative Medicine, Faculty of Health, Miskolc University, Miskolc, Hungary; SeroScience Ltd., Budapest, Hungary.
| |
Collapse
|
25
|
Abstract
During pathogenesis, viruses come in contact with the microbiota that colonizes the mucosal sites they infect. The intestinal microbiota has emerged as a critical factor in intestinal viral susceptibility. While the interaction of virus-intestinal commensal bacteria can lead to enhanced or decreased viral infection capacity, several scientific studies support the use of probiotics as antiviral therapies. Thus, probiotics and the modulation of the intestinal microbiota are envisaged as therapeutic strategies in the prevention and treatment of viral infection.
Collapse
|
26
|
Minard G, Tran FH, Van VT, Goubert C, Bellet C, Lambert G, Kim KLH, Thuy THT, Mavingui P, Valiente Moro C. French invasive Asian tiger mosquito populations harbor reduced bacterial microbiota and genetic diversity compared to Vietnamese autochthonous relatives. Front Microbiol 2015; 6:970. [PMID: 26441903 PMCID: PMC4585046 DOI: 10.3389/fmicb.2015.00970] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/01/2015] [Indexed: 01/16/2023] Open
Abstract
The Asian tiger mosquito Aedes albopictus is one of the most significant pathogen vectors of the twenty-first century. Originating from Asia, it has invaded a wide range of eco-climatic regions worldwide. The insect-associated microbiota is now recognized to play a significant role in host biology. While genetic diversity bottlenecks are known to result from biological invasions, the resulting shifts in host-associated microbiota diversity has not been thoroughly investigated. To address this subject, we compared four autochthonous Ae. albopictus populations in Vietnam, the native area of Ae. albopictus, and three populations recently introduced to Metropolitan France, with the aim of documenting whether these populations display differences in host genotype and bacterial microbiota. Population-level genetic diversity (microsatellite markers and COI haplotype) and bacterial diversity (16S rDNA metabarcoding) were compared between field-caught mosquitoes. Bacterial microbiota from the whole insect bodies were largely dominated by Wolbachia pipientis. Targeted analysis of the gut microbiota revealed a greater bacterial diversity in which a fraction was common between French and Vietnamese populations. The genus Dysgonomonas was the most prevalent and abundant across all studied populations. Overall genetic diversities of both hosts and bacterial microbiota were significantly reduced in recently established populations of France compared to the autochthonous populations of Vietnam. These results open up many important avenues of investigation in order to link the process of geographical invasion to shifts in commensal and symbiotic microbiome communities, as such shifts may have dramatic impacts on the biology and/or vector competence of invading hematophagous insects.
Collapse
Affiliation(s)
- G Minard
- Ecologie Microbienne, UMR Centre National de la Recherche Scientifique 5557, USC INRA 1364, VetAgro Sup, FR41 BioEnvironment and Health, Université Claude Bernard Lyon 1 Villeurbanne, France
| | - F H Tran
- Ecologie Microbienne, UMR Centre National de la Recherche Scientifique 5557, USC INRA 1364, VetAgro Sup, FR41 BioEnvironment and Health, Université Claude Bernard Lyon 1 Villeurbanne, France
| | - Van Tran Van
- Ecologie Microbienne, UMR Centre National de la Recherche Scientifique 5557, USC INRA 1364, VetAgro Sup, FR41 BioEnvironment and Health, Université Claude Bernard Lyon 1 Villeurbanne, France
| | - C Goubert
- Laboratoire de Biométrie et Biologie Evolutive, UMR 5558, CNRS, INRIA, VetAgro Sup Villeurbanne, France
| | - C Bellet
- Entente Interdépartementale Rhône-Alpes pour la Démoustication Chindrieux, France
| | - G Lambert
- Entente Interdépartementale de Démoustication du Littoral Méditerranéen Montpellier, France
| | - Khanh Ly Huynh Kim
- Department of Medical Entomology and Zoonotics, Pasteur Institute in Ho Chi Minh City Vietnam
| | - Trang Huynh Thi Thuy
- Department of Medical Entomology and Zoonotics, Pasteur Institute in Ho Chi Minh City Vietnam
| | - P Mavingui
- Ecologie Microbienne, UMR Centre National de la Recherche Scientifique 5557, USC INRA 1364, VetAgro Sup, FR41 BioEnvironment and Health, Université Claude Bernard Lyon 1 Villeurbanne, France ; Université de La Réunion, UMR PIMIT, INSERM U1187, CNRS 9192, IRD 249, Plateforme Technologique CYROI Saint-Denis, France
| | - C Valiente Moro
- Ecologie Microbienne, UMR Centre National de la Recherche Scientifique 5557, USC INRA 1364, VetAgro Sup, FR41 BioEnvironment and Health, Université Claude Bernard Lyon 1 Villeurbanne, France
| |
Collapse
|
27
|
Cani PD, Everard A. Talking microbes: When gut bacteria interact with diet and host organs. Mol Nutr Food Res 2015; 60:58-66. [PMID: 26178924 PMCID: PMC5014210 DOI: 10.1002/mnfr.201500406] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/03/2015] [Accepted: 07/07/2015] [Indexed: 12/12/2022]
Abstract
Obesity and diabetes have reached epidemic proportions. Evidence suggests that besides dietary habits and physical activity, other environmental factors, such as gut microbes, are recognized as additional partners implicated in the control of energy homeostasis. Studies on the human gut microbiota have shown that the general population can be stratified on the sole basis of three dominant bacteria (i.e., the concept of enterotypes), while some others have suggested categorizing the population according to their microbiome gene richness. Both aspects have been strengthened by recent studies investigating the impact of nutrients (e.g., dietary fibers, fat feeding) and dietary habits (i.e., vegans versus omnivores) of different populations. Using preclinical models, quite a few novel mechanisms have been proposed in these gut microbiota–host interactions, including the role of novel bacteria, the regulation of antimicrobial peptide production, the maintenance of the gut barrier function and intestinal innate immunity. In this review, we discuss several of the aforementioned aspects. Nonetheless, determining the overall mechanisms by which microbes dialogue with host cells will require further investigations before anticipating the development of next‐generation nutritional interventions using prebiotics, probiotics, synbiotics, or even specific nutrients for promoting health benefits.
Collapse
Affiliation(s)
- Patrice D Cani
- Metabolism and Nutrition Research Group, WELBIO- Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Amandine Everard
- Metabolism and Nutrition Research Group, WELBIO- Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
28
|
Dobbs SM, Dobbs RJ, Weller C, Charlett A, Augustin A, Taylor D, Ibrahim MAA, Bjarnason I. Peripheral aetiopathogenic drivers and mediators of Parkinson's disease and co-morbidities: role of gastrointestinal microbiota. J Neurovirol 2015; 22:22-32. [PMID: 26092111 PMCID: PMC4729788 DOI: 10.1007/s13365-015-0357-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/17/2015] [Accepted: 05/21/2015] [Indexed: 12/26/2022]
Abstract
We seek an aetiopathogenic model for the spectrum of Parkinson's disease (PD), functional bowel disease, depression and cognitive impairment. The adopted concept is that systemic immuno-inflammatory processes mediate neuro-inflammation. The model would be based on phenotype, exposome (including gastrointestinal microbiome), milieu (immuno-inflammatory and metabolome), human genetics and their interactions. It would enable a patient's position, to be understood in terms of drivers, perpetuators and mediators, and a future position, with and without intervention, predicted. Even the cardinal facets of PD may have different drivers: halting one may allow escape down subordinate pathways. Peptic ulceration is prodromal to PD. In our randomised placebo-controlled trial, hypokinesia improved over the year following biopsy-proven Helicobacter pylori eradication and rigidity worsened. This was independent of any (stable, long t½) antiparkinsonian medication. There are pointers to an autoimmune process: for example, surveillance-confirmed hypokinesia effect was indication specific. During surveillance, successive antimicrobial courses, other than for Helicobacter, were associated with cumulative increase in rigidity. Exhibiting laxatives appeared to stem the overall temporal increase, despite antiparkinsonian medication, in rigidity. Thus, intestinal dysbiosis may be a major source of bystander neuronal damage. There are biological gradients of objective measures of PD facets on circulating inflammatory markers and leucocyte subset counts. Moreover, lactulose hydrogen breath test positivity for small-intestinal bacterial overgrowth (present in two thirds of PD patients) is associated with the same subsets: higher natural killer and total CD4+ counts and lower neutrophils. With greater aetiopathogenic understanding, relatively low cost and on-the-shelf medication could have a major impact. A new generation of animal models, based on the gut microbiome, is envisaged.
Collapse
Affiliation(s)
- Sylvia M Dobbs
- Pharmaceutical Sciences, King's College London, London, UK. .,The Maudsley Hospital, London, UK. .,Department of Gastroenterology, King's College Hospital, London, UK.
| | - R John Dobbs
- Pharmaceutical Sciences, King's College London, London, UK.,The Maudsley Hospital, London, UK.,Department of Gastroenterology, King's College Hospital, London, UK
| | - Clive Weller
- Pharmaceutical Sciences, King's College London, London, UK
| | - André Charlett
- Pharmaceutical Sciences, King's College London, London, UK.,Statistics Unit, National Infection Service, Public Health England, London, UK
| | - Aisha Augustin
- Pharmaceutical Sciences, King's College London, London, UK.,The Maudsley Hospital, London, UK
| | - David Taylor
- Pharmaceutical Sciences, King's College London, London, UK.,The Maudsley Hospital, London, UK
| | - Mohammad A A Ibrahim
- Diagnostic Immunology Laboratory, King's College and St Thomas's Hospitals, London, UK
| | - Ingvar Bjarnason
- Department of Gastroenterology, King's College Hospital, London, UK
| |
Collapse
|
29
|
Marzorati M, Qin B, Hildebrand F, Klosterbuer A, Roughead Z, Roessle C, Rochat F, Raes J, Possemiers S. Addition of acacia gum to a FOS/inulin blend improves its fermentation profile in the Simulator of the Human Intestinal Microbial Ecosystem (SHIME®). J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.04.039] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
30
|
Standardised animal models of host microbial mutualism. Mucosal Immunol 2015; 8:476-86. [PMID: 25492472 PMCID: PMC4424382 DOI: 10.1038/mi.2014.113] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 10/09/2014] [Indexed: 02/04/2023]
Abstract
An appreciation of the importance of interactions between microbes and multicellular organisms is currently driving research in biology and biomedicine. Many human diseases involve interactions between the host and the microbiota, so investigating the mechanisms involved is important for human health. Although microbial ecology measurements capture considerable diversity of the communities between individuals, this diversity is highly problematic for reproducible experimental animal models that seek to establish the mechanistic basis for interactions within the overall host-microbial superorganism. Conflicting experimental results may be explained away through unknown differences in the microbiota composition between vivaria or between the microenvironment of different isolated cages. In this position paper, we propose standardised criteria for stabilised and defined experimental animal microbiotas to generate reproducible models of human disease that are suitable for systematic experimentation and are reproducible across different institutions.
Collapse
|
31
|
de Souza AZZ, Zambom AZ, Abboud KY, Reis SK, Tannihão F, Guadagnini D, Saad MJA, Prada PO. Oral supplementation with L-glutamine alters gut microbiota of obese and overweight adults: A pilot study. Nutrition 2015; 31:884-9. [PMID: 25933498 DOI: 10.1016/j.nut.2015.01.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 01/09/2015] [Accepted: 01/13/2015] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The aim of this study was to determine whether oral supplementation with L-glutamine (GLN) modifies the gut microbiota composition in overweight and obese adults. METHODS Thirty-three overweight and obese adults, ages between 23 and 59 y and body mass index between 25.03 and 47.12 kg/m(2), were randomly assigned to receive either oral supplementation with 30 g of L-alanine (ALA group control) or 30 g of GLN (GLN group) daily for 14 d. We analyzed the gut microbiota composition with new-generation sequencing techniques and bioinformatics analysis. RESULTS After 14 d of supplementation, adults in the GLN group exhibited statistically significant differences in the Firmicutes and Actinobacteria phyla compared with those in the ALA group. The ratio of Firmicutes to Bacteroidetes, a good biomarker for obesity, decreased in the GLN group from 0.85 to 0.57, whereas it increased from 0.91 to 1.12 in the ALA group. At the genus level, Dialister, Dorea, Pseudobutyrivibrio, and Veillonella, belonging to the Firmicutes phylum, had statistically significant reduction. CONCLUSION Oral supplementation with GLN, for a short time, altered the composition of the gut microbiota in overweight and obese humans reducing the Firmicutes to Bacteroidetes ratio, which resembled weight loss programs already seen in the literature.
Collapse
Affiliation(s)
| | - Adriano Zanin Zambom
- Department of Statistics, State University of Campinas, Campinas, São Paulo, Brazil
| | | | - Sabrina Karen Reis
- School of Applied Sciences, State University of Campinas, Campinas, São Paulo, Brazil
| | - Fabiana Tannihão
- School of Applied Sciences, State University of Campinas, Campinas, São Paulo, Brazil
| | - Dioze Guadagnini
- Department of Internal Medicine, State University of Campinas, Campinas, São Paulo, Brazil
| | - Mario J A Saad
- Department of Internal Medicine, State University of Campinas, Campinas, São Paulo, Brazil
| | - Patricia Oliveira Prada
- School of Applied Sciences, State University of Campinas, Campinas, São Paulo, Brazil; Department of Internal Medicine, State University of Campinas, Campinas, São Paulo, Brazil.
| |
Collapse
|
32
|
Wang WL, Xu SY, Ren ZG, Tao L, Jiang JW, Zheng SS. Application of metagenomics in the human gut microbiome. World J Gastroenterol 2015; 21:803-814. [PMID: 25624713 PMCID: PMC4299332 DOI: 10.3748/wjg.v21.i3.803] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/30/2014] [Accepted: 11/07/2014] [Indexed: 02/06/2023] Open
Abstract
There are more than 1000 microbial species living in the complex human intestine. The gut microbial community plays an important role in protecting the host against pathogenic microbes, modulating immunity, regulating metabolic processes, and is even regarded as an endocrine organ. However, traditional culture methods are very limited for identifying microbes. With the application of molecular biologic technology in the field of the intestinal microbiome, especially metagenomic sequencing of the next-generation sequencing technology, progress has been made in the study of the human intestinal microbiome. Metagenomics can be used to study intestinal microbiome diversity and dysbiosis, as well as its relationship to health and disease. Moreover, functional metagenomics can identify novel functional genes, microbial pathways, antibiotic resistance genes, functional dysbiosis of the intestinal microbiome, and determine interactions and co-evolution between microbiota and host, though there are still some limitations. Metatranscriptomics, metaproteomics and metabolomics represent enormous complements to the understanding of the human gut microbiome. This review aims to demonstrate that metagenomics can be a powerful tool in studying the human gut microbiome with encouraging prospects. The limitations of metagenomics to be overcome are also discussed. Metatranscriptomics, metaproteomics and metabolomics in relation to the study of the human gut microbiome are also briefly discussed.
Collapse
|
33
|
Doré J, Blottière H. The influence of diet on the gut microbiota and its consequences for health. Curr Opin Biotechnol 2015; 32:195-199. [PMID: 25615931 DOI: 10.1016/j.copbio.2015.01.002] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 12/29/2014] [Accepted: 01/03/2015] [Indexed: 02/07/2023]
Abstract
Man is an intimate symbiosis between 10 trillion human cells and some 100 trillion bacteria, most of which inhabit the intestine where they constitute an extremely dense and diverse microbiota. This symbiotic balance that has to be established within each newborn is key to the maintenance of health and well being. Its development is markedly influenced by microbial exposure encountered very early in life. Mode of infant feeding, and the post-weaning transition to habitual diet will further shape the microbiota. Recent studies support the concept that diet should be viewed as a means to prevent potentially durable alterations of symbiosis observed in immune-mediated metabolic and inflammatory diseases. Non-digestible dietary fiber will play a major role in this context.
Collapse
Affiliation(s)
- Joël Doré
- INRA, Micalis & MetaGenoPolis, Jouy-en-Josas, France.
| | | |
Collapse
|
34
|
Soueidan H, Schmitt LA, Candresse T, Nikolski M. Finding and identifying the viral needle in the metagenomic haystack: trends and challenges. Front Microbiol 2015; 5:739. [PMID: 25610431 PMCID: PMC4285800 DOI: 10.3389/fmicb.2014.00739] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 12/05/2014] [Indexed: 11/13/2022] Open
Abstract
Collectively, viruses have the greatest genetic diversity on Earth, occupy extremely varied niches and are likely able to infect all living organisms. Viral infections are an important issue for human health and cause considerable economic losses when agriculturally important crops or husbandry animals are infected. The advent of metagenomics has provided a precious tool to study viruses by sampling them in natural environments and identifying the genomic composition of a sample. However, reaching a clear recognition and taxonomic assignment of the identified viruses has been hampered by the computational difficulty of these problems. In this perspective paper we examine the trends in current research for the identification of viral sequences in a metagenomic sample, pinpoint the intrinsic computational difficulties for the identification of novel viral sequences within metagenomic samples, and suggest possible avenues to overcome them.
Collapse
Affiliation(s)
- Hayssam Soueidan
- Bordeaux Bioinformatics Center, Université de Bordeaux Bordeaux, France ; INSERM U1035, Université de Bordeaux Bordeaux, France
| | - Louise-Amélie Schmitt
- Bordeaux Bioinformatics Center, Université de Bordeaux Bordeaux, France ; Centre National de la Recherche Scientifique/Laboratoire Bordelais de Recherche en Informatique, Université de Bordeaux Talence, France
| | - Thierry Candresse
- Institut National de la Recherche Agronomique, UMR 1332 Biologie du Fruit et Pathologie Villenave d'Ornon, France ; UMR 1332 Biologie du Fruit et Pathologie, Université de Bordeaux Villenave d'Ornon, France
| | - Macha Nikolski
- Bordeaux Bioinformatics Center, Université de Bordeaux Bordeaux, France ; Centre National de la Recherche Scientifique/Laboratoire Bordelais de Recherche en Informatique, Université de Bordeaux Talence, France
| |
Collapse
|
35
|
Minard G, Tran FH, Van VT, Goubert C, Bellet C, Lambert G, Kim KLH, Thuy THT, Mavingui P, Valiente Moro C. French invasive Asian tiger mosquito populations harbor reduced bacterial microbiota and genetic diversity compared to Vietnamese autochthonous relatives. Front Microbiol 2015; 6:970. [PMID: 26441903 DOI: 10.3389/fmicb.2015.00970/abstract] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/01/2015] [Indexed: 05/19/2023] Open
Abstract
The Asian tiger mosquito Aedes albopictus is one of the most significant pathogen vectors of the twenty-first century. Originating from Asia, it has invaded a wide range of eco-climatic regions worldwide. The insect-associated microbiota is now recognized to play a significant role in host biology. While genetic diversity bottlenecks are known to result from biological invasions, the resulting shifts in host-associated microbiota diversity has not been thoroughly investigated. To address this subject, we compared four autochthonous Ae. albopictus populations in Vietnam, the native area of Ae. albopictus, and three populations recently introduced to Metropolitan France, with the aim of documenting whether these populations display differences in host genotype and bacterial microbiota. Population-level genetic diversity (microsatellite markers and COI haplotype) and bacterial diversity (16S rDNA metabarcoding) were compared between field-caught mosquitoes. Bacterial microbiota from the whole insect bodies were largely dominated by Wolbachia pipientis. Targeted analysis of the gut microbiota revealed a greater bacterial diversity in which a fraction was common between French and Vietnamese populations. The genus Dysgonomonas was the most prevalent and abundant across all studied populations. Overall genetic diversities of both hosts and bacterial microbiota were significantly reduced in recently established populations of France compared to the autochthonous populations of Vietnam. These results open up many important avenues of investigation in order to link the process of geographical invasion to shifts in commensal and symbiotic microbiome communities, as such shifts may have dramatic impacts on the biology and/or vector competence of invading hematophagous insects.
Collapse
Affiliation(s)
- G Minard
- Ecologie Microbienne, UMR Centre National de la Recherche Scientifique 5557, USC INRA 1364, VetAgro Sup, FR41 BioEnvironment and Health, Université Claude Bernard Lyon 1 Villeurbanne, France
| | - F H Tran
- Ecologie Microbienne, UMR Centre National de la Recherche Scientifique 5557, USC INRA 1364, VetAgro Sup, FR41 BioEnvironment and Health, Université Claude Bernard Lyon 1 Villeurbanne, France
| | - Van Tran Van
- Ecologie Microbienne, UMR Centre National de la Recherche Scientifique 5557, USC INRA 1364, VetAgro Sup, FR41 BioEnvironment and Health, Université Claude Bernard Lyon 1 Villeurbanne, France
| | - C Goubert
- Laboratoire de Biométrie et Biologie Evolutive, UMR 5558, CNRS, INRIA, VetAgro Sup Villeurbanne, France
| | - C Bellet
- Entente Interdépartementale Rhône-Alpes pour la Démoustication Chindrieux, France
| | - G Lambert
- Entente Interdépartementale de Démoustication du Littoral Méditerranéen Montpellier, France
| | - Khanh Ly Huynh Kim
- Department of Medical Entomology and Zoonotics, Pasteur Institute in Ho Chi Minh City Vietnam
| | - Trang Huynh Thi Thuy
- Department of Medical Entomology and Zoonotics, Pasteur Institute in Ho Chi Minh City Vietnam
| | - P Mavingui
- Ecologie Microbienne, UMR Centre National de la Recherche Scientifique 5557, USC INRA 1364, VetAgro Sup, FR41 BioEnvironment and Health, Université Claude Bernard Lyon 1 Villeurbanne, France ; Université de La Réunion, UMR PIMIT, INSERM U1187, CNRS 9192, IRD 249, Plateforme Technologique CYROI Saint-Denis, France
| | - C Valiente Moro
- Ecologie Microbienne, UMR Centre National de la Recherche Scientifique 5557, USC INRA 1364, VetAgro Sup, FR41 BioEnvironment and Health, Université Claude Bernard Lyon 1 Villeurbanne, France
| |
Collapse
|
36
|
Tralau T, Sowada J, Luch A. Insights on the human microbiome and its xenobiotic metabolism: what is known about its effects on human physiology? Expert Opin Drug Metab Toxicol 2014; 11:411-25. [PMID: 25476418 DOI: 10.1517/17425255.2015.990437] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Our microbiome harbours a metabolic capacity far beyond our own. Moreover, its gene pool is highly adaptable and subject to selective pressure, including host exposure to xenobiotics. Yet, the resulting adaptations do not necessarily follow host well-being and can therefore contribute to disease or unfavourable metabolite production. AREAS COVERED This review provides an overview of our host-microbiome relationship in light of bacterial (xenobiotic) metabolism, community dynamics, entero-endocrine crosstalk, dysbiosis and potential therapeutic targets. In addition, it will highlight the need for a systematic analysis of the microbiome's potential for substance toxification. EXPERT OPINION The influence of our microbiota reaches from primary metabolites to secondary effects such as substrate competition or the activation of eukaryotic Phase I and Phase II enzymes. Further on it plays a hitherto underestimated role in drug metabolism, toxicity and pathogenesis. These effects are partly caused by entero-endocrine crosstalk and interference with eukaryotic regulatory networks. On first sight, the resulting concept of a metabolically competent microbiome adds enormous complexity to human physiology. Yet, the potential specificity of microbial targets harbours therapeutic promise for diseases such as diabetes, cancer and psychiatric disorders. A better physiological and biochemical understanding of the microbiome is thus of high priority for academia and biomedical research.
Collapse
Affiliation(s)
- Tewes Tralau
- German Federal Institute for Risk Assessment (BfR), Department of Chemicals and Product Safety , Max-Dohrn Strasse 8-10, 10589 Berlin , Germany
| | | | | |
Collapse
|
37
|
Abstract
The recent developments of metagenomics permit an extremely high-resolution molecular scan of the intestinal microbiota giving new insights and opening perspectives for clinical applications. Beyond the unprecedented vision of the intestinal microbiota given by large-scale quantitative metagenomics studies, such as the EU MetaHIT project, functional metagenomics tools allow the exploration of fine interactions between food constituents, microbiota and host, leading to the identification of signals and intimate mechanisms of crosstalk, especially between bacteria and human cells. Cloning of large genome fragments, either from complex intestinal communities or from selected bacteria, allows the screening of these biological resources for bioactivity towards complex plant polymers or functional food such as prebiotics. This permitted identification of novel carbohydrate-active enzyme families involved in dietary fibre and host glycan breakdown, and highlighted unsuspected bacterial players at the top of the intestinal microbial food chain. Similarly, exposure of fractions from genomic and metagenomic clones onto human cells engineered with reporter systems to track modulation of immune response, cell proliferation or cell metabolism has allowed the identification of bioactive clones modulating key cell signalling pathways or the induction of specific genes. This opens the possibility to decipher mechanisms by which commensal bacteria or candidate probiotics can modulate the activity of cells in the intestinal epithelium or even in distal organs such as the liver, adipose tissue or the brain. Hence, in spite of our inability to culture many of the dominant microbes of the human intestine, functional metagenomics open a new window for the exploration of food–microbe–host crosstalk.
Collapse
|
38
|
de Wouters T, Ledue F, Nepelska M, Doré J, Blottière HM, Lapaque N. A robust and adaptable high throughput screening method to study host-microbiota interactions in the human intestine. PLoS One 2014; 9:e105598. [PMID: 25141006 PMCID: PMC4139392 DOI: 10.1371/journal.pone.0105598] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/24/2014] [Indexed: 12/21/2022] Open
Abstract
The intestinal microbiota has many beneficial roles for its host. However, the precise mechanisms developed by the microbiota to influence the host intestinal cell responses are only partially known. The complexity of the ecosystem and our inability to culture most of these micro-organisms have led to the development of molecular approaches such as functional metagenomics, i.e. the heterologous expression of a metagenome in order to identify functions. This elegant strategy coupled to high throughput screening allowed to identify novel enzymes from different ecosystems where culture methods have not yet been adapted to isolate the candidate microorganisms. We have proposed to use this functional metagenomic approach in order to model the microbiota's interaction with the host by combining this heterologous expression with intestinal reporter cell lines. The addition of the cellular component to this functional metagenomic approach introduced a second important source of variability resulting in a novel challenge for high throughput screening. First attempts of high throughput screening with various reporter cell-lines showed a high distribution of the response and consequent difficulties to reproduce the response, impairing an easy and clear identification of confirmed hits. In this study, we developed a robust and reproducible methodology to combine these two biological systems for high throughput application. We optimized experimental setups and completed them by appropriate statistical analysis tools allowing the use this innovative approach in a high throughput manner and on a broad range of reporter assays. We herewith present a methodology allowing a high throughput screening combining two biological systems. Therefore ideal conditions for homogeneity, sensitivity and reproducibility of both metagenomic clones as well as reporter cell lines have been identified and validated. We believe that this innovative method will allow the identification of new bioactive microbial molecules and, subsequently, will promote understanding of host-microbiota interactions.
Collapse
Affiliation(s)
- Tomas de Wouters
- INRA, UMR 1319 MICALIS, Domaine de Vilvert, Jouy-en-Josas, France
- AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - Florence Ledue
- INRA, UMR 1319 MICALIS, Domaine de Vilvert, Jouy-en-Josas, France
- AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - Malgorzata Nepelska
- INRA, UMR 1319 MICALIS, Domaine de Vilvert, Jouy-en-Josas, France
- AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - Joël Doré
- INRA, UMR 1319 MICALIS, Domaine de Vilvert, Jouy-en-Josas, France
- AgroParisTech, UMR Micalis, Jouy-en-Josas, France
- INRA, US 1367 MetaGenoPoliS, Jouy-en-Josas, France
| | - Hervé M. Blottière
- INRA, UMR 1319 MICALIS, Domaine de Vilvert, Jouy-en-Josas, France
- AgroParisTech, UMR Micalis, Jouy-en-Josas, France
- INRA, US 1367 MetaGenoPoliS, Jouy-en-Josas, France
| | - Nicolas Lapaque
- INRA, UMR 1319 MICALIS, Domaine de Vilvert, Jouy-en-Josas, France
- AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| |
Collapse
|
39
|
Martín R, Miquel S, Langella P, Bermúdez-Humarán LG. The role of metagenomics in understanding the human microbiome in health and disease. Virulence 2014; 5:413-23. [PMID: 24429972 DOI: 10.4161/viru.27864] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The term microbiome refers to the genetic material of the catalog of microbial taxa associated with humans. As in all ecosystems, the microbiota reaches a dynamic equilibrium in the human body, which can be altered by environmental factors and external stimuli. Metagenomics is a relatively new field of study of microbial genomes within diverse environmental samples, which is of increasing importance in microbiology. The introduction of this ecological perception of microbiology is the key to achieving real knowledge about the influence of the microbiota in human health and disease. The aim of this review is to summarize the link between the human microbiota (focusing on the intestinal, vaginal, skin, and airway body sites) and health from this ecological point of view, highlighting the contribution of metagenomics in the advance of this field.
Collapse
Affiliation(s)
- Rebeca Martín
- INRA; UMR1319 Micalis; Jouy-en-Josas, France; AgroParisTech; UMR Micalis; Jouy-en-Josas, France
| | - Sylvie Miquel
- INRA; UMR1319 Micalis; Jouy-en-Josas, France; AgroParisTech; UMR Micalis; Jouy-en-Josas, France
| | - Philippe Langella
- INRA; UMR1319 Micalis; Jouy-en-Josas, France; AgroParisTech; UMR Micalis; Jouy-en-Josas, France
| | - Luis G Bermúdez-Humarán
- INRA; UMR1319 Micalis; Jouy-en-Josas, France; AgroParisTech; UMR Micalis; Jouy-en-Josas, France
| |
Collapse
|
40
|
Vieira AT, Teixeira MM, Martins FS. The role of probiotics and prebiotics in inducing gut immunity. Front Immunol 2013; 4:445. [PMID: 24376446 PMCID: PMC3859913 DOI: 10.3389/fimmu.2013.00445] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 11/26/2013] [Indexed: 12/13/2022] Open
Abstract
The gut immune system is influenced by many factors, including dietary components and commensal bacteria. Nutrients that affect gut immunity and strategies that restore a healthy gut microbial community by affecting the microbial composition are being developed as new therapeutic approaches to treat several inflammatory diseases. Although probiotics (live microorganisms) and prebiotics (food components) have shown promise as treatments for several diseases in both clinical and animal studies, an understanding of the molecular mechanisms behind the direct and indirect effects on the gut immune response will facilitate better and possibly more efficient therapy for diseases. In this review, we will first describe the concept of prebiotics, probiotics, and symbiotics and cover the most recently well-established scientific findings regarding the direct and indirect mechanisms by which these dietary approaches can influence gut immunity. Emphasis will be placed on the relationship of diet, the microbiota, and the gut immune system. Second, we will highlight recent results from our group, which suggest a new dietary manipulation that includes the use of nutrient products (organic selenium and Lithothamnium muelleri) and probiotics (Saccharomyces boulardii UFMG 905 and Bifidobacterium sp.) that can stimulate and manipulate the gut immune response, inducing intestinal homeostasis. Furthermore, the purpose of this review is to discuss and translate all of this knowledge into therapeutic strategies and into treatment for extra-intestinal compartment pathologies. We will conclude by discussing perspectives and molecular advances regarding the use of prebiotics or probiotics as new therapeutic strategies that manipulate the microbial composition and the gut immune responses of the host.
Collapse
Affiliation(s)
- Angélica T Vieira
- Immunopharmacology Group, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil
| | - Mauro M Teixeira
- Immunopharmacology Group, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil
| | - Flaviano S Martins
- Immunopharmacology Group, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil ; Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais , Belo Horizonte , Brazil
| |
Collapse
|