101
|
Ramirez Garcia A, Zhang J, Greppi A, Constancias F, Wortmann E, Wandres M, Hurley K, Pascual-García A, Ruscheweyh HJ, Sturla SJ, Lacroix C, Schwab C. Impact of manipulation of glycerol/diol dehydratase activity on intestinal microbiota ecology and metabolism. Environ Microbiol 2021; 23:1765-1779. [PMID: 33587772 DOI: 10.1111/1462-2920.15431] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/19/2022]
Abstract
Glycerol/diol dehydratases (GDH) are enzymes that catalyse the production of propionate from 1,2-propanediol, and acrolein from glycerol. Acrolein reacts with dietary carcinogenic heterocyclic amines (HCA), reducing HCA mutagenicity, but is itself also an antimicrobial agent and toxicant. Gut microbial GDH activity has been suggested as an endogenous acrolein source; however, there is limited information on the potential of the intestinal microbiota to have GDH activity, and what impact it can have on the intestinal ecosystem and host health. We hypothesized that GDH activity of gut microbiota is determined by the abundance and distribution of GDH-active taxa and can be enhanced by supplementation of the GDH active Anaerobutyricum hallii, and tested this hypothesis combining quantitative profiling of gdh, model batch fermentations, microbiota manipulation, and kinetic modelling of acrolein formation. Our results suggest that GDH activity is a common trait of intestinal microbiota shared by a few taxa, which was dependent on overall gdh abundance. Anaerobutyricum hallii was identified as a key taxon in GDH metabolism, and its supplementation increased the rate of GDH activity and acrolein release, which enhanced the transformation of HCA and reduced fermentation activity. The findings of this first systematic study on acrolein release by intestinal microbiota indicate that dietary and microbial modulation might impact GDH activity, which may influence host health.
Collapse
Affiliation(s)
- Alejandro Ramirez Garcia
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.,Laboratory of Toxicology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Jianbo Zhang
- Laboratory of Toxicology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anna Greppi
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Florentin Constancias
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Esther Wortmann
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Muriel Wandres
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Katherine Hurley
- Laboratory of Toxicology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | | | - Hans-Joachim Ruscheweyh
- Institute of Microbiology, Department of Biology, and Swiss Institute of Bioinformatics, ETH Zürich, Zürich, Switzerland
| | - Shana J Sturla
- Laboratory of Toxicology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Clarissa Schwab
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland.,Department of Biological and Chemical Engineering, Aarhus University, Aarhus, Denmark
| |
Collapse
|
102
|
Identification of New Potential Biotherapeutics from Human Gut Microbiota-Derived Bacteria. Microorganisms 2021; 9:microorganisms9030565. [PMID: 33803291 PMCID: PMC7998412 DOI: 10.3390/microorganisms9030565] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
The role of the gut microbiota in health and disease is well recognized and the microbiota dysbiosis observed in many chronic diseases became a new therapeutic target. The challenge is to get a better insight into the functionality of commensal bacteria and to use this knowledge to select live biotherapeutics as new preventive or therapeutic products. In this study, we set up a screening approach to evaluate the functional capacities of a set of 21 strains isolated from the gut microbiota of neonates and adults. For this purpose, we selected key biological processes involved in the microbiome-host symbiosis and known to impact the host physiology i.e., the production of short-chain fatty acids and the ability to strengthen an epithelial barrier (Caco-2), to induce the release of the anti-inflammatory IL-10 cytokine after co-culture with human immune cells (PBMC) or to increase GLP-1 production from STC-1 endocrine cell line. This strategy highlighted fifteen strains exhibiting beneficial activities among which seven strains combined several of them. Interestingly, this work revealed for the first time a high prevalence of potential health-promoting functions among intestinal commensal strains and identified several appealing novel candidates for the management of chronic diseases, notably obesity and inflammatory bowel diseases.
Collapse
|
103
|
Morales D, Shetty SA, López-Plaza B, Gómez-Candela C, Smidt H, Marín FR, Soler-Rivas C. Modulation of human intestinal microbiota in a clinical trial by consumption of a β-D-glucan-enriched extract obtained from Lentinula edodes. Eur J Nutr 2021; 60:3249-3265. [PMID: 33580297 DOI: 10.1007/s00394-021-02504-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 01/28/2021] [Indexed: 12/23/2022]
Abstract
PURPOSE The aim of this study was to evaluate the hypocholesterolemic, immune- and microbiota-modulatory effect of a mushroom extract in hypercholesterolemic subjects. METHODS A randomized, controlled, double-blind, and parallel clinical trial was carried out with subjects from 18 to 65 years old (n = 52) with untreated mild hypercholesterolemia. Volunteers consumed a β-D-glucan-enriched (BGE) mixture (10.4 g/day) obtained from shiitake mushrooms (Lentinula edodes) ensuring a 3.5 g/day of fungal β-D-glucans or a placebo incorporated in three different commercial creams. RESULTS This mixture showed hypocholesterolemic activities in vitro and in animal studies. After eight weeks intervention, no significant differences in lipid- or cholesterol-related parameters were found compared to placebo subjects as well as before and after the BGE mixture administration. No inflammatory or immunomodulatory responses were noticed and no changes in IL-1β, IL-6, TNF-α or oxLDL were recorded. However, consumption of the BGE mixture was safe and managed to achieve the dietary fibre intake recommended as cardiovascular protective diet. Moreover, the BGE mixture modulated the colonic microbiota differently compared to placebo. Microbial community composition varied from before to after the intervention with several genera being positively or negatively correlated with some biomarkers related to cholesterol metabolism. CONCLUSION These results suggested a relation between cholesterol metabolism, microbiota and BGE administration. Nevertheless, the precise significance of this differential modulation was not fully elucidated and requires further studies.
Collapse
Affiliation(s)
- Diego Morales
- Department of Production and Characterization of Novel Foods, Institute of Food Science Research - CIAL (UAM+CSIC), Universidad Autónoma de Madrid, C/ Nicolas Cabrera 9, Campus de Cantoblanco, 28049, Madrid, Spain.
| | - Sudarshan A Shetty
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Bricia López-Plaza
- Nutrition Research Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046, Madrid, Spain
| | - Carmen Gómez-Candela
- Nutrition Research Group, Hospital La Paz Institute for Health Research (IdiPAZ), 28046, Madrid, Spain
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Francisco Ramón Marín
- Department of Production and Characterization of Novel Foods, Institute of Food Science Research - CIAL (UAM+CSIC), Universidad Autónoma de Madrid, C/ Nicolas Cabrera 9, Campus de Cantoblanco, 28049, Madrid, Spain
| | - Cristina Soler-Rivas
- Department of Production and Characterization of Novel Foods, Institute of Food Science Research - CIAL (UAM+CSIC), Universidad Autónoma de Madrid, C/ Nicolas Cabrera 9, Campus de Cantoblanco, 28049, Madrid, Spain
| |
Collapse
|
104
|
Wang JM, Yang MX, Wu QF, Chen J, Deng SF, Chen L, Wei DN, Liang FR. Improvement of intestinal flora: accompany with the antihypertensive effect of electroacupuncture on stage 1 hypertension. Chin Med 2021; 16:7. [PMID: 33413552 PMCID: PMC7792359 DOI: 10.1186/s13020-020-00417-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 12/22/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Increasing evidence have indicated the relationship between intestinal dysbiosis and hypertension. We aimed to evaluate the effect of the electroacupuncture (EA) on intestinal microbiota in patients with stage 1 hypertension. METHODS 93 hypertensive patients and 15 healthy subjects were enrolled in this study. Applying a highly accurate oscillometric device to evaluate the antihypertensive effect of EA. 16S rRNA sequencing was used to profile stool microbial communities from Healthy group, Before treatment (BT) group and After treatment (AT) group, and various multivariate analysis approaches were used to assess diversity, composition and abundance of intestinal microbiota. RESULTS In this study, EA significantly decreased the blood pressure (BP) of hypertensive patients. Higher abundance of Firmicutes and lower Bacteroidetes abundance were observed in the BT group compared to the Healthy group. And EA treatment significantly decreased the Firmicutes/Bacteroidetes ratio compared to the BT group. Moreover, at the genus level, there was an increased abundance of Escherichia-Shigella in patients with hypertension, while Blautia were decreased, and EA reversed these changes. CONCLUSIONS Our study indicates that EA can effectively lower BP and improve the structure of intestinal microbiota which are correlate with the alteration of blood pressure by electroacupuncture. TRIAL REGISTRATION Clinicaltrial.gov, NCT01701726. Registered 5 October 2012, https://clinicaltrials.gov/ct2/show/study/NCT01701726.
Collapse
Affiliation(s)
- Jun-Meng Wang
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, No.37, Road Shi-Er-Qiao, Jinniu District, Chengdu, 610075, Sichuan, China
| | - Ming-Xiao Yang
- School of Chinese Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, SAR, China
- Department of Chinese Medicine, The University of Hong Kong-Shenzhen Hospital, Hai yuan Road, Futian District, Shenzhen, 518053, Guangdong, China
| | - Qiao-Feng Wu
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, No.37, Road Shi-Er-Qiao, Jinniu District, Chengdu, 610075, Sichuan, China.
- Institute of Acupuncture and Homeostasis Regulation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China.
| | - Ji Chen
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, No.37, Road Shi-Er-Qiao, Jinniu District, Chengdu, 610075, Sichuan, China
| | - Shu-Fang Deng
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, No.37, Road Shi-Er-Qiao, Jinniu District, Chengdu, 610075, Sichuan, China
| | - Lin Chen
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, No.37, Road Shi-Er-Qiao, Jinniu District, Chengdu, 610075, Sichuan, China
| | - Da-Neng Wei
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, No.37, Road Shi-Er-Qiao, Jinniu District, Chengdu, 610075, Sichuan, China
| | - Fan-Rong Liang
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, No.37, Road Shi-Er-Qiao, Jinniu District, Chengdu, 610075, Sichuan, China.
| |
Collapse
|
105
|
Micó-Carnero M, Rojano-Alfonso C, Álvarez-Mercado AI, Gracia-Sancho J, Casillas-Ramírez A, Peralta C. Effects of Gut Metabolites and Microbiota in Healthy and Marginal Livers Submitted to Surgery. Int J Mol Sci 2020; 22:44. [PMID: 33375200 PMCID: PMC7793124 DOI: 10.3390/ijms22010044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/18/2020] [Accepted: 12/20/2020] [Indexed: 12/12/2022] Open
Abstract
Microbiota is defined as the collection of microorganisms within the gastrointestinal ecosystem. These microbes are strongly implicated in the stimulation of immune responses. An unbalanced microbiota, termed dysbiosis, is related to the development of several liver diseases. The bidirectional relationship between the gut, its microbiota and the liver is referred to as the gut-liver axis. The translocation of bacterial products from the intestine to the liver induces inflammation in different cell types such as Kupffer cells, and a fibrotic response in hepatic stellate cells, resulting in deleterious effects on hepatocytes. Moreover, ischemia-reperfusion injury, a consequence of liver surgery, alters the microbiota profile, affecting inflammation, the immune response and even liver regeneration. Microbiota also seems to play an important role in post-operative outcomes (i.e., liver transplantation or liver resection). Nonetheless, studies to determine changes in the gut microbial populations produced during and after surgery, and affecting liver function and regeneration are scarce. In the present review we analyze and discuss the preclinical and clinical studies reported in the literature focused on the evaluation of alterations in microbiota and its products as well as their effects on post-operative outcomes in hepatic surgery.
Collapse
Affiliation(s)
- Marc Micó-Carnero
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (M.M.-C.); (C.R.-A.)
| | - Carlos Rojano-Alfonso
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (M.M.-C.); (C.R.-A.)
| | - Ana Isabel Álvarez-Mercado
- Departamento de Bioquímica y Biología Molecular II, Escuela de Farmacia, Universidad de Granada, 18071 Granada, Spain;
- Institut of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria ibs, GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Laboratory IDIBAPS, 03036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08036 Barcelona, Spain
| | - Araní Casillas-Ramírez
- Hospital Regional de Alta Especialidad de Ciudad Victoria “Bicentenario 2010”, Ciudad Victoria 87087, Mexico;
- Facultad de Medicina e Ingeniería en Sistemas Computacionales de Matamoros, Universidad Autónoma de Tamaulipas, Matamoros 87300, Mexico
| | - Carmen Peralta
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (M.M.-C.); (C.R.-A.)
| |
Collapse
|
106
|
Li GH, Huang SJ, Li X, Liu XS, Du QL. Response of gut microbiota to serum metabolome changes in intrahepatic cholestasis of pregnant patients. World J Gastroenterol 2020; 26:7338-7351. [PMID: 33362388 PMCID: PMC7739160 DOI: 10.3748/wjg.v26.i46.7338] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/09/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Intrahepatic cholestasis in pregnancy (ICP) is the most common liver disease during pregnancy, and its exact etiology and course of progression are still poorly understood.
AIM To investigate the link between the gut microbiota and serum metabolome in ICP patients.
METHODS In this study, a total of 30 patients were recruited, including 15 patients with ICP (disease group) and 15 healthy pregnant patients (healthy group). The serum nontarget metabolomes from both groups were determined. Amplification of the 16S rRNA V3-V4 region was performed using fecal samples from the disease and healthy groups. By comparing the differences in the microbiota and metabolite compositions between the two groups, the relationship between the gut microbiota and serum metabolites was also investigated.
RESULTS The Kyoto Encyclopedia of Genes and Genomes analysis results showed that the primary bile acid biosynthesis, bile secretion and taurine and hypotaurine metabolism pathways were enriched in the ICP patients compared with the healthy controls. In addition, some pathways related to protein metabolism were also enriched in the ICP patients. The principal coordination analysis results showed that there was a distinct difference in the gut microbiota composition (beta diversity) between the ICP patients and healthy controls. At the phylum level, we observed that the relative abundance of Firmicutes was higher in the healthy group, while Bacteroidetes were enriched in the disease group. At the genus level, most of the bacteria depleted in ICP are able to produce short-chain fatty acids (e.g., Faecalibacterium, Blautia and Eubacterium hallii), while the bacteria enriched in ICP are associated with bile acid metabolism (e.g., Parabacteroides and Bilophila). Our results also showed that specific genera were associated with the serum metabolome.
CONCLUSION Our study showed that the serum metabolome was altered in ICP patients compared to healthy controls, with significant differences in the bile, taurine and hypotaurine metabolite pathways. Alterations in the metabolization of these pathways may lead to disturbances in the gut microbiota, which may further affect the course of progression of ICP.
Collapse
Affiliation(s)
- Guo-Hua Li
- Department of Reproductive Immunology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, China
| | - Shi-Jia Huang
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, China
| | - Xiang Li
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, China
| | - Xiao-Song Liu
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, China
| | - Qiao-Ling Du
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, China
| |
Collapse
|
107
|
Ballini A, Scacco S, Boccellino M, Santacroce L, Arrigoni R. Microbiota and Obesity: Where Are We Now? BIOLOGY 2020; 9:biology9120415. [PMID: 33255588 PMCID: PMC7761345 DOI: 10.3390/biology9120415] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/14/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023]
Abstract
Simple Summary Emerging new data reported in the international scientific literature show that specific alterations in the human gut microbiota are characteristic in obesity and obesity-related metabolic diseases. Obesity is conditioned by a multitude of factors, and the microbiota is certainly an important player. The analysis of the data obtained from experimental studies allow us to hypothesize that changes in the composition of the microbiota may be the cause, and not simply the consequence, of alterations in human metabolism. Clinical trials on wide samples that investigate the role of diet-induced modulation of the gut microbiota on the host metabolism are needed to understand the interactions at the molecular level for the observed correlations between metabolism and microbiota changes. Abstract Genetic and environmental factors are underlying causes of obesity and other metabolic diseases, so it is therefore difficult to find suitable and effective medical treatments. However, without a doubt, the gut microbiota—and also the bacteria present in the oral cavity—act as key factors in the development of these pathologies, yet the mechanisms have not been fully described. Certainly, a more detailed knowledge of the structure of the microbiota—composition, intra- and inter-species relationships, metabolic functions—could be of great help in counteracting the onset of obesity. Identifying key bacterial species will allow us to create a database of “healthy” bacteria, making it possible to manipulate the bacterial community according to metabolic and clinical needs. Targeting gut microbiota in clinical care as treatment for obesity and health-related complications—even just for weight loss has become a real possibility. In this topical review we provide an overview of the role of the microbiota on host energy homeostasis and obesity-related metabolic diseases, therefore addressing the therapeutic potential of novel and existing strategies (impact of nutrition/dietary modulation, and fecal microbiota transplantation) in the treatment of metabolic disease.
Collapse
Affiliation(s)
- Andrea Ballini
- Department of Biosciences, Biotechnologies and Biopharmaceutics, Campus Universitario, University of Bari “Aldo Moro”, 70125 Bari, Italy;
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70121 Bari, Italy
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Salvatore Scacco
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70121 Bari, Italy
- Correspondence: (S.S.); (M.B.); (R.A.)
| | - Mariarosaria Boccellino
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Correspondence: (S.S.); (M.B.); (R.A.)
| | - Luigi Santacroce
- Microbiology and Virology Laboratory, Ionian Department, Policlinico University Hospital, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Roberto Arrigoni
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), 70124 Bari, Italy
- Correspondence: (S.S.); (M.B.); (R.A.)
| |
Collapse
|
108
|
Mukherjee A, Lordan C, Ross RP, Cotter PD. Gut microbes from the phylogenetically diverse genus Eubacterium and their various contributions to gut health. Gut Microbes 2020; 12:1802866. [PMID: 32835590 PMCID: PMC7524325 DOI: 10.1080/19490976.2020.1802866] [Citation(s) in RCA: 341] [Impact Index Per Article: 68.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/10/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023] Open
Abstract
Over the last two decades our understanding of the gut microbiota and its contribution to health and disease has been transformed. Among a new 'generation' of potentially beneficial microbes to have been recognized are members of the genus Eubacterium, who form a part of the core human gut microbiome. The genus consists of phylogenetically, and quite frequently phenotypically, diverse species, making Eubacterium a taxonomically unique and challenging genus. Several members of the genus produce butyrate, which plays a critical role in energy homeostasis, colonic motility, immunomodulation and suppression of inflammation in the gut. Eubacterium spp. also carry out bile acid and cholesterol transformations in the gut, thereby contributing to their homeostasis. Gut dysbiosis and a consequently modified representation of Eubacterium spp. in the gut, have been linked with various human disease states. This review provides an overview of Eubacterium species from a phylogenetic perspective, describes how they alter with diet and age and summarizes its association with the human gut and various health conditions.
Collapse
Affiliation(s)
- Arghya Mukherjee
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Cathy Lordan
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - R. Paul Ross
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Paul D. Cotter
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
109
|
Van Hul M, Le Roy T, Prifti E, Dao MC, Paquot A, Zucker JD, Delzenne NM, Muccioli GG, Clément K, Cani PD. From correlation to causality: the case of Subdoligranulum. Gut Microbes 2020; 12:1-13. [PMID: 33323004 PMCID: PMC7744154 DOI: 10.1080/19490976.2020.1849998] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gut microbes are considered as major factors contributing to human health. Nowadays, the vast majority of the data available in the literature are mostly exhibiting negative or positive correlations between specific bacteria and metabolic parameters. From these observations, putative detrimental or beneficial effects are then inferred. Akkermansia muciniphila is one of the unique examples for which the correlations with health benefits have been causally validated in vivo in rodents and humans. In this study, based on available metagenomic data in overweight/obese population and clinical variables that we obtained from two cohorts of individuals (n = 108) we identified several metagenomic species (MGS) strongly associated with A. muciniphila with one standing out: Subdoligranulum. By analyzing both qPCR and shotgun metagenomic data, we discovered that the abundance of Subdoligranulum was correlated positively with microbial richness and HDL-cholesterol levels and negatively correlated with fat mass, adipocyte diameter, insulin resistance, levels of leptin, insulin, CRP, and IL6 in humans. Therefore, to further explore whether these strong correlations could be translated into causation, we investigated the effects of the unique cultivated strain of Subdoligranulum (Subdoligranulum variabile DSM 15176 T) in obese and diabetic mice as a proof-of-concept. Strikingly, there were no significant difference in any of the hallmarks of obesity and diabetes measured (e.g., body weight gain, fat mass gain, glucose tolerance, liver weight, plasma lipids) at the end of the 8 weeks of treatment. Therefore, the absence of effect following the supplementation with S. variabile indicates that increasing the intestinal abundance of this bacterium is not translated into beneficial effects in mice. In conclusion, we demonstrated that despite the fact that numerous strong correlations exist between a given bacteria and health, proof-of-concept experiments are required to be further validated or not in vivo. Hence, an urgent need for causality studies is warranted to move from human observations to preclinical validations.
Collapse
Affiliation(s)
- Matthias Van Hul
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Tiphaine Le Roy
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Edi Prifti
- Institut de Recherche et Developpement, IRD, Sorbonne Unive.rsity, UMMISCO, Bondy, France,Sorbonne Université, INSERM, Nutrition and Obesities: Systemic Approaches (Nutriomics) Research Unit, Paris, France
| | - Maria Carlota Dao
- Sorbonne Université, INSERM, Nutrition and Obesities: Systemic Approaches (Nutriomics) Research Unit, Paris, France
| | - Adrien Paquot
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Jean-Daniel Zucker
- Institut de Recherche et Developpement, IRD, Sorbonne Unive.rsity, UMMISCO, Bondy, France,Sorbonne Université, INSERM, Nutrition and Obesities: Systemic Approaches (Nutriomics) Research Unit, Paris, France
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Giulio G. Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Karine Clément
- Sorbonne Université, INSERM, Nutrition and Obesities: Systemic Approaches (Nutriomics) Research Unit, Paris, France,Assistance Publique Hôpitaux de Paris, Nutrition Department, Pitié-Salpêtrière Hospital, CRNH Ile de France, Paris, France
| | - Patrice D. Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université Catholique de Louvain, Brussels, Belgium,CONTACT Patrice D. Cani UCLouvain, Université Catholique de Louvain, LDRI, Metabolism and Nutrition Research Group, BrusselsB-1200, Belgium
| |
Collapse
|
110
|
Van de Guchte M, Burz SD, Cadiou J, Wu J, Mondot S, Blottière HM, Doré J. Alternative stable states in the intestinal ecosystem: proof of concept in a rat model and a perspective of therapeutic implications. MICROBIOME 2020; 8:153. [PMID: 33158453 PMCID: PMC7646066 DOI: 10.1186/s40168-020-00933-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/06/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Chronic immune-mediated diseases are rapidly expanding and notoriously difficult to cure. Altered relatively stable intestinal microbiota configurations are associated with several of these diseases, and with a possible pre-disease condition (more susceptible to disease development) of the host-microbiota ecosystem. These observations are reminiscent of the behavior of an ecosystem with alternative stable states (different stable configurations that can exist under identical external conditions), and we recently postulated that health, pre-disease and disease represent such alternative states. Here, our aim was to examine if alternative stable states indeed exist in the intestinal ecosystem. RESULTS Rats were exposed to varying concentrations of DSS in order to create a wide range of mildly inflammatory conditions, in a context of diet-induced low microbiota diversity. The consequences for the intestinal microbiota were traced by 16S rRNA gene profiling over time, and inflammation of the distal colon was evaluated at sacrifice, 45 days after the last DSS treatment. The results provide the first formal experimental proof for the existence of alternative stable states in the rat intestinal ecosystem, taking both microbiota and host inflammatory status into consideration. The alternative states are host-microbiota ecosystem states rather than independent and dissociated microbiota and host states, and inflammation can prompt stable state-transition. Based on these results, we propose a conceptual model providing new insights in the interplay between host inflammatory status and microbiota status. These new insights call for innovative therapeutic strategies to cure (pre-)disease. CONCLUSIONS We provide proof of concept showing the existence of alternative stable states in the rat intestinal ecosystem. We further propose a model which, if validated in humans, will support innovative diagnosis, therapeutic strategy, and monitoring in the treatment of chronic inflammatory conditions. This model provides a strong rationale for the application of combinatorial therapeutic strategies, targeting host and microbiota rather than only one of the two in chronic immune-mediated diseases. Video Abstract.
Collapse
Affiliation(s)
- Maarten Van de Guchte
- University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| | - Sebastian D. Burz
- University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
- University Paris-Saclay, INRAE, Metagenopolis, 78350 Jouy-en-Josas, France
| | - Julie Cadiou
- University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| | - Jiangbo Wu
- University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| | - Stanislas Mondot
- University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| | - Hervé M. Blottière
- University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
- University Paris-Saclay, INRAE, Metagenopolis, 78350 Jouy-en-Josas, France
| | - Joël Doré
- University Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
- University Paris-Saclay, INRAE, Metagenopolis, 78350 Jouy-en-Josas, France
| |
Collapse
|
111
|
Chen L, van den Munckhof ICL, Schraa K, Ter Horst R, Koehorst M, van Faassen M, van der Ley C, Doestzada M, Zhernakova DV, Kurilshikov A, Bloks VW, Groen AK, Riksen NP, Rutten JHW, Joosten LAB, Wijmenga C, Zhernakova A, Netea MG, Fu J, Kuipers F. Genetic and Microbial Associations to Plasma and Fecal Bile Acids in Obesity Relate to Plasma Lipids and Liver Fat Content. Cell Rep 2020; 33:108212. [PMID: 33027657 DOI: 10.1016/j.celrep.2020.108212] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/31/2020] [Accepted: 09/09/2020] [Indexed: 12/21/2022] Open
Abstract
Bile acids (BAs) are implicated in the etiology of obesity-related conditions such as non-alcoholic fatty liver disease. Differently structured BA species display variable signaling activities via farnesoid X receptor (FXR) and Takeda G protein-coupled BA receptor 1 (TGR5). This study profiles plasma and fecal BAs and plasma 7α-hydroxy-4-cholesten-3-one (C4) in 297 persons with obesity, identifies underlying genetic and microbial determinants, and establishes BA correlations with liver fat and plasma lipid parameters. We identify 27 genetic associations (p < 5 × 10-8) and 439 microbial correlations (FDR < 0.05) for 50 BA entities. Additionally, we report 111 correlations between BA and 88 lipid parameters (FDR < 0.05), mainly for C4 reflecting hepatic BA synthesis. Inter-individual variability in the plasma BA profile does not reflect hepatic BA synthetic pathways, but rather transport and metabolism within the enterohepatic circulation. Our study reveals genetic and microbial determinants of BAs in obesity and their relationship to disease-relevant lipid parameters that are important for the design of personalized therapies targeting BA-signaling pathways.
Collapse
Affiliation(s)
- Lianmin Chen
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands; Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands
| | - Inge C L van den Munckhof
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500HB, the Netherlands
| | - Kiki Schraa
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500HB, the Netherlands
| | - Rob Ter Horst
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500HB, the Netherlands
| | - Martijn Koehorst
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands
| | - Martijn van Faassen
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands
| | - Claude van der Ley
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands
| | - Marwah Doestzada
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands; Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands
| | - Daria V Zhernakova
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands; Laboratory of Genomic Diversity, Center for Computer Technologies, ITMO University, St. Petersburg 197101, Russia
| | - Alexander Kurilshikov
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands
| | - Vincent W Bloks
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands
| | - Albert K Groen
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands; Department of Vascular Medicine, University of Amsterdam, Amsterdam University Medical Center, Amsterdam 1012WX, the Netherlands
| | | | - Niels P Riksen
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500HB, the Netherlands
| | - Joost H W Rutten
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500HB, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500HB, the Netherlands; Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania
| | - Cisca Wijmenga
- University of Groningen, Groningen 9712CP, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500HB, the Netherlands; Department for Genomics & Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn 53113, Germany; Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova 200349, Romania
| | - Jingyuan Fu
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands; Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands.
| | - Folkert Kuipers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen 9713AV, the Netherlands.
| |
Collapse
|
112
|
Cuffaro B, Assohoun ALW, Boutillier D, Súkeníková L, Desramaut J, Boudebbouze S, Salomé-Desnoulez S, Hrdý J, Waligora-Dupriet AJ, Maguin E, Grangette C. In Vitro Characterization of Gut Microbiota-Derived Commensal Strains: Selection of Parabacteroides distasonis Strains Alleviating TNBS-Induced Colitis in Mice. Cells 2020; 9:cells9092104. [PMID: 32947881 PMCID: PMC7565435 DOI: 10.3390/cells9092104] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/15/2020] [Accepted: 07/20/2020] [Indexed: 12/17/2022] Open
Abstract
Alterations in the gut microbiota composition and diversity seem to play a role in the development of chronic diseases, including inflammatory bowel disease (IBD), leading to gut barrier disruption and induction of proinflammatory immune responses. This opens the door for the use of novel health-promoting bacteria. We selected five Parabacteroides distasonis strains isolated from human adult and neonates gut microbiota. We evaluated in vitro their immunomodulation capacities and their ability to reinforce the gut barrier and characterized in vivo their protective effects in an acute murine model of colitis. The in vitro beneficial activities were highly strain dependent: two strains exhibited a potent anti-inflammatory potential and restored the gut barrier while a third strain reinstated the epithelial barrier. While their survival to in vitro gastric conditions was variable, the levels of P. distasonis DNA were higher in the stools of bacteria-treated animals. The strains that were positively scored in vitro displayed a strong ability to rescue mice from colitis. We further showed that two strains primed dendritic cells to induce regulatory T lymphocytes from naïve CD4+ T cells. This study provides better insights on the functionality of commensal bacteria and crucial clues to design live biotherapeutics able to target inflammatory chronic diseases such as IBD.
Collapse
Affiliation(s)
- Bernardo Cuffaro
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Centre d’Infection et d’Immunité de Lille, 59000 Lille, France; (B.C.); (D.B.); (J.D.)
- Institut Micalis, MIHA Team, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France; (A.L.W.A.); (S.B.)
| | - Aka L. W. Assohoun
- Institut Micalis, MIHA Team, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France; (A.L.W.A.); (S.B.)
- Laboratoire de Biotechnologie et Microbiologie des Aliments, UFR en Sciences et Technologies des Aliments, Université Nangui Abrogoua, Abidjan 00225, Côte d’Ivoire
| | - Denise Boutillier
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Centre d’Infection et d’Immunité de Lille, 59000 Lille, France; (B.C.); (D.B.); (J.D.)
| | - Lenka Súkeníková
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital, 121 08 Prague, Czech Republic; (L.S.); (J.H.)
| | - Jérémy Desramaut
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Centre d’Infection et d’Immunité de Lille, 59000 Lille, France; (B.C.); (D.B.); (J.D.)
| | - Samira Boudebbouze
- Institut Micalis, MIHA Team, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France; (A.L.W.A.); (S.B.)
| | - Sophie Salomé-Desnoulez
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41-UMS 2014-PLBS, 59000 Lille, France;
| | - Jiří Hrdý
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital, 121 08 Prague, Czech Republic; (L.S.); (J.H.)
| | | | - Emmanuelle Maguin
- Institut Micalis, MIHA Team, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France; (A.L.W.A.); (S.B.)
- Correspondence: (E.M.); (C.G.); Tel.: +33-681-151-925 (E.M.); +33-320-877-392 (C.G.)
| | - Corinne Grangette
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Centre d’Infection et d’Immunité de Lille, 59000 Lille, France; (B.C.); (D.B.); (J.D.)
- Correspondence: (E.M.); (C.G.); Tel.: +33-681-151-925 (E.M.); +33-320-877-392 (C.G.)
| |
Collapse
|
113
|
Ntemiri A, Ghosh TS, Gheller ME, Tran TTT, Blum JE, Pellanda P, Vlckova K, Neto MC, Howell A, Thalacker-Mercer A, O’Toole PW. Whole Blueberry and Isolated Polyphenol-Rich Fractions Modulate Specific Gut Microbes in an In Vitro Colon Model and in a Pilot Study in Human Consumers. Nutrients 2020; 12:E2800. [PMID: 32932733 PMCID: PMC7551244 DOI: 10.3390/nu12092800] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/08/2020] [Accepted: 09/08/2020] [Indexed: 12/24/2022] Open
Abstract
Blueberry (BB) consumption is linked to improved health. The bioconversion of the polyphenolic content of BB by fermentative bacteria in the large intestine may be a necessary step for the health benefits attributed to BB consumption. The identification of specific gut microbiota taxa that respond to BB consumption and that mediate the bioconversion of consumed polyphenolic compounds into bioactive forms is required to improve our understanding of how polyphenols impact human health. We tested the ability of polyphenol-rich fractions purified from whole BB-namely, anthocyanins/flavonol glycosides (ANTH/FLAV), proanthocyanidins (PACs), the sugar/acid fraction (S/A), and total polyphenols (TPP)-to modulate the fecal microbiota composition of healthy adults in an in vitro colon system. In a parallel pilot study, we tested the effect of consuming 38 g of freeze-dried BB powder per day for 6 weeks on the fecal microbiota of 17 women in two age groups (i.e., young and older). The BB ingredients had a distinct effect on the fecal microbiota composition in the artificial colon model. The ANTH/FLAV and PAC fractions were more effective in promoting microbiome alpha diversity compared to S/A and TPP, and these effects were attributed to differentially responsive taxa. Dietary enrichment with BB resulted in a moderate increase in the diversity of the microbiota of the older subjects but not in younger subjects, and certain health-relevant taxa were significantly associated with BB consumption. Alterations in the abundance of some gut bacteria correlated not only with BB consumption but also with increased antioxidant activity in blood. Collectively, these pilot data support the notion that BB consumption is associated with gut microbiota changes and health benefits.
Collapse
Affiliation(s)
- Alexandra Ntemiri
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (A.N.); (T.S.G.); (T.T.T.T.); (P.P.); (K.V.); (M.C.N.)
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
| | - Tarini S. Ghosh
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (A.N.); (T.S.G.); (T.T.T.T.); (P.P.); (K.V.); (M.C.N.)
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
| | - Molly E. Gheller
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; (M.E.G.); (J.E.B.); (A.T.-M.)
| | - Tam T. T. Tran
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (A.N.); (T.S.G.); (T.T.T.T.); (P.P.); (K.V.); (M.C.N.)
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
| | - Jamie E. Blum
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; (M.E.G.); (J.E.B.); (A.T.-M.)
| | - Paola Pellanda
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (A.N.); (T.S.G.); (T.T.T.T.); (P.P.); (K.V.); (M.C.N.)
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
| | - Klara Vlckova
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (A.N.); (T.S.G.); (T.T.T.T.); (P.P.); (K.V.); (M.C.N.)
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
| | - Marta C. Neto
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (A.N.); (T.S.G.); (T.T.T.T.); (P.P.); (K.V.); (M.C.N.)
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
| | - Amy Howell
- Marucci Center for Blueberry Cranberry Research, Rutgers University, Chatsworth, NJ 08019, USA;
| | - Anna Thalacker-Mercer
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; (M.E.G.); (J.E.B.); (A.T.-M.)
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, AL 35294, USA
| | - Paul W. O’Toole
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (A.N.); (T.S.G.); (T.T.T.T.); (P.P.); (K.V.); (M.C.N.)
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
| |
Collapse
|
114
|
Pivotal Roles for pH, Lactate, and Lactate-Utilizing Bacteria in the Stability of a Human Colonic Microbial Ecosystem. mSystems 2020; 5:5/5/e00645-20. [PMID: 32900872 PMCID: PMC7483512 DOI: 10.1128/msystems.00645-20] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lactate can be produced by many gut bacteria, but in adults its accumulation in the colon is often an indicator of microbiota perturbation. Using continuous culture anaerobic fermentor systems, we found that lactate concentrations remained low in communities of human colonic bacteria maintained at pH 6.5, even when dl-lactate was infused at 10 or 20 mM. In contrast, lower pH (5.5) led to periodic lactate accumulation following lactate infusion in three fecal microbial communities examined. Lactate accumulation was concomitant with greatly reduced butyrate and propionate production and major shifts in microbiota composition, with Bacteroidetes and anaerobic Firmicutes being replaced by Actinobacteria, lactobacilli, and Proteobacteria Pure-culture experiments confirmed that Bacteroides and Firmicutes isolates were susceptible to growth inhibition by relevant concentrations of lactate and acetate, whereas the lactate-producer Bifidobacterium adolescentis was resistant. To investigate system behavior further, we used a mathematical model (microPop) based on 10 microbial functional groups. By incorporating differential growth inhibition, our model reproduced the chaotic behavior of the system, including the potential for lactate infusion both to promote and to rescue the perturbed system. The modeling revealed that system behavior is critically dependent on the proportion of the community able to convert lactate into butyrate or propionate. Communities with low numbers of lactate-utilizing bacteria are inherently less stable and more prone to lactate-induced perturbations. These findings can help us to understand the consequences of interindividual microbiota variation for dietary responses and microbiota changes associated with disease states.IMPORTANCE Lactate is formed by many species of colonic bacteria, and can accumulate to high levels in the colons of inflammatory bowel disease subjects. Conversely, in healthy colons lactate is metabolized by lactate-utilizing species to the short-chain fatty acids butyrate and propionate, which are beneficial for the host. Here, we investigated the impact of continuous lactate infusions (up to 20 mM) at two pH values (6.5 and 5.5) on human colonic microbiota responsiveness and metabolic outputs. At pH 5.5 in particular, lactate tended to accumulate in tandem with decreases in butyrate and propionate and with corresponding changes in microbial composition. Moreover, microbial communities with low numbers of lactate-utilizing bacteria were inherently less stable and therefore more prone to lactate-induced perturbations. These investigations provide clear evidence of the important role these lactate utilizers may play in health maintenance. These should therefore be considered as potential new therapeutic probiotics to combat microbiota perturbations.
Collapse
|
115
|
Li H, Yu Q, Li T, Shao L, Su M, Zhou H, Qu J. Rumen Microbiome and Metabolome of Tibetan Sheep ( Ovis aries) Reflect Animal Age and Nutritional Requirement. Front Vet Sci 2020; 7:609. [PMID: 32984417 PMCID: PMC7492597 DOI: 10.3389/fvets.2020.00609] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 07/28/2020] [Indexed: 01/26/2023] Open
Abstract
The rumen microbiota plays an important role in animal functional attributes. These microbes are indispensable for the normal physiological development of the rumen, and may also convert the plant polysaccharides from grass into available milk and meat, making it highly valuable to humans. Exploring the microbial composition and metabolites of rumen across developmental stages is important for understanding ruminant nutrition and metabolism. However, relatively few reports have investigated the microbiome and metabolites across developmental stages in ruminants. Using 16S rRNA gene sequnecing, metabolomics and high-performance liquid chromatography techniques, we compared the rumen microbiota, metabolites and short chain fatty acids (SCFAs) between lambs and sub-adult Tibetan sheep (Ovis aries) from Qinghai-Tibetan Plateau. Bacteroidetes and Spirochaetae were enriched in sub-adult sheep, while Firmicutes and Tenericutes were more abundant in young individuals. The sub-adult individuals had higher alpha diversity values than those in young sheep. Metabolomics analysis showed that the content of essential amino acids and related gene functional pathways in rumen were different between the lambs and sub-adult population. L-Leucine that participates in valine, leucine and isoleucine biosynthesis was more abundant in the lambs, while phenylethylamine that takes part in phenylalanine metabolism was more enriched in the sub-adults. Both rumen microbial community structures and metabolite profiles were impacted by age, but rumen SCFA concentration was relatively stable between different age stages. Some specific microbes (e.g., Clostridium and Ruminococcaceae) were positively associated with L-Leucine but negatively correlated with phenylethylamine, implying that rumen microbes may play different roles for metabolite production at different ages. Mantel test analysis showed that rumen microbiota was significantly correlated with metabolomics and SCFA profiles. Our results indicates the close relationship between microbial composition and metabolites, and also reveal different nutritional requirement for different ages in ruminants, thus having important significance for regulating animal nutrition and metabolism by microbiome intervention.
Collapse
Affiliation(s)
- Huan Li
- School of Public Health, Lanzhou University, Lanzhou, China.,Key Laboratory of Restoration Ecology for Cold Regions in Qinghai, Xining, China
| | - Qiaoling Yu
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Tongtong Li
- Department of Applied Biology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Liye Shao
- Key Laboratory of Health Aquaculture and Product Processing in Dongting Lake Area of Hunan Province, Zoology Key Laboratory of Hunan Higher Education, Hunan University of Arts and Science, Changde, China
| | - Ming Su
- Central South Inventory and Planning Institute of National Forestry and Grassland Administration, Changsha, China
| | - Huakun Zhou
- Key Laboratory of Restoration Ecology for Cold Regions in Qinghai, Xining, China.,Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
| | - Jiapeng Qu
- Key Laboratory of Restoration Ecology for Cold Regions in Qinghai, Xining, China.,Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
| |
Collapse
|
116
|
Yang J, Yang Y, Ishii M, Nagata M, Aw W, Obana N, Tomita M, Nomura N, Fukuda S. Does the Gut Microbiota Modulate Host Physiology through Polymicrobial Biofilms? Microbes Environ 2020; 35. [PMID: 32624527 PMCID: PMC7511787 DOI: 10.1264/jsme2.me20037] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Microbes inhabit various environments, such as soil, water environments, plants, and animals. Humans harbor a complex commensal microbial community in the gastrointestinal tract, which is known as the gut microbiota. The gut microbiota participates not only in various metabolic processes in the human body, it also plays a critical role in host immune responses. Gut microbes that inhabit the intestinal epithelial surface form polymicrobial biofilms. In the last decade, it has been widely reported that gut microbial biofilms and gut microbiota-derived products, such as metabolites and bacterial membrane vesicles, not only directly affect the host intestinal environment, but also indirectly influence the health of the host. In this review, we discuss the most recent findings from human and animal studies on the interactions between the gut microbiota and hosts, and their associations with various disorders, including inflammatory diseases, atopic dermatitis, metabolic disorders, and psychiatric and neurological diseases. The integrated approach of metabologenomics together with biofilm imaging may provide valuable insights into the gut microbiota and suggest remedies that may lead to a healthier society.
Collapse
Affiliation(s)
- Jiayue Yang
- Institute for Advanced Biosciences, Keio University.,Systems Biology Program, Graduate School of Media and Governance, Keio University
| | | | - Manami Ishii
- Institute for Advanced Biosciences, Keio University.,Systems Biology Program, Graduate School of Media and Governance, Keio University
| | - Mayuko Nagata
- Institute for Advanced Biosciences, Keio University.,Faculty of Environment and Information Studies, Keio University
| | - Wanping Aw
- Institute for Advanced Biosciences, Keio University.,Systems Biology Program, Graduate School of Media and Governance, Keio University
| | - Nozomu Obana
- Transborder Medical Research Center, University of Tsukuba
| | - Masaru Tomita
- Institute for Advanced Biosciences, Keio University.,Systems Biology Program, Graduate School of Media and Governance, Keio University.,Faculty of Environment and Information Studies, Keio University
| | - Nobuhiko Nomura
- Microbiology Research Center for Sustainability, University of Tsukuba.,Faculty of Life and Environmental Sciences, University of Tsukuba
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University.,Systems Biology Program, Graduate School of Media and Governance, Keio University.,Transborder Medical Research Center, University of Tsukuba.,Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology.,Metabologenomics, Inc
| |
Collapse
|
117
|
Changes of saliva microbiota in the onset and after the treatment of diabetes in patients with periodontitis. Aging (Albany NY) 2020; 12:13090-13114. [PMID: 32634783 PMCID: PMC7377876 DOI: 10.18632/aging.103399] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/25/2020] [Indexed: 01/06/2023]
Abstract
The relationship between type 2 diabetes mellitus (T2DM) and oral microbiota is still insufficiently recognized. In the present study, we compared the salivary microbiome of nondiabetic individuals, treatment-naïve diabetic patients, and diabetic patients treated with metformin or a combination of insulin and other drugs. The α- and β-diversity demonstrated significant differences in the salivary microbiome between the nondiabetic people and patients with a history of diabetes, while little divergence was found among individuals with a history of diabetes. After characterizing the effects of periodontitis on the microbial composition of each group, the salivary microbiome of the treatment-naïve diabetic patient group was compared with that of nondiabetic people and the metformin/combined treatment groups. The results revealed changes in the contents of certain bacteria after both the onset and the treatment of diabetes; among these differential bacteria, Blautia_wexlerae, Lactobacillus_fermentum, Nocardia_coeliaca and Selenomonas_artemidis varied in all processes. A subsequent correlational analysis of the differential bacteria and clinical characteristics demonstrated that salivary microbes were related to drug treatment and certain pathological changes. Finally, the four common differential bacteria were employed for distinguishing the treatment-naïve diabetic patients from the nondiabetic people and the treated patients, with prediction accuracies of 83.3%, 75% and 75%, respectively.
Collapse
|
118
|
A Novel Non-Digestible, Carrot-Derived Polysaccharide (cRG-I) Selectively Modulates the Human Gut Microbiota while Promoting Gut Barrier Integrity: An Integrated in Vitro Approach. Nutrients 2020; 12:nu12071917. [PMID: 32610452 PMCID: PMC7400138 DOI: 10.3390/nu12071917] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 12/21/2022] Open
Abstract
Modulation of the gut microbiome as a means to improve human health has recently gained increasing interest. In this study, it was investigated whether cRG-I, a carrot-derived pectic polysaccharide, enriched in rhamnogalacturonan-I (RG-I) classifies as a potential prebiotic ingredient using novel in vitro models. First, digestion methods involving α-amylase/brush border enzymes demonstrated the non-digestibility of cRG-I by host-derived enzymes versus digestible (starch/maltose) and non-digestible controls (inulin). Then, a recently developed short-term (48 h) colonic incubation strategy was applied and revealed that cRG-I fermentation increased levels of health-promoting short-chain fatty acids (SCFA; mainly acetate and propionate) and lactate comparable but not identical to the reference prebiotic inulin. Upon upgrading this fermentation model by inclusion of a simulated mucosal environment while applying quantitative 16S-targeted Illumina sequencing, cRG-I was additionally shown to specifically stimulate operational taxonomic units (OTUs) related to health-associated species such as Bifidobacterium longum, Bifidobacterium adolescentis, Bacteroides dorei, Bacteroides ovatus, Roseburia hominis, Faecalibacterium prausnitzii, and Eubacterium hallii. Finally, in a novel model to assess host–microbe interactions (Caco-2/peripheral blood mononuclear cells (PBMC) co-culture) fermented cRG-I increased barrier integrity while decreasing markers for inflammation. In conclusion, by using novel in vitro models, cRG-I was identified as a promising prebiotic candidate to proceed to clinical studies.
Collapse
|
119
|
Andrade JC, Almeida D, Domingos M, Seabra CL, Machado D, Freitas AC, Gomes AM. Commensal Obligate Anaerobic Bacteria and Health: Production, Storage, and Delivery Strategies. Front Bioeng Biotechnol 2020; 8:550. [PMID: 32582673 PMCID: PMC7291883 DOI: 10.3389/fbioe.2020.00550] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
In the last years several human commensals have emerged from the gut microbiota studies as potential probiotics or therapeutic agents. Strains of human gut inhabitants such as Akkermansia, Bacteroides, or Faecalibacterium have shown several interesting bioactivities and are thus currently being considered as food supplements or as live biotherapeutics, as is already the case with other human commensals such as bifidobacteria. The large-scale use of these bacteria will pose many challenges and drawbacks mainly because they are quite sensitive to oxygen and/or very difficult to cultivate. This review highlights the properties of some of the most promising human commensals bacteria and summarizes the most up-to-date knowledge on their potential health effects. A comprehensive outlook on the potential strategies currently employed and/or available to produce, stabilize, and deliver these microorganisms is also presented.
Collapse
Affiliation(s)
- José Carlos Andrade
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Gandra, Portugal
| | - Diana Almeida
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Melany Domingos
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Catarina Leal Seabra
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Daniela Machado
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Ana Cristina Freitas
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Ana Maria Gomes
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| |
Collapse
|
120
|
Wijdeveld M, Nieuwdorp M, IJzerman R. The interaction between microbiome and host central nervous system: the gut-brain axis as a potential new therapeutic target in the treatment of obesity and cardiometabolic disease. Expert Opin Ther Targets 2020; 24:639-653. [PMID: 32441559 DOI: 10.1080/14728222.2020.1761958] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The role of the intestinal microbiota in host cardiometabolic health and disease has gained significant attention over recent decades. Previous studies have shown effects on metabolic health through gut microbiota modulation; this suggests diverse interaction pathways that constitute the communication between gut microbiota and host central nervous system, the so-called gut-brain axis. AREAS COVERED This article provides an overview of the various mechanisms that may mediate the gut-brain axis. It places an emphasis on cardiometabolic health, including effects of short-chain fatty acids (SCFA), alterations in neurotransmitters and gut peptides and microbial effects on chronic inflammation and immune function. Moreover, this paper sheds light on whether these mechanisms afford therapeutic targets to promote metabolic health. To this end, a PubMed search with the terms 'gut microbiota,' 'obesity' and 'insulin sensitivity' was performed. EXPERT OPINION Many properties of the human gut microbiome are associated with the central regulation of appetite and metabolic status. Some of these relationships are causal and there are positive effects from certain intervention methods. Microbial manipulation may offer a means to prevent or treat obesity and associated co-morbidities. However, to establish direct causal relations between altered gut microbiota and metabolic disease, clinical intervention studies are necessary.
Collapse
Affiliation(s)
- Madelief Wijdeveld
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers , Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers , Amsterdam, The Netherlands
| | - Richard IJzerman
- Department of Endocrinology, Amsterdam University Medical Centers , Amsterdam, The Netherlands
| |
Collapse
|
121
|
Crobach MJT, Ducarmon QR, Terveer EM, Harmanus C, Sanders IMJG, Verduin KM, Kuijper EJ, Zwittink RD. The Bacterial Gut Microbiota of Adult Patients Infected, Colonized or Noncolonized by Clostridioides difficile. Microorganisms 2020; 8:microorganisms8050677. [PMID: 32384826 PMCID: PMC7284656 DOI: 10.3390/microorganisms8050677] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 12/15/2022] Open
Abstract
: Gut microbiota composition in patients with Clostridioides difficile colonization is not well investigated. We aimed to identify bacterial signatures associated with resistance and susceptibility to C. difficile colonization (CDC) and infection (CDI). Therefore, gut microbiota composition from patients with CDC (n = 41), with CDI (n = 41), and without CDC (controls, n = 43) was determined through 16S rRNA gene amplicon sequencing. Bacterial diversity was decreased in CDC and CDI patients (p<0.01). Overall microbiota composition was significantly different between control, CDC, and CDI patients (p = 0.001). Relative abundance of Clostridioides (most likely C. difficile) increased stepwise from controls to CDC and CDI patients. In addition, differential abundance analysis revealed that CDI patients' gut microbiota was characterized by significantly higher relative abundance of Bacteroides and Veillonella than CDC patients and controls. Control patients had significantly higher Eubacterium hallii and Fusicatenibacter abundance than colonized patients. Network analysis indicated that Fusicatenibacter was negatively associated with Clostridioides in CDI patients, while Veillonella was positively associated with Clostridioides in CDC patients. Bacterial microbiota diversity decreased in both CDC and CDI patients, but harbored a distinct microbiota. Eubacterium hallii and Fusicatenibacter may indicate resistance against C. difficile colonization and subsequent infection, while Veillonella may indicate susceptibility to colonization and infection by C. difficile.
Collapse
Affiliation(s)
- Monique J. T. Crobach
- Experimental Bacteriology, Department of Medical Microbiology, Center for Infectious Diseases, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (M.J.T.C.); (Q.R.D.); (E.M.T.); (C.H.); (I.M.J.G.S.); (E.J.K.)
| | - Quinten R. Ducarmon
- Experimental Bacteriology, Department of Medical Microbiology, Center for Infectious Diseases, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (M.J.T.C.); (Q.R.D.); (E.M.T.); (C.H.); (I.M.J.G.S.); (E.J.K.)
- Center for Microbiome Analyses and Therapeutics, Department of Medical Microbiology, Center for Infectious Diseases, Leiden University Medical Center, 2333ZA Leiden, The Netherlands
| | - Elisabeth M. Terveer
- Experimental Bacteriology, Department of Medical Microbiology, Center for Infectious Diseases, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (M.J.T.C.); (Q.R.D.); (E.M.T.); (C.H.); (I.M.J.G.S.); (E.J.K.)
- Center for Microbiome Analyses and Therapeutics, Department of Medical Microbiology, Center for Infectious Diseases, Leiden University Medical Center, 2333ZA Leiden, The Netherlands
- Netherlands Donor Feces Bank, 2333ZA Leiden, The Netherlands
| | - Celine Harmanus
- Experimental Bacteriology, Department of Medical Microbiology, Center for Infectious Diseases, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (M.J.T.C.); (Q.R.D.); (E.M.T.); (C.H.); (I.M.J.G.S.); (E.J.K.)
| | - Ingrid M. J. G. Sanders
- Experimental Bacteriology, Department of Medical Microbiology, Center for Infectious Diseases, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (M.J.T.C.); (Q.R.D.); (E.M.T.); (C.H.); (I.M.J.G.S.); (E.J.K.)
| | - Kees M. Verduin
- Department of Microbiology and Infection Prevention, Amphia Hospital, 4818CK Breda, The Netherlands;
| | - Ed J. Kuijper
- Experimental Bacteriology, Department of Medical Microbiology, Center for Infectious Diseases, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (M.J.T.C.); (Q.R.D.); (E.M.T.); (C.H.); (I.M.J.G.S.); (E.J.K.)
- Center for Microbiome Analyses and Therapeutics, Department of Medical Microbiology, Center for Infectious Diseases, Leiden University Medical Center, 2333ZA Leiden, The Netherlands
- Netherlands Donor Feces Bank, 2333ZA Leiden, The Netherlands
| | - Romy D. Zwittink
- Experimental Bacteriology, Department of Medical Microbiology, Center for Infectious Diseases, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (M.J.T.C.); (Q.R.D.); (E.M.T.); (C.H.); (I.M.J.G.S.); (E.J.K.)
- Center for Microbiome Analyses and Therapeutics, Department of Medical Microbiology, Center for Infectious Diseases, Leiden University Medical Center, 2333ZA Leiden, The Netherlands
- Correspondence: ; Tel.: +31-(0)71-526-4830
| |
Collapse
|
122
|
Hsieh PS, Ho HH, Hsieh SH, Kuo YW, Tseng HY, Kao HF, Wang JY. Lactobacillus salivarius AP-32 and Lactobacillus reuteri GL-104 decrease glycemic levels and attenuate diabetes-mediated liver and kidney injury in db/db mice. BMJ Open Diabetes Res Care 2020; 8:8/1/e001028. [PMID: 32332068 PMCID: PMC7202753 DOI: 10.1136/bmjdrc-2019-001028] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/27/2020] [Accepted: 03/24/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Patients with type 2 diabetes mellitus (T2DM) exhibit strong insulin resistance or abnormal insulin production. Probiotics, which are beneficial live micro-organisms residing naturally in the intestinal tract, play indispensable roles in the regulation of host metabolism. However, the detailed mechanisms remain unclear. Here, we evaluate the mechanisms by which probiotic strains mediate glycemic regulation in the host. The findings should enable the development of a safe and natural treatment for patients with T2DM. RESEARCH DESIGNS AND METHODS Sugar consumption by more than 20 strains of Lactobacillus species was first evaluated. The probiotic strains that exhibited high efficiency of sugar consumption were further coincubated with Caco-2 cells to evaluate the regulation of sugar absorption in gut epithelial cells. Finally, potential probiotic strains were selected and introduced into a T2DM animal model to study their therapeutic efficacy. RESULTS Among the tested strains, Lactobacillus salivarius AP-32 and L. reuteri GL-104 had higher monosaccharide consumption rates and regulated the expression of monosaccharide transporters. Glucose transporter type-5 and Na+-coupled glucose transporter mRNAs were downregulated in Caco-2 cells after AP-32 and GL-104 treatment, resulting in the modulation of intestinal hexose uptake. Animal studies revealed that diabetic mice treated with AP-32, GL-104, or both showed significantly decreased fasting blood glucose levels, improved glucose tolerance and blood lipid profiles, and attenuated diabetes-mediated liver and kidney injury. CONCLUSION Our data elucidate a novel role for probiotics in glycemic regulation in the host. L. salivarius AP-32 and L. reuteri GL-104 directly reduce monosaccharide transporter expression in gut cells and have potential as therapeutic probiotics for patients with T2DM.
Collapse
Affiliation(s)
- Pei-Shan Hsieh
- Research and Development Department, Glac Biotech Co., Ltd, Tainan, Taiwan
| | - Hsieh-Hsun Ho
- Research and Development Department, Glac Biotech Co., Ltd, Tainan, Taiwan
| | - Shih-Hung Hsieh
- Research and Development Department, Glac Biotech Co., Ltd, Tainan, Taiwan
| | - Yi-Wei Kuo
- Research and Development Department, Glac Biotech Co., Ltd, Tainan, Taiwan
| | - Hsiu-Ying Tseng
- Department of Pediatrics, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Hui-Fang Kao
- Department of Pediatrics, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Jiu-Yao Wang
- Department of Pediatrics, National Cheng Kung University Hospital, Tainan, Taiwan
| |
Collapse
|
123
|
Gilijamse PW, Hartstra AV, Levin E, Wortelboer K, Serlie MJ, Ackermans MT, Herrema H, Nederveen AJ, Imangaliyev S, Aalvink S, Sommer M, Levels H, Stroes ESG, Groen AK, Kemper M, de Vos WM, Nieuwdorp M, Prodan A. Treatment with Anaerobutyricum soehngenii: a pilot study of safety and dose-response effects on glucose metabolism in human subjects with metabolic syndrome. NPJ Biofilms Microbiomes 2020; 6:16. [PMID: 32221294 PMCID: PMC7101376 DOI: 10.1038/s41522-020-0127-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/09/2020] [Indexed: 01/25/2023] Open
Abstract
Dysbiosis of the intestinal microbiota has been implicated in insulin resistance, although evidence regarding causality in humans is scarce. We performed a phase I/II dose-finding and safety study on the effect of oral intake of the anaerobic butyrogenic strain Anaerobutyricum soehngenii on glucose metabolism in 24 subjects with metabolic syndrome. We found that treatment with A. soehngenii was safe and observed a significant correlation between the measured fecal abundance of administered A. soehngenii and improvement in peripheral insulin sensitivity after 4 weeks of treatment. This was accompanied by an altered microbiota composition and a change in bile acid metabolism. Finally, we show that metabolic response upon administration of A. soehngenii (defined as improved insulin sensitivity 4 weeks after A. soehngenii intake) is dependent on microbiota composition at baseline. These data in humans are promising, but additional studies are needed to reproduce our findings and to investigate long-term effects, as well as other modes of delivery.
Collapse
Affiliation(s)
- Pim W Gilijamse
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands.,Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Annick V Hartstra
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Evgeni Levin
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Koen Wortelboer
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Mireille J Serlie
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Mariette T Ackermans
- Laboratory of Endocrinology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Hilde Herrema
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Aart J Nederveen
- Department of Radiology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Sultan Imangaliyev
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Steven Aalvink
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | | | - Han Levels
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Albert K Groen
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Marleen Kemper
- Department of Clinical Pharmacy, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Willem M de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands.,Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Andrei Prodan
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands.
| |
Collapse
|
124
|
Benítez-Páez A, Gómez Del Pugar EM, López-Almela I, Moya-Pérez Á, Codoñer-Franch P, Sanz Y. Depletion of Blautia Species in the Microbiota of Obese Children Relates to Intestinal Inflammation and Metabolic Phenotype Worsening. mSystems 2020; 5:e00857-19. [PMID: 32209719 PMCID: PMC7093825 DOI: 10.1128/msystems.00857-19] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/06/2020] [Indexed: 12/16/2022] Open
Abstract
Cross-sectional studies conducted with obese and control subjects have suggested associations between gut microbiota alterations and obesity, but the links with specific disease phenotypes and proofs of causality are still scarce. The present study aimed to profile the gut microbiota of lean and obese children with and without insulin resistance to characterize associations with specific obesity-related complications and understand the role played in metabolic inflammation. Through massive sequencing of 16S rRNA gene amplicons and data analysis using a novel permutation approach, we have detected decreased incidence of Blautia species, especially Blautia luti and B. wexlerae, in the gut microbiota of obese children, which was even more pronounced in cases with both obesity and insulin resistance. There was also a parallel increase in proinflammatory cytokines and chemokines (gamma interferon [IFN-γ], tumor necrosis factor alpha [TNF-α], and monocyte chemoattractant protein 1 [MCP-1]) in feces of obese children compared to those of lean ones. B. luti and B. wexlerae were also shown to exert an anti-inflammatory effect in peripheral blood mononuclear cell cultures in vitro, compared to non-obesity-associated species. We suggest that the depletion of B. luti and B. wexlerae species in the gut ecosystem may occur in cases of obesity and contribute to metabolic inflammation leading to insulin resistance.IMPORTANCE Child obesity constitutes a risk factor for developing insulin resistance which, if sustained, could lead to more severe conditions like type 2 diabetes (T2D) in adulthood. Our study identified previously unknown species whose depletion (Blautia luti and Blautia wexlerae) is associated with insulin resistance in obese individuals. Our results also indicate that these bacterial species might help to reduce inflammation causally linked to obesity-related complications. Childhood is considered a window of opportunity to tackle obesity. These new findings provide, therefore, valuable information for the future design of microbiota-based strategies for the early prevention of obesity-related complications.
Collapse
Affiliation(s)
- Alfonso Benítez-Páez
- Microbial Ecology, Nutrition, and Health Research Unit, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| | - Eva M Gómez Del Pugar
- Microbial Ecology, Nutrition, and Health Research Unit, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| | - Inmaculada López-Almela
- Microbial Ecology, Nutrition, and Health Research Unit, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| | - Ángela Moya-Pérez
- Microbial Ecology, Nutrition, and Health Research Unit, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| | - Pilar Codoñer-Franch
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain
- Department of Pediatrics, Dr. Peset University Hospital, Valencia, Spain
| | - Yolanda Sanz
- Microbial Ecology, Nutrition, and Health Research Unit, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Paterna-Valencia, Spain
| |
Collapse
|
125
|
Liu Z, Dai X, Zhang H, Shi R, Hui Y, Jin X, Zhang W, Wang L, Wang Q, Wang D, Wang J, Tan X, Ren B, Liu X, Zhao T, Wang J, Pan J, Yuan T, Chu C, Lan L, Yin F, Cadenas E, Shi L, Zhao S, Liu X. Gut microbiota mediates intermittent-fasting alleviation of diabetes-induced cognitive impairment. Nat Commun 2020; 11:855. [PMID: 32071312 PMCID: PMC7029019 DOI: 10.1038/s41467-020-14676-4] [Citation(s) in RCA: 319] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 01/27/2020] [Indexed: 12/14/2022] Open
Abstract
Cognitive decline is one of the complications of type 2 diabetes (T2D). Intermittent fasting (IF) is a promising dietary intervention for alleviating T2D symptoms, but its protective effect on diabetes-driven cognitive dysfunction remains elusive. Here, we find that a 28-day IF regimen for diabetic mice improves behavioral impairment via a microbiota-metabolites-brain axis: IF enhances mitochondrial biogenesis and energy metabolism gene expression in hippocampus, re-structures the gut microbiota, and improves microbial metabolites that are related to cognitive function. Moreover, strong connections are observed between IF affected genes, microbiota and metabolites, as assessed by integrative modelling. Removing gut microbiota with antibiotics partly abolishes the neuroprotective effects of IF. Administration of 3-indolepropionic acid, serotonin, short chain fatty acids or tauroursodeoxycholic acid shows a similar effect to IF in terms of improving cognitive function. Together, our study purports the microbiota-metabolites-brain axis as a mechanism that can enable therapeutic strategies against metabolism-implicated cognitive pathophysiologies. Intermittent fasting (IF) has been shown beneficial in reducing metabolic diseases. Here, using a multi-omics approach in a T2D mouse model, the authors report that IF alters the composition of the gut microbiota and improves metabolic phenotypes that correlate with cognitive behavior.
Collapse
Affiliation(s)
- Zhigang Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China.
| | - Xiaoshuang Dai
- BGI Institute of Applied Agriculture, BGI-Shenzhen, Shenzhen, 518120, China
| | - Hongbo Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Renjie Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Yan Hui
- BGI Institute of Applied Agriculture, BGI-Shenzhen, Shenzhen, 518120, China.,Department of Food Science, University of Copenhagen, Copenhagen, 1958, Denmark
| | - Xin Jin
- BGI Institute of Applied Agriculture, BGI-Shenzhen, Shenzhen, 518120, China
| | - Wentong Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Luanfeng Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Qianxu Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Danna Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Jia Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Xintong Tan
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Bo Ren
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Xiaoning Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Tong Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Jiamin Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Junru Pan
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Tian Yuan
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Chuanqi Chu
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China
| | - Lei Lan
- BGI Institute of Applied Agriculture, BGI-Shenzhen, Shenzhen, 518120, China
| | - Fei Yin
- Center for Innovation in Brain Science and Department of Pharmacology, University of Arizona, Tucson, 85721, AZ, USA
| | - Enrique Cadenas
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, 90089, CA, USA
| | - Lin Shi
- Division Food and Nutrition Science, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, SE-412 96, Sweden.
| | - Shancen Zhao
- BGI Institute of Applied Agriculture, BGI-Shenzhen, Shenzhen, 518120, China.
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
126
|
Short-chain fatty acids accompanying changes in the gut microbiome contribute to the development of hypertension in patients with preeclampsia. Clin Sci (Lond) 2020; 134:289-302. [PMID: 31961431 DOI: 10.1042/cs20191253] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/15/2022]
Abstract
Preeclampsia (PE) is regarded as a pregnancy-associated hypertension disorder that is related to excessive inflammatory responses. Although the gut microbiota (GM) and short-chain fatty acids (SCFAs) have been related to hypertension, their effects on PE remain unknown. We determined the GM abundance and faecal SCFA levels by 16S ribosomal RNA (rRNA) sequencing and gas chromatography, respectively, using faecal samples from 27 patients with severe PE and 36 healthy, pregnant control subjects. We found that patients with PE had significantly decreased GM diversity and altered GM abundance. At the phylum level, patients with PE exhibited decreased abundance of Firmicutes albeit increased abundance of Proteobacteria; at the genus level, patients with PE had lower abundance of Blautia, Eubacterium_rectale, Eubacterium_hallii, Streptococcus, Bifidobacterium, Collinsella, Alistipes, and Subdoligranulum, albeit higher abundance of Enterobacter and Escherichia_Shigella. The faecal levels of butyric and valeric acids were significantly decreased in patients with PE and significantly correlated with the above-mentioned differential GM abundance. We predicted significantly increased abundance of the lipopolysaccharide (LPS)-synthesis pathway and significantly decreased abundance of the G protein-coupled receptor (GPCR) pathway in patients with PE, based on phylogenetic reconstruction of unobserved states (PICRUSt). Finally, we evaluated the effects of oral butyrate on LPS-induced hypertension in pregnant rats. We found that butyrate significantly reduced the blood pressure (BP) in these rats. In summary, we provide the first evidence linking GM dysbiosis and reduced faecal SCFA to PE and demonstrate that butyrate can directly regulate BP in vivo, suggesting its potential as a therapeutic agent for PE.
Collapse
|
127
|
Chen J, Wang Q, Wang A, Lin Z. Structural and Functional Characterization of the Gut Microbiota in Elderly Women With Migraine. Front Cell Infect Microbiol 2020; 9:470. [PMID: 32083024 PMCID: PMC7001586 DOI: 10.3389/fcimb.2019.00470] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 12/20/2019] [Indexed: 12/17/2022] Open
Abstract
Migraine is a very common, multifactorial, and recurrent central nervous system disorder that causes throbbing headache, photophobia, phonophobia, nausea, and disability. Migraine occurs more often in females, and its complex physiopathology is not yet fully understood. An increasing number of gastrointestinal disorders have been linked to the occurrence of migraine suggesting that gut microbiota might play a pivotal role in migraine through the gut–brain axis. In the present work, we performed a metagenome-wide association study (MWAS) to determine the relationship between gut microbiota and migraine by analyzing 108 shotgun-sequenced fecal samples obtained from elderly women who suffer from migraine and matched healthy controls. Notably, the alpha diversity was significantly decreased in the migraine group at species, genus, and Kyoto Encyclopedia of Genes and Genomes (KEGG) orthologous levels. Firmicutes, especially the “unfriendly” Clostridium spp., were significantly enriched in the migraine group. Conversely, the healthy controls held more beneficial microorganisms, such as Faecalibacterium prausnitzii, Bifidobacterium adolescentis, and Methanobrevibacter smithii. For functional modules, the migraine group was enriched in gut–brain modules (GBMs) including kynurenine degradation and γ-aminobutyric acid (GABA) synthesis. However, the healthy controls held higher gut metabolic modules (GMMs) including glycolysis, homoacetogenesis, and GBMs including quinolinic acid degradation and S-adenosyl methionine (SAM) synthesis. The differences in gut microbiota composition and function between the migraine and healthy groups provided new information as well as novel therapeutic targets and strategies for migraine treatment, which could help to improve the early diagnosis of the disease, as well as the long-term prognosis and the life quality of patients suffering from migraine.
Collapse
Affiliation(s)
- Juanjuan Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China.,BGI-Shenzhen, Shenzhen, China
| | - Qi Wang
- BGI-Shenzhen, Shenzhen, China.,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing, China
| | | | - Zhanglin Lin
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
128
|
Xu C, Cheng C, Zhang X, Peng J. Inclusion of Soluble Fiber in the Gestation Diet Changes the Gut Microbiota, Affects Plasma Propionate and Odd-Chain Fatty Acids Levels, and Improves Insulin Sensitivity in Sows. Int J Mol Sci 2020; 21:ijms21020635. [PMID: 31963640 PMCID: PMC7013540 DOI: 10.3390/ijms21020635] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/06/2020] [Accepted: 01/15/2020] [Indexed: 12/31/2022] Open
Abstract
The transition from pregnancy to lactation is characterized by a progressive decrease in insulin sensitivity. Propionate increases with dietary fiber consumption and has been shown to improve insulin sensitivity. Recent studies suggest that plasma odd-chain fatty acids [OCFAs; pentadecanoic acid (C15:0) and heptadecanoic acid (C17:0)] that inversely correlated with insulin resistance are synthesized endogenously from gut-derived propionate. The present study investigated the effects of soluble fiber during gestation on gut microbiota, plasma non-esterified fatty acids and insulin sensitivity in sows. Sows were allocated to either control or 2.0% guar gum plus pregelatinized waxy maize starch (SF) dietary treatment during gestation. The SF addition changes the structure and composition of gut microbiota in sows. Genus Eubacterium increased by SF addition may promote intestinal propionate production. Moreover, the dietary SF increased circulating levels of plasma OCFAs, especially C17:0. The SF-fed sows had a higher insulin sensitivity and a lower systemic inflammation level during perinatal period. Furthermore, the plasma C15:0 and C17:0 was negatively correlated with the area under curve of plasma glucose after meal and plasma interleukin-6. In conclusion, dietary SF improves insulin sensitivity and alleviates systemic inflammation in perinatal sows, potentially related to its stimulating effect on propionate and OCFAs production.
Collapse
Affiliation(s)
- Chuanhui Xu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (C.X.); (C.C.); (X.Z.)
| | - Chuanshang Cheng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (C.X.); (C.C.); (X.Z.)
- Key Laboratory of Animal Nutrition and Feed Science, Ministry of Agriculture and Rural Affairs, WENS Research Institute (Technology center), Yunfu 527300, China
| | - Xiu Zhang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (C.X.); (C.C.); (X.Z.)
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (C.X.); (C.C.); (X.Z.)
- The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan 430070, China
- Correspondence:
| |
Collapse
|
129
|
Wang J, He Y, Yu D, Jin L, Gong X, Zhang B. Perilla oil regulates intestinal microbiota and alleviates insulin resistance through the PI3K/AKT signaling pathway in type-2 diabetic KKAy mice. Food Chem Toxicol 2020; 135:110965. [DOI: 10.1016/j.fct.2019.110965] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 11/08/2019] [Accepted: 11/10/2019] [Indexed: 01/10/2023]
|
130
|
Lemoinne S, Kemgang A, Ben Belkacem K, Straube M, Jegou S, Corpechot C, Chazouillères O, Housset C, Sokol H. Fungi participate in the dysbiosis of gut microbiota in patients with primary sclerosing cholangitis. Gut 2020; 69:92-102. [PMID: 31003979 DOI: 10.1136/gutjnl-2018-317791] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/21/2019] [Accepted: 03/19/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Patients with primary sclerosing cholangitis (PSC) were previously shown to display a bacterial gut dysbiosis but fungal microbiota has never been examined in these patients. The aim of this study was to assess the fungal gut microbiota in patients with PSC. DESIGN We analysed the faecal microbiota of patients with PSC and concomitant IBD (n=27), patients with PSC and no IBD (n=22), patients with IBD and no PSC (n=33) and healthy subjects (n=30). Bacterial and fungal composition of the faecal microbiota was determined using 16S and ITS2 sequencing, respectively. RESULTS We found that patients with PSC harboured bacterial dysbiosis characterised by a decreased biodiversity, an altered composition and a decreased correlation network density. These alterations of the microbiota were associated with PSC, independently of IBD status. For the first time, we showed that patients with PSC displayed a fungal gut dysbiosis, characterised by a relative increase in biodiversity and an altered composition. Notably, we observed an increased proportion of Exophiala and a decreased proportion of Saccharomyces cerevisiae. Compared with patients with IBD and healthy subjects, the gut microbiota of patients with PSC exhibited a strong disruption in bacteria-fungi correlation network, suggesting an alteration in the interkingdom crosstalk. CONCLUSION This study demonstrates that bacteria and fungi contribute to gut dysbiosis in PSC.
Collapse
Affiliation(s)
- Sara Lemoinne
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris, France.,Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris, France.,Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis (MIVB-H), Department of Hepatology, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Astrid Kemgang
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris, France.,Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris, France.,Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis (MIVB-H), Department of Hepatology, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Karima Ben Belkacem
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris, France.,Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris, France.,Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis (MIVB-H), Department of Hepatology, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Marjolène Straube
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris, France.,Department of Gastroenterology, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Sarah Jegou
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris, France.,Department of Gastroenterology, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Christophe Corpechot
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris, France.,Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris, France.,Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis (MIVB-H), Department of Hepatology, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | | | - Olivier Chazouillères
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris, France.,Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris, France.,Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis (MIVB-H), Department of Hepatology, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Chantal Housset
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris, France.,Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, INSERM, Paris, France.,Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis (MIVB-H), Department of Hepatology, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Harry Sokol
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, INSERM, Paris, France.,Department of Gastroenterology, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.,UMR1319 Micalis, AgroParisTech, INRA, Jouy-en-Josas, France
| |
Collapse
|
131
|
Patterson E, Ryan PM, Wiley N, Carafa I, Sherwin E, Moloney G, Franciosi E, Mandal R, Wishart DS, Tuohy K, Ross RP, Cryan JF, Dinan TG, Stanton C. Gamma-aminobutyric acid-producing lactobacilli positively affect metabolism and depressive-like behaviour in a mouse model of metabolic syndrome. Sci Rep 2019; 9:16323. [PMID: 31704943 PMCID: PMC6841999 DOI: 10.1038/s41598-019-51781-x] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 09/20/2019] [Indexed: 12/22/2022] Open
Abstract
Metabolic and neuroactive metabolite production represents one of the mechanisms through which the gut microbiota can impact health. One such metabolite, gamma-aminobutyric acid (GABA), can modulate glucose homeostasis and alter behavioural patterns in the host. We previously demonstrated that oral administration of GABA-producing Lactobacillus brevis DPC6108 has the potential to increase levels of circulating insulin in healthy rats. Therefore, the objective of this study was to assess the efficacy of endogenous microbial GABA production in improving metabolic and behavioural outcomes in a mouse model of metabolic dysfunction. Diet-induced obese and metabolically dysfunctional mice received one of two GABA-producing strains, L. brevis DPC6108 or L. brevis DSM32386, daily for 12 weeks. After 8 and 10 weeks of intervention, the behavioural and metabolic profiles of the mice were respectively assessed. Intervention with both L. brevis strains attenuated several abnormalities associated with metabolic dysfunction, causing a reduction in the accumulation of mesenteric adipose tissue, increased insulin secretion following glucose challenge, improved plasma cholesterol clearance and reduced despair-like behaviour and basal corticosterone production during the forced swim test. Taken together, this exploratory dataset indicates that intervention with GABA-producing lactobacilli has the potential to improve metabolic and depressive- like behavioural abnormalities associated with metabolic syndrome in mice.
Collapse
Affiliation(s)
- E Patterson
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - P M Ryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - N Wiley
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - I Carafa
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland.,Department of Food Quality and Nutrition, Research and Innovation Centre-Fondazione Edmund Mach, San Michele all'Adige, Trento, Italy
| | - E Sherwin
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - G Moloney
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - E Franciosi
- Department of Food Quality and Nutrition, Research and Innovation Centre-Fondazione Edmund Mach, San Michele all'Adige, Trento, Italy
| | - R Mandal
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - D S Wishart
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.,Department of Computing Science, University of Alberta, Edmonton, Alberta, Canada.,National Institute for Nanotechnology, Edmonton, Alberta, Canada
| | - K Tuohy
- Department of Food Quality and Nutrition, Research and Innovation Centre-Fondazione Edmund Mach, San Michele all'Adige, Trento, Italy
| | - R P Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - J F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - T G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - C Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland. .,Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland.
| |
Collapse
|
132
|
Wosinska L, Cotter PD, O'Sullivan O, Guinane C. The Potential Impact of Probiotics on the Gut Microbiome of Athletes. Nutrients 2019; 11:E2270. [PMID: 31546638 PMCID: PMC6835687 DOI: 10.3390/nu11102270] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/05/2019] [Accepted: 09/19/2019] [Indexed: 12/19/2022] Open
Abstract
There is accumulating evidence that physical fitness influences the gut microbiome and as a result, promotes health. Indeed, exercise-induced alterations in the gut microbiome can influence health parameters crucial to athletic performance, specifically, immune function, lower susceptibility to infection, inflammatory response and tissue repair. Consequently, maintenance of a healthy gut microbiome is essential for an athlete's health, training and performance. This review explores the effect of exercise on the microbiome while also investigating the effect of probiotics on various potential consequences associated with over-training in athletes, as well as their associated health benefits.
Collapse
Affiliation(s)
- Laura Wosinska
- Department of Biological Sciences, Cork Institute of Technology, Bishopstown, T12 P928 Cork, Ireland.
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Cork, Ireland.
- APC Microbiome Ireland, T12 YT20 Cork, Ireland.
| | - Paul D Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Cork, Ireland.
- APC Microbiome Ireland, T12 YT20 Cork, Ireland.
| | - Orla O'Sullivan
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Cork, Ireland.
- APC Microbiome Ireland, T12 YT20 Cork, Ireland.
| | - Caitriona Guinane
- Department of Biological Sciences, Cork Institute of Technology, Bishopstown, T12 P928 Cork, Ireland.
| |
Collapse
|
133
|
Koopman N, Molinaro A, Nieuwdorp M, Holleboom AG. Review article: can bugs be drugs? The potential of probiotics and prebiotics as treatment for non-alcoholic fatty liver disease. Aliment Pharmacol Ther 2019; 50:628-639. [PMID: 31373710 DOI: 10.1111/apt.15416] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 05/06/2019] [Accepted: 06/23/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver condition. A major current research effort is ongoing to find potential strategies to treat NAFLD-non-alcoholic steatohepatitis (NASH), with special attention to the gut microbiota. Multiple animal studies and pilot clinical trials are assessing different gut microbiota modulating strategies such as faecal microbiota transplantation, antibiotics, probiotics, prebiotics and synbiotics. AIM To review the role of microbiota in NAFLD-NASH and determine whether pro- and prebiotics have potential as treatment METHODS: Information was obtained from critically reviewing literature on PubMed on targeting the gut microbiota in NAFLD. Search terms included NAFLD, NASH, non-alcoholic fatty liver disease, steatohepatitis; combined with microbiome, microbiota, gut bacteria, probiotics and prebiotics. RESULTS Animal studies and the first emerging studies in humans show promising results for both the common probiotics Lactobacillus, Bifidobacterium and Streptococci as for short chain fatty acid (SCFA) butyrate-producing bacteria. Also, prebiotics have positive effects on different mechanisms underlying NAFLD-NASH. CONCLUSIONS The most promising strategies thus far developed to alter the microbiome in NAFLD-NASH are probiotics and prebiotics. However, pre- and probiotic treatment of NAFLD-NASH is relatively new and still under development. Actual understanding of the involved mechanisms is lacking and changes in the intestinal microbiota composition after treatment are rarely measured. Furthermore, large clinical trials with comparative endpoints are unavailable. Personalised treatment based on metagenomics gut microbiota analysis will probably be part of the future diagnosis and treatment of NAFLD-NASH.
Collapse
Affiliation(s)
- Nienke Koopman
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Antonio Molinaro
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Max Nieuwdorp
- Vascular Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Adriaan G Holleboom
- Vascular Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
134
|
Tilg H, Zmora N, Adolph TE, Elinav E. The intestinal microbiota fuelling metabolic inflammation. Nat Rev Immunol 2019; 20:40-54. [DOI: 10.1038/s41577-019-0198-4] [Citation(s) in RCA: 377] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2019] [Indexed: 02/06/2023]
|
135
|
Novik G, Savich V. Beneficial microbiota. Probiotics and pharmaceutical products in functional nutrition and medicine. Microbes Infect 2019; 22:8-18. [PMID: 31233819 DOI: 10.1016/j.micinf.2019.06.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 12/12/2022]
Abstract
The article is mainly devoted to such representatives of gut microbiota as lactic acid bacteria and bifidobacteria, with minor accent on less frequently used or new probiotic microorganisms. Positive effects in treatment and prevention of diseases by different microbial groups, their metabolites and mechanisms of action, management and market of probiotic products are considered.
Collapse
Affiliation(s)
- Galina Novik
- Belarusian Collection of Microorganisms, Institute of Microbiology, National Academy of Sciences of Belarus, 2 Academician V.F. Kuprevich Street, 220141 Minsk, the Republic of Belarus.
| | - Victoria Savich
- Belarusian Collection of Microorganisms, Institute of Microbiology, National Academy of Sciences of Belarus, 2 Academician V.F. Kuprevich Street, 220141 Minsk, the Republic of Belarus
| |
Collapse
|
136
|
Yuan T, Chu C, Shi R, Cui T, Zhang X, Zhao Y, Shi X, Hui Y, Pan J, Qian R, Dai X, Liu Z, Liu X. ApoE-Dependent Protective Effects of Sesamol on High-Fat Diet-Induced Behavioral Disorders: Regulation of the Microbiome-Gut-Brain Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:6190-6201. [PMID: 31117496 DOI: 10.1021/acs.jafc.9b01436] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Sesamol, an antioxidant lignan from sesame oil, possesses neuroprotective bioactivities. The present work was aimed to elucidate the systemic protective effects of sesamol on cognitive deficits and to determine the possible link between gut and brain. Wildtype and ApoE-/- mice were treated with a high-fat diet and sesamol (0.05%, w/v, in drinking water) for 10 weeks. Behavioral tests including Morris-water maze, Y-maze, and elevated plus maze tests indicated that sesamol could only improve cognitive deficits and anxiety behaviors in wildtype. Consistently, sesamol improved synapse ultrastructure and inhibited Aβ accumulation in an ApoE-dependent manner. Moreover, sesamol prevented dietary-induced gut barrier damages and systemic inflammation. Sesamol also reshaped gut microbiome and improved the generation of microbial metabolites short-chain fatty acids. To summarize, this study revealed that the possible mechanism of neuroprotective effects of sesamol might be ApoE-dependent, and its beneficial effects on gut microbiota/metabolites could be translated into neurodegenerative diseases treatment.
Collapse
Affiliation(s)
- Tian Yuan
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| | - Chuanqi Chu
- School of Food Science and Technology , Jiangnan University , Wuxi , China
| | - Rubing Shi
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| | - Tianlin Cui
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| | - Xinglin Zhang
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| | - Yihang Zhao
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| | - Xu Shi
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| | - Yan Hui
- Department of Food Science , University of Copenhagen , Copenhagen , Denmark
| | - Junru Pan
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| | - Rui Qian
- Food Analysis and Development Center , Beijing ZhiYunDa Technology, Co., LTD. , Beijing , China
| | - Xiaoshuang Dai
- BGI Institute of Applied Agriculture , BGI-Shenzhen , Shenzhen , China
| | - Zhigang Liu
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| | - Xuebo Liu
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| |
Collapse
|
137
|
|
138
|
Almeida D, Machado D, Andrade JC, Mendo S, Gomes AM, Freitas AC. Evolving trends in next-generation probiotics: a 5W1H perspective. Crit Rev Food Sci Nutr 2019; 60:1783-1796. [PMID: 31062600 DOI: 10.1080/10408398.2019.1599812] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In recent years, scientific community has been gathering increasingly more insight on the dynamics that are at play in metabolic and inflammatory disorders. These rapidly growing conditions are reaching epidemic proportions, bringing clinicians and researcher's new challenges. The specific roles and modulating properties that beneficial/probiotic bacteria hold in the context of the gut ecosystem seem to be key to avert these inflammatory and diet-related disorders. Faecalibacterium prausnitzii, Akkermansia muciniphila and Eubacterium hallii have been identified as candidates for next generation probiotics (NGPs) with exciting potential for the prevention and treatment of such of dysbiosis-associated diseases. The challenges of these non-conventional native gut bacteria lie mainly on their extreme sensitivity to O2 traces. If these strains are to be used successfully in food, supplements or drugs they need to be stable and active in humans. In the present review, we present an overall perspective of the most updated scientific literature on the newly called NGPs through the 5W1H (What, Why, Who, Where, When, and How) method, an innovative and attractive problem-solving approach that provides the reader an effective understanding of the issue at hand.
Collapse
Affiliation(s)
- Diana Almeida
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal.,Department of Biology and CESAM, University of Aveiro, Aveiro, Portugal
| | - Daniela Machado
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - José Carlos Andrade
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Gandra PRD, Portugal
| | - Sónia Mendo
- Department of Biology and CESAM, University of Aveiro, Aveiro, Portugal
| | - Ana Maria Gomes
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Ana Cristina Freitas
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| |
Collapse
|
139
|
Faecal Microbiota Are Related to Insulin Sensitivity and Secretion in Overweight or Obese Adults. J Clin Med 2019; 8:jcm8040452. [PMID: 30987356 PMCID: PMC6518043 DOI: 10.3390/jcm8040452] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/18/2019] [Accepted: 03/27/2019] [Indexed: 12/18/2022] Open
Abstract
Emerging evidence suggests a role for the gut microbiota in glucose metabolism and diabetes. Few studies have examined the associations between the faecal microbiome and insulin sensitivity and secretion using gold-standard methods in high-risk populations prior to diabetes onset. We investigated the relationships between faecal microbiota composition (16S rRNA sequencing) and gold-standard measures of insulin sensitivity (hyperinsulinaemic-euglycaemic clamp) and insulin secretion (intravenous glucose tolerance test) in 38 overweight or obese otherwise healthy individuals. Genus Clostridium was positively associated with insulin sensitivity, and genera Dialister and Phascolarctobacterium were related to both insulin sensitivity and secretion. Insulin sensitivity was associated with a higher abundance of Phascolarctobacterium and lower abundance of Dialister. Those with higher insulin secretion had a higher abundance of Dialister and lower abundance of Bifidobacterium, compared to those with lower insulin secretion. Body mass index (BMI) was positively correlated with Streptococcus abundance whereas Coprococcus abundance was negatively correlated to BMI and percent body fat. These results suggest that faecal microbiota is related to insulin sensitivity and secretion in overweight or obese adults. These correlations are distinct although partially overlapping, suggesting different pathophysiological pathways. Our findings can inform future trials aiming to manipulate gut microbiome to improve insulin sensitivity and secretion and prevent type 2 diabetes.
Collapse
|
140
|
Douillard FP, de Vos WM. Biotechnology of health-promoting bacteria. Biotechnol Adv 2019; 37:107369. [PMID: 30876799 DOI: 10.1016/j.biotechadv.2019.03.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/15/2019] [Accepted: 03/11/2019] [Indexed: 12/20/2022]
Abstract
Over the last decade, there has been an increasing scientific and public interest in bacteria that may positively contribute to human gut health and well-being. This interest is reflected by the ever-increasing number of developed functional food products containing health-promoting bacteria and reaching the market place as well as by the growing revenue and profits of notably bacterial supplements worldwide. Traditionally, the origin of probiotic-marketed bacteria was limited to a rather small number of bacterial species that mostly belong to lactic acid bacteria and bifidobacteria. Intensifying research efforts on the human gut microbiome offered novel insights into the role of human gut microbiota in health and disease, while also providing a deep and increasingly comprehensive understanding of the bacterial communities present in this complex ecosystem and their interactions with the gut-liver-brain axis. This resulted in rational and systematic approaches to select novel health-promoting bacteria or to engineer existing bacteria with enhanced probiotic properties. In parallel, the field of gut microbiomics developed into a fertile framework for the identification, isolation and characterization of a phylogenetically diverse array of health-promoting bacterial species, also called next-generation therapeutic bacteria. The present review will address these developments with specific attention for the selection and improvement of a selected number of health-promoting bacterial species and strains that are extensively studied or hold promise for future food or pharma product development.
Collapse
Affiliation(s)
- François P Douillard
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Willem M de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands.
| |
Collapse
|
141
|
Zhang Q, Xiao X, Zheng J, Li M, Yu M, Ping F, Wang T, Wang X. Influence of Maternal Inulin-Type Prebiotic Intervention on Glucose Metabolism and Gut Microbiota in the Offspring of C57BL Mice. Front Endocrinol (Lausanne) 2019; 10:675. [PMID: 31632351 PMCID: PMC6779716 DOI: 10.3389/fendo.2019.00675] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/17/2019] [Indexed: 01/14/2023] Open
Abstract
Scope: Maternal obesity leads to glucose intolerance in the offspring. Changes in the gut microbiota are being increasingly implicated in the pathogenesis of diabetes. We hypothesized that inulin intervention during gestation and lactation improves glucose metabolism disorders in mouse offspring from high-fat diet (HD)-fed dams. Procedures: Female C57BL mice were fed a control diet or HD for 4 weeks before mating. After mating, pregnant mice were randomly divided into three groups through gestation and lactation: control diet (CD) group, HD group, and HD treated with inulin (HD-inulin) group. At weaning, glucose metabolic status was assessed. Gut microbial DNA from offspring cecal contents was isolated and processed for metagenomic shotgun sequencing, and taxonomic and functional profiling were performed. Results: Offspring from dams in the HD-inulin groups demonstrated reduced fasting blood glucose, decreased blood glucose area under the curve during the oral glucose tolerance test, and reduced fasting serum insulin and HOMA-IR compared to offspring from dams in the HD group. Nineteen differentially abundant bacterial species were identified between the HD-inulin and HD groups. The HD-inulin group displayed significantly greater abundances of Bacteroides_acidifaciens, Eubacterium_sp_CAG_786, Clostridium_sp_CAG_343, and Bifidobacterium_breve species and lower abundances of Oscillibacter_sp_1_3, Ruminococcus_gnavus_CAG_126, and Bacteroides_massiliensis species. Differentially abundant bacterial species among the three groups were involved in 38 metabolic pathways, including several glucose and lipid metabolism pathways. Conclusion: Our results show that early inulin intervention in HD-fed mouse dams moderates offspring glucose metabolism and gut dysbiosis.
Collapse
|
142
|
Grabherr F, Grander C, Effenberger M, Adolph TE, Tilg H. Gut Dysfunction and Non-alcoholic Fatty Liver Disease. Front Endocrinol (Lausanne) 2019; 10:611. [PMID: 31555219 PMCID: PMC6742694 DOI: 10.3389/fendo.2019.00611] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 08/22/2019] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as one of the leading liver diseases worldwide. NAFLD is characterized by hepatic steatosis and may progress to an inflammatory condition termed non-alcoholic steatohepatitis (NASH), liver cirrhosis, and hepatocellular carcinoma. It became evident in the last years that NAFLD pathophysiology is complex and involves diverse immunological and metabolic pathways. An association between intestinal signals (e.g., derived from the gut microbiota) and the development of obesity and its metabolic consequences such as NAFLD are increasingly recognized. Pre-clinical studies have shown that germ-free mice are protected against obesity and hepatic steatosis. Several human studies from the past years have demonstrated that NAFLD contains a disease-specific gut microbiome signature. Controlled studies propose that certain bacteria with rather pro-inflammatory features such as Proteobacteria or Escherichia coli are dominantly present in these patients. In contrast, rather protective bacteria such as Faecalibacterium prausnitzii are decreased in NAFLD patients. Furthermore, various bacterial metabolites and microbiota-generated secondary bile acids are involved in NAFLD-associated metabolic dysfunction. Although these findings are exciting, research currently lack evidence that interference at the level of the gut microbiome is beneficial for these diseases. Further preclinical and clinical studies are needed to advance this aspect of NAFLD research and to support the notion that the intestinal microbiota is indeed of major relevance in this disorder.
Collapse
|
143
|
Predicting the associations between microbes and diseases by integrating multiple data sources and path-based HeteSim scores. Neurocomputing 2019. [DOI: 10.1016/j.neucom.2018.09.054] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
144
|
Wan Y, Tong W, Zhou R, Li J, Yuan J, Wang F, Li D. Habitual animal fat consumption in shaping gut microbiota and microbial metabolites. Food Funct 2019; 10:7973-7982. [PMID: 31776537 DOI: 10.1039/c9fo01490j] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A diet with higher animal-based fat consumption is likely to be associated with moderately unfavorable impacts on gut microbial diversity, community, and regulation of fecal short-chain fatty acids.
Collapse
Affiliation(s)
- Yi Wan
- Institute of Nutrition and Health
- Qingdao University
- Qingdao
- China
- Department of Food Science and Nutrition
| | - Wenfeng Tong
- Department of Food Science and Nutrition
- Zhejiang University
- Hangzhou
- China
| | - Renke Zhou
- Department of Food Science and Nutrition
- Zhejiang University
- Hangzhou
- China
| | - Jie Li
- Department of Nutrition
- Chinese People's Liberation Army General Hospital
- Beijing
- China
| | - Jihong Yuan
- Department of Nutrition
- Chinese People's Liberation Army General Hospital
- Beijing
- China
| | - Fenglei Wang
- Department of Food Science and Nutrition
- Zhejiang University
- Hangzhou
- China
| | - Duo Li
- Institute of Nutrition and Health
- Qingdao University
- Qingdao
- China
| |
Collapse
|
145
|
Shetty SA, Zuffa S, Bui TPN, Aalvink S, Smidt H, De Vos WM. Reclassification of Eubacterium hallii as Anaerobutyricum hallii gen. nov., comb. nov., and description of Anaerobutyricum soehngenii sp. nov., a butyrate and propionate-producing bacterium from infant faeces. Int J Syst Evol Microbiol 2018; 68:3741-3746. [PMID: 30351260 DOI: 10.1099/ijsem.0.003041] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
A bacterial strain designated L2-7T, phylogenetically related to Eubacterium hallii DSM 3353T, was previously isolated from infant faeces. The complete genome of strain L2-7T contains eight copies of the 16S rRNA gene with only 98.0-98.5 % similarity to the 16S rRNA gene of the previously described type strain E. hallii. The next closest validly described species is Anaerostipes hadrus DSM 3319T (90.7 % 16S rRNA gene similarity). A polyphasic taxonomic approach showed strain L2-7T to be a novel species, related to type strain E. hallii DSM 3353T. The experimentally observed DNA-DNA hybridization value between strain L2-7T and E. hallii DSM 3353T was 26.25 %, close to that calculated from the genomes (34.3 %). The G+C content of the chromosomal DNA of strain L2-7T was 38.6 mol%. The major fatty acids were C16 : 0, C16 : 1cis9 and a component with summed feature 10 (C18 : 1c11/t9/t6c). Strain L2-7T had higher amounts of C16 : 0 (30.6 %) compared to E. hallii DSM 3353T (19.5 %) and its membrane contained phosphatidylglycerol and phosphatidylethanolamine, which were not detected in E. hallii DSM 3353T. Furthermore, 16S rRNA gene phylogenetic analysis advocates that E. hallii DSM 3353T is misclassified, and its reclassification as a member of the family Lachnospiraceae is necessary. Using a polyphasic approach, we propose that E. hallii (=DSM 3353T=ATCC 27751T) be reclassified as the type strain of a novel genus Anaerobutyricum sp. nov., comb. nov. and we propose that strain L2-7T should be classified as a novel species, Anaerobutyricum soehngenii sp. nov. The type strain is L2-7T (=DSM 17630T=KCTC 15707T).
Collapse
Affiliation(s)
- Sudarshan A Shetty
- 1Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, Building 124, 6708 WE Wageningen, The Netherlands
| | - Simone Zuffa
- 1Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, Building 124, 6708 WE Wageningen, The Netherlands
| | - Thi Phuong Nam Bui
- 1Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, Building 124, 6708 WE Wageningen, The Netherlands
| | - Steven Aalvink
- 1Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, Building 124, 6708 WE Wageningen, The Netherlands
| | - Hauke Smidt
- 1Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, Building 124, 6708 WE Wageningen, The Netherlands
| | - Willem M De Vos
- 1Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, Building 124, 6708 WE Wageningen, The Netherlands.,2Department of Veterinary Biosciences, Division of Microbiology and Epidemiology, University of Helsinki, Helsinki, Finland.,3RPU Immunobiology, Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
146
|
Cinek O, Kramna L, Mazankova K, Odeh R, Alassaf A, Ibekwe MU, Ahmadov G, Elmahi BME, Mekki H, Lebl J, Abdullah MA. The bacteriome at the onset of type 1 diabetes: A study from four geographically distant African and Asian countries. Diabetes Res Clin Pract 2018; 144:51-62. [PMID: 30121305 DOI: 10.1016/j.diabres.2018.08.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/22/2018] [Accepted: 08/13/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Gut bacteriome profiling studies in type 1 diabetes (T1D) to date are mostly limited to populations of Europe, with two studies from China and one study each from Mexico and the USA. We therefore sought to characterize the stool bacteriome in children after onset of T1D along with age- and place-matched control subjects from four geographically distant African and Asian countries. METHODS Samples were collected from 73 children and adolescents shortly after T1D onset (Azerbaijan 19, Jordan 20, Nigeria 14, Sudan 20) and 104 matched control subjects of similar age and locale. Genotyping of major T1D susceptibility genes was performed using saliva or blood samples. The bacteriome was profiled by next-generation sequencing of 16S rDNA. Negative binomial regression was used to model associations, with adjustment for the matched structure of the study. RESULTS A significant positive association with T1D was noted for the genus Escherichia (class Gammaproteobacteria, phylum Proteobacteria), whereas Eubacterium and Roseburia, two genera of class Clostridia, phylum Firmicutes, were inversely associated with T1D. We also confirmed a previously observed inverse association with Clostridium clusters IV or XIVa. No associations were noted for richness, evenness, or enterotypes. CONCLUSIONS Based on our results, some type of distortion of the gut bacteriome appears to be a global feature of T1D, and our findings for four distant populations add new candidates to the existing list of bacteria. It remains to be established whether the observed associations are markers or causative factors.
Collapse
Affiliation(s)
- Ondrej Cinek
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, V Uvalu 84, Prague 5, Czech Republic.
| | - Lenka Kramna
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, V Uvalu 84, Prague 5, Czech Republic.
| | - Karla Mazankova
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, V Uvalu 84, Prague 5, Czech Republic.
| | - Rasha Odeh
- Department of Pediatrics, School of Medicine, University of Jordan, Amman, Jordan.
| | - Abeer Alassaf
- Department of Pediatrics, School of Medicine, University of Jordan, Amman, Jordan.
| | - MaryAnn Ugochi Ibekwe
- Department of Pediatrics, Federal Teaching Hospital Abakaliki, Ebonyi State University, Abakaliki, Nigeria.
| | - Gunduz Ahmadov
- Endocrine Centre Baku, Str. I. Hashimov 4A, AZ1114 Baku, Azerbaijan.
| | - Bashir Mukhtar Elwasila Elmahi
- Department of Paediatrics and Child Health, University of Khartoum, Faculty of Medicine, Khartoum, Sudan; Sudan Childhood Diabetes Center, Khartoum, Sudan.
| | - Hanan Mekki
- Department of Paediatrics and Child Health, University of Khartoum, Faculty of Medicine, Khartoum, Sudan.
| | - Jan Lebl
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, V Uvalu 84, Prague 5, Czech Republic.
| | - Mohammed Ahmed Abdullah
- Department of Paediatrics and Child Health, University of Khartoum, Faculty of Medicine, Khartoum, Sudan; Sudan Childhood Diabetes Center, Khartoum, Sudan.
| |
Collapse
|
147
|
Daliri EBM, Tango CN, Lee BH, Oh DH. Human microbiome restoration and safety. Int J Med Microbiol 2018; 308:487-497. [DOI: 10.1016/j.ijmm.2018.05.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 04/04/2018] [Accepted: 05/07/2018] [Indexed: 02/07/2023] Open
|
148
|
Martin G, Kolida S, Marchesi JR, Want E, Sidaway JE, Swann JR. In Vitro Modeling of Bile Acid Processing by the Human Fecal Microbiota. Front Microbiol 2018; 9:1153. [PMID: 29922256 PMCID: PMC5996868 DOI: 10.3389/fmicb.2018.01153] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 05/14/2018] [Indexed: 12/22/2022] Open
Abstract
Bile acids, the products of concerted host and gut bacterial metabolism, have important signaling functions within the mammalian metabolic system and a key role in digestion. Given the complexity of the mega-variate bacterial community residing in the gastrointestinal tract, studying associations between individual bacterial genera and bile acid processing remains a challenge. Here, we present a novel in vitro approach to determine the bacterial genera associated with the metabolism of different primary bile acids and their potential to contribute to inter-individual variation in this processing. Anaerobic, pH-controlled batch cultures were inoculated with human fecal microbiota and treated with individual conjugated primary bile acids (500 μg/ml) to serve as the sole substrate for 24 h. Samples were collected throughout the experiment (0, 5, 10, and 24 h) and the bacterial composition was determined by 16S rRNA gene sequencing and the bile acid signatures were characterized using a targeted ultra-performance liquid chromatography-mass spectrometry (UPLC-MS) approach. Data fusion techniques were used to identify statistical bacterial-metabolic linkages. An increase in gut bacteria associated bile acids was observed over 24 h with variation in the rate of bile acid metabolism across the volunteers (n = 7). Correlation analysis identified a significant association between the Gemmiger genus and the deconjugation of glycine conjugated bile acids while the deconjugation of taurocholic acid was associated with bacteria from the Eubacterium and Ruminococcus genera. A positive correlation between Dorea and deoxycholic acid production suggest a potential role for this genus in cholic acid dehydroxylation. A slower deconjugation of taurocholic acid was observed in individuals with a greater abundance of Parasutterella and Akkermansia. This work demonstrates the utility of integrating compositional (metataxonomics) and functional (metabonomics) systems biology approaches, coupled to in vitro model systems, to study the biochemical capabilities of bacteria within complex ecosystems. Characterizing the dynamic interactions between the gut microbiota and the bile acid pool enables a greater understanding of how variation in the gut microbiota influences host bile acid signatures, their associated functions and their implications for health.
Collapse
Affiliation(s)
- Glynn Martin
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Sofia Kolida
- OptiBiotix Health PLC, Innovation Centre, York, United Kingdom
| | - Julian R Marchesi
- Division of Integrative Systems Medicine and Digestive Diseases, Imperial College London, London, United Kingdom.,Centre for Digestive and Gut Health, Imperial College London, London, United Kingdom.,School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Elizabeth Want
- Division of Integrative Systems Medicine and Digestive Diseases, Imperial College London, London, United Kingdom
| | | | - Jonathan R Swann
- Division of Integrative Systems Medicine and Digestive Diseases, Imperial College London, London, United Kingdom
| |
Collapse
|
149
|
Bircher L, Geirnaert A, Hammes F, Lacroix C, Schwab C. Effect of cryopreservation and lyophilization on viability and growth of strict anaerobic human gut microbes. Microb Biotechnol 2018; 11:721-733. [PMID: 29663668 PMCID: PMC6011992 DOI: 10.1111/1751-7915.13265] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/22/2018] [Accepted: 03/08/2018] [Indexed: 01/22/2023] Open
Abstract
Strict anaerobic gut microbes have been suggested as ‘next‐generation probiotics’ for treating several intestinal disorders. The development of preservation techniques is of major importance for therapeutic application. This study investigated cryopreservation (−80°C) and lyophilization survival and storage stability (4°C for 3 months) of the strict anaerobic gut microbes Bacteroides thetaiotaomicron, Faecalibacterium prausnitzii, Roseburia intestinalis, Anaerostipes caccae, Eubacterium hallii and Blautia obeum. To improve preservation survival, protectants sucrose and inulin (both 5% w/v) were added for lyophilization and were also combined with glycerol (15% v/v) for cryopreservation. Bacterial fitness, evaluated by maximum growth rate and lag phase, viability and membrane integrity were determined using a standardized growth assay and by flow cytometry as markers for preservation resistance. Lyophilization was more detrimental to viability and fitness than cryopreservation, but led to better storage stability. Adding sucrose and inulin enhanced viability and the proportion of intact cells during lyophilization of all strains. Viability of protectant‐free B. thetaiotaomicron, A. caccae and F. prausnitzii was above 50% after cryopreservation and storage and increased to above 80% if protectants were present. The addition of glycerol, sucrose and inulin strongly enhanced the viability of B. obeum, E. hallii and R. intestinalis from 0.03–2% in protectant‐free cultures to 11–37%. This is the first study that quantitatively compared the effect of cryopreservation and lyophilization and the addition of selected protectants on viability and fitness of six strict anaerobic gut microbes. Our results suggest that efficiency of protectants is process‐ and species‐specific.
Collapse
Affiliation(s)
- Lea Bircher
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zürich, Schmelzbergstrasse 7, 8092, Zürich, Switzerland
| | - Annelies Geirnaert
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zürich, Schmelzbergstrasse 7, 8092, Zürich, Switzerland
| | | | - Christophe Lacroix
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zürich, Schmelzbergstrasse 7, 8092, Zürich, Switzerland
| | - Clarissa Schwab
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zürich, Schmelzbergstrasse 7, 8092, Zürich, Switzerland
| |
Collapse
|
150
|
Romaní-Pérez M, Agusti A, Sanz Y. Innovation in microbiome-based strategies for promoting metabolic health. Curr Opin Clin Nutr Metab Care 2017; 20:484-491. [PMID: 28862999 DOI: 10.1097/mco.0000000000000419] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Update on the development of microbiome-based interventions and dietary supplements to combat obesity and related comorbidities, which are leading causes of global mortality. RECENT FINDINGS The role of intestinal dysbiosis, partly resulting from unhealthy diets, in the development of obesity and metabolic disorders, is well documented by recent translational research. Human experimental trials with whole-faecal transplants are ongoing, and their results will be crucial as proof of concept that interventions intended to modulate the microbiome composition and function could be alternatives for the management of obesity and related comorbidities. Potential next-generation probiotic bacteria (Akkermansia, Bacteroides spp., Eubacterium halli) and microbiota-derived molecules (e.g. membrane proteins, short-chain fatty acids) are being evaluated in preclinical and clinical trials to promote the development of innovative dietary supplements. The fact that live or inactivated bacteria and their products can regulate pathways that increase energy expenditure, and reduce energy intake, and absorption and systemic inflammation make them attractive research targets from a nutritional and clinical perspective. SUMMARY Understanding which are the beneficial bacteria and their bioactive products is helping us to envisage innovative microbiome-based dietary interventions to tackle obesity. Advances will likely result from future refinements of these strategies according to the individual's microbiome configuration and its particular response to interventions, thereby progressing towards personalized nutrition.
Collapse
Affiliation(s)
- Marina Romaní-Pérez
- Microbial Ecology, Nutrition & Health Research Unit. Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | | | | |
Collapse
|