551
|
Paraiso IL, Plagmann LS, Yang L, Zielke R, Gombart AF, Maier CS, Sikora AE, Blakemore PR, Stevens JF. Reductive Metabolism of Xanthohumol and 8-Prenylnaringenin by the Intestinal Bacterium Eubacterium ramulus. Mol Nutr Food Res 2018; 63:e1800923. [PMID: 30471194 DOI: 10.1002/mnfr.201800923] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/14/2018] [Indexed: 12/16/2022]
Abstract
SCOPE The intestinal microbiota transforms a wide range of available substrates, including polyphenols. Microbial catabolites of polyphenols can contribute in significant ways to the health-promoting properties of their parent polyphenols. This work aims to identify intestinal metabolites of xanthohumol (XN), a prenylated flavonoid found in hops (Humulus lupulus) and beer, as well as to identify pathways of metabolism of XN in the gut. METHODS AND RESULTS To investigate intestinal metabolism, XN and related prenylated flavonoids, isoxanthohumol (IX), and 8-prenylnaringenin (8PN) were added to growing cultures of intestinal bacteria, Eubacterium ramulus and E. limosum. Liquid chromatography coupled with mass spectrometry was used to identify metabolites of the flavonoids from the cultures. The metabolic capacity of E. limosum appears to be limited to O-demethylation. Evidence from the study indicates that E. ramulus hydrogenates XN to form α,β-dihydroxanthohumol (DXN) and metabolizes the potent phytoestrogen 8PN into the chalcones, O-desmethylxanthohumol (DMX) and O-desmethyl-α,β-dihydroxanthohumol (DDXN). CONCLUSION Microbial metabolism is likely to affect both activity and toxicity of XN and derivatives. This study along with others highlights that attention should be focused on metabolites, in particular, products of intestinal microbial metabolism.
Collapse
Affiliation(s)
- Ines L Paraiso
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, 97331, USA.,Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA
| | - Layhna S Plagmann
- Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA.,Department of Chemistry, Oregon State University, Corvallis, OR, 97331, USA
| | - Liping Yang
- Department of Chemistry, Oregon State University, Corvallis, OR, 97331, USA
| | - Ryszard Zielke
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Adrian F Gombart
- Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA.,Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, 97331, USA
| | - Claudia S Maier
- Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA.,Department of Chemistry, Oregon State University, Corvallis, OR, 97331, USA
| | - Aleksandra E Sikora
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, 97331, USA.,Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | - Paul R Blakemore
- Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA.,Department of Chemistry, Oregon State University, Corvallis, OR, 97331, USA
| | - Jan F Stevens
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, 97331, USA.,Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA
| |
Collapse
|
552
|
Abstract
The intestinal microbiome plays a crucial role in the development of the immune system and regulation of immune responses. Many factors influence the composition of the infant intestinal microbiome and therefore the development and function of the immune system. This, in turn, may alter the risk of subsequent allergies, autoimmune diseases and other adverse health outcomes. Here, we review factors that influence the composition of the intestinal microbiome during the first year of life, including birth location, gestational age, delivery mode, feeding method, hospitalization, antibiotic or probiotic intake and living conditions. Understanding how the early intestinal microbiome is established and how this is perturbed provides many opportunities for interventions to improve health.
Collapse
|
553
|
Onori R, Marín M, Rodríguez-Sánchez B, Oliver C, Muñoz P, Bouza E, Alcalá L. First isolation of Skermanella aerolata from a human sample. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2018; 31:552-553. [PMID: 30364926 PMCID: PMC6254487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Raffaella Onori
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid
| | - Mercedes Marín
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid
| | - Belén Rodríguez-Sánchez
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid
| | - Cristina Oliver
- Department of Obstetrics and Gynecology, Hospital General Universitario Gregorio Marañón, Madrid
| | - Patricia Muñoz
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid,CIBER de Enfermedades Respiratorias (CIBERES CB06/06/0058), Spain,Medicine Department, School of Medicine, Universidad Complutense de Madrid
| | - Emilio Bouza
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid,CIBER de Enfermedades Respiratorias (CIBERES CB06/06/0058), Spain,Medicine Department, School of Medicine, Universidad Complutense de Madrid
| | - Luis Alcalá
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid,CIBER de Enfermedades Respiratorias (CIBERES CB06/06/0058), Spain
| |
Collapse
|
554
|
Bao J, Zheng H, Wang Y, Zheng X, He L, Qi W, Wang T, Guo B, Guo G, Zhang Z, Zhang W, Li J, McManus DP. Echinococcus granulosus Infection Results in an Increase in Eisenbergiella and Parabacteroides Genera in the Gut of Mice. Front Microbiol 2018; 9:2890. [PMID: 30555437 PMCID: PMC6281689 DOI: 10.3389/fmicb.2018.02890] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 11/12/2018] [Indexed: 12/15/2022] Open
Abstract
Cystic echinococcosis (CE) is a chronic infectious disease caused by Echinococcus granulosus. To confirm whether the infection impacts on the gut microbiota, we established a mouse model of E. granulosus infection in this study whereby BALB/c mice were infected with micro-cysts of E. granulosus. After 4 months of infection, fecal samples were collected for high-throughput sequencing of the hypervariable regions of the 16S rRNA gene. Sequence analysis revealed a total of 13,353 operational taxonomic units (OTUs) with only 40.6% of the OTUs having genera reference information and 101 of the OTUs were significantly increased in infected mice. Bioinformatics analysis showed that the common core microbiota were not significantly changed at family level. However, two genera (Eisenbergiella and Parabacteroides) were enriched in the infected mice (P AMOV A < 0.05) at genus level. Functional analysis indicated that seven pathways were altered in the E. granulosus Infection Group compared with the Uninfected Group. Spearman correlation analysis showed strong correlations of IgG, IgG1 and IgG2a with nine major genera. E. granulosus cyst infection may change the gut microbiota which may be associated with metabolic pathways.
Collapse
Affiliation(s)
- Jianling Bao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,College of Public Health, Xinjiang Medical University, Urumqi, China
| | - Huajun Zheng
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai, China.,Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Yuezhu Wang
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai, China
| | - Xueting Zheng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Li He
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Wenjing Qi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Tian Wang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Baoping Guo
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Gang Guo
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zhaoxia Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Wenbao Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,College of Public Health, Xinjiang Medical University, Urumqi, China
| | - Jun Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Donald P McManus
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
555
|
Moen AEF, Lindstrøm JC, Tannæs TM, Vatn S, Ricanek P, Vatn MH, Jahnsen J. The prevalence and transcriptional activity of the mucosal microbiota of ulcerative colitis patients. Sci Rep 2018; 8:17278. [PMID: 30467421 PMCID: PMC6250705 DOI: 10.1038/s41598-018-35243-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/01/2018] [Indexed: 12/16/2022] Open
Abstract
Active microbes likely have larger impact on gut health status compared to inactive or dormant microbes. We investigate the composition of active and total mucosal microbiota of treatment-naïve ulcerative colitis (UC) patients to determine the microbial picture at the start-up phase of disease, using both a 16S rRNA transcript and gene amplicon sequencing. DNA and RNA were isolated from the same mucosal colonic biopsies. Our aim was to identify active microbial members of the microbiota in early stages of disease and reveal which members are present, but do not act as major players. We demonstrated differences in active and total microbiota of UC patients when comparing inflamed to non-inflamed tissue. Several taxa, among them the Proteobacteria phyla and families therein, revealed lower transcriptional activity despite a high presence. The Bifidobacteriaceae family of the Actinobacteria phylum showed lower abundance in the active microbiota, although no difference in presence was detected. The most abundant microbiota members of the inflamed tissue in UC patients were not the most active. Knowledge of active members of microbiota in UC patients could enhance our understanding of disease etiology. The active microbial community composition did not deviate from the total when comparing UC patients to non-IBD controls.
Collapse
Affiliation(s)
- Aina E Fossum Moen
- Department of Clinical Molecular Biology (EpiGen), Division of Medicine, Akershus University Hospital, Lørenskog, and University of Oslo, Oslo, Norway
| | - Jonas Christoffer Lindstrøm
- Health Services Research Unit, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
| | - Tone Møller Tannæs
- Department of Clinical Molecular Biology (EpiGen), Division of Medicine, Akershus University Hospital, Lørenskog, and University of Oslo, Oslo, Norway.
| | - Simen Vatn
- Department of Gastroenterology, Division of Medicine, Akershus University Hospital, Lørenskog, and University of Oslo, Oslo, Norway
| | - Petr Ricanek
- Department of Gastroenterology, Division of Medicine, Akershus University Hospital, Lørenskog, and University of Oslo, Oslo, Norway
| | - Morten H Vatn
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
| | - Jørgen Jahnsen
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Division of Medicine, Akershus University Hospital, Lørenskog, and University of Oslo, Oslo, Norway
| | | |
Collapse
|
556
|
Possible Prevention of Diabetes with a Gluten-Free Diet. Nutrients 2018; 10:nu10111746. [PMID: 30428550 PMCID: PMC6266002 DOI: 10.3390/nu10111746] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/06/2018] [Accepted: 11/07/2018] [Indexed: 02/07/2023] Open
Abstract
Gluten seems a potentially important determinant in type 1 diabetes (T1D) and type 2 diabetes (T2D). Intake of gluten, a major component of wheat, rye, and barley, affects the microbiota and increases the intestinal permeability. Moreover, studies have demonstrated that gluten peptides, after crossing the intestinal barrier, lead to a more inflammatory milieu. Gluten peptides enter the pancreas where they affect the morphology and might induce beta-cell stress by enhancing glucose- and palmitate-stimulated insulin secretion. Interestingly, animal studies and a human study have demonstrated that a gluten-free (GF) diet during pregnancy reduces the risk of T1D. Evidence regarding the role of a GF diet in T2D is less clear. Some studies have linked intake of a GF diet to reduced obesity and T2D and suggested a role in reducing leptin- and insulin-resistance and increasing beta-cell volume. The current knowledge indicates that gluten, among many environmental factors, may be an aetiopathogenic factors for development of T1D and T2D. However, human intervention trials are needed to confirm this and the proposed mechanisms.
Collapse
|
557
|
Silverman JD, Durand HK, Bloom RJ, Mukherjee S, David LA. Dynamic linear models guide design and analysis of microbiota studies within artificial human guts. MICROBIOME 2018; 6:202. [PMID: 30419949 PMCID: PMC6233358 DOI: 10.1186/s40168-018-0584-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 10/23/2018] [Indexed: 05/19/2023]
Abstract
BACKGROUND Artificial gut models provide unique opportunities to study human-associated microbiota. Outstanding questions for these models' fundamental biology include the timescales on which microbiota vary and the factors that drive such change. Answering these questions though requires overcoming analytical obstacles like estimating the effects of technical variation on observed microbiota dynamics, as well as the lack of appropriate benchmark datasets. RESULTS To address these obstacles, we created a modeling framework based on multinomial logistic-normal dynamic linear models (MALLARDs) and performed dense longitudinal sampling of four replicate artificial human guts over the course of 1 month. The resulting analyses revealed how the ratio of biological variation to technical variation from sample processing depends on sampling frequency. In particular, we find that at hourly sampling frequencies, 76% of observed variation could be ascribed to technical sources, which could also skew the observed covariation between taxa. We also found that the artificial guts demonstrated replicable trajectories even after a recovery from a transient feed disruption. Additionally, we observed irregular sub-daily oscillatory dynamics associated with the bacterial family Enterobacteriaceae within all four replicate vessels. CONCLUSIONS Our analyses suggest that, beyond variation due to sequence counting, technical variation from sample processing can obscure temporal variation from biological sources in artificial gut studies. Our analyses also supported hypotheses that human gut microbiota fluctuates on sub-daily timescales in the absence of a host and that microbiota can follow replicable trajectories in the presence of environmental driving forces. Finally, multiple aspects of our approach are generalizable and could ultimately be used to facilitate the design and analysis of longitudinal microbiota studies in vivo.
Collapse
Affiliation(s)
- Justin D. Silverman
- Program in Computational Biology and Bioinformatics, Duke University, CIEMAS, Room 2171, 101 Science Drive, Box 3382, Durham, NC 27708 USA
- Medical Scientist Training Program, Duke University, Durham, NC 27708 USA
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708 USA
| | - Heather K. Durand
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708 USA
| | - Rachael J. Bloom
- University Program in Genetics and Genomics, Duke University, Durham, NC 27708 USA
| | - Sayan Mukherjee
- Program in Computational Biology and Bioinformatics, Duke University, CIEMAS, Room 2171, 101 Science Drive, Box 3382, Durham, NC 27708 USA
- Departments of Statistical Science, Mathematics, Computer Science, Biostatistics & Bioinformatics, Duke University, Durham, NC 27708 USA
| | - Lawrence A. David
- Program in Computational Biology and Bioinformatics, Duke University, CIEMAS, Room 2171, 101 Science Drive, Box 3382, Durham, NC 27708 USA
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708 USA
- University Program in Genetics and Genomics, Duke University, Durham, NC 27708 USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708 USA
| |
Collapse
|
558
|
Anisimova E, Yarullina D. Characterization of Erythromycin and Tetracycline Resistance in Lactobacillus fermentum Strains. Int J Microbiol 2018; 2018:3912326. [PMID: 30534155 PMCID: PMC6252201 DOI: 10.1155/2018/3912326] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/10/2018] [Accepted: 10/17/2018] [Indexed: 11/23/2022] Open
Abstract
Lactobacillus fermentum colonizing gastrointestinal and urogenital tracts of humans and animals is widely used in manufacturing of fermented products and as probiotics. These bacteria may function as vehicles of antibiotic resistance genes, which can be transferred to pathogenic bacteria. Therefore, monitoring and control of transmissible antibiotic resistance determinants in these microorganisms is necessary to approve their safety status. The aim of this study was to characterize erythromycin and tetracycline resistance of L. fermentum isolates and to estimate the potential transfer of resistance genes from lactobacilli to the other Gram-positive and Gram-negative bacteria. Among six L. fermentum strains isolated from human feces and commercial dairy products, five strains demonstrated phenotypic resistance to tetracycline. PCR screening for antibiotic resistance determinants revealed plasmid-located tetracycline resistance genes tet(K) and tet(M) in all strains and erythromycin resistance genes erm(B) in the chromosome of L. fermentum 5-1 and erm(C) in the plasmid of L. fermentum 3-4. All tested lactobacilli lacked conjugative transposon Tn916 and were not able to transfer tetracycline resistance genes to Staphylococcus aureus, Staphylococcus epidermidis, Listeria monocytogenes, Acinetobacter baumannii, Citrobacter freundii, and Escherichia coli by filter mating. Staphylococcus haemolyticus did not accept erythromycin resistance genes from corresponding Lactobacillus strains. Thus, in the present study, L. fermentum was not implicated in the spread of erythromycin and tetracycline resistance, but still these strains pose the threat to the environment and human health because they harbored erythromycin and tetracycline resistance genes in their plasmids and therefore should not be used in foods and probiotics.
Collapse
Affiliation(s)
- Elizaveta Anisimova
- Department of Microbiology, Kazan Federal University, Kremlevskaya Str. 18, Kazan 420008, Russia
| | - Dina Yarullina
- Department of Microbiology, Kazan Federal University, Kremlevskaya Str. 18, Kazan 420008, Russia
| |
Collapse
|
559
|
Bozzi Cionci N, Baffoni L, Gaggìa F, Di Gioia D. Therapeutic Microbiology: The Role of Bifidobacterium breve as Food Supplement for the Prevention/Treatment of Paediatric Diseases. Nutrients 2018; 10:E1723. [PMID: 30423810 PMCID: PMC6265827 DOI: 10.3390/nu10111723] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/05/2018] [Accepted: 11/08/2018] [Indexed: 02/07/2023] Open
Abstract
The human intestinal microbiota, establishing a symbiotic relationship with the host, plays a significant role for human health. It is also well known that a disease status is frequently characterized by a dysbiotic condition of the gut microbiota. A probiotic treatment can represent an alternative therapy for enteric disorders and human pathologies not apparently linked to the gastrointestinal tract. Among bifidobacteria, strains of the species Bifidobacterium breve are widely used in paediatrics. B. breve is the dominant species in the gut of breast-fed infants and it has also been isolated from human milk. It has antimicrobial activity against human pathogens, it does not possess transmissible antibiotic resistance traits, it is not cytotoxic and it has immuno-stimulating abilities. This review describes the applications of B. breve strains mainly for the prevention/treatment of paediatric pathologies. The target pathologies range from widespread gut diseases, including diarrhoea and infant colics, to celiac disease, obesity, allergic and neurological disorders. Moreover, B. breve strains are used for the prevention of side infections in preterm newborns and during antibiotic treatments or chemotherapy. With this documentation, we hope to increase knowledge on this species to boost the interest in the emerging discipline known as "therapeutic microbiology".
Collapse
Affiliation(s)
- Nicole Bozzi Cionci
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum-Università di Bologna, Viale Fanin 42, 40127 Bologna, Italy.
| | - Loredana Baffoni
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum-Università di Bologna, Viale Fanin 42, 40127 Bologna, Italy.
| | - Francesca Gaggìa
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum-Università di Bologna, Viale Fanin 42, 40127 Bologna, Italy.
| | - Diana Di Gioia
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum-Università di Bologna, Viale Fanin 42, 40127 Bologna, Italy.
| |
Collapse
|
560
|
Tetz G, Tetz V. Tetz's theory and law of longevity. Theory Biosci 2018; 137:145-154. [PMID: 29978435 PMCID: PMC6208853 DOI: 10.1007/s12064-018-0267-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 06/29/2018] [Indexed: 11/30/2022]
Abstract
Here, we present new theory and law of longevity intended to evaluate fundamental factors that control lifespan. This theory is based on the fact that genes affecting host organism longevity are represented by subpopulations: genes of host eukaryotic cells, commensal microbiota, and non-living genetic elements. Based on Tetz's theory of longevity, we propose that lifespan and aging are defined by the accumulation of alterations over all genes of macroorganism and microbiome and the non-living genetic elements associated with them. Tetz's law of longevity states that longevity is limited by the accumulation of alterations to the limiting value that is not compatible with life. Based on theory and law, we also propose a novel model to calculate several parameters, including the rate of aging and the remaining lifespan of individuals. We suggest that this theory and model have explanatory and predictive potential to eukaryotic organisms, allowing the influence of diseases, medication, and medical procedures to be re-examined in relation to longevity. Such estimates also provide a framework to evaluate new fundamental aspects that control aging and lifespan.
Collapse
Affiliation(s)
- George Tetz
- Human Microbiology Institute, 423 West 127 Street, New York, NY, 10027, USA.
| | - Victor Tetz
- Human Microbiology Institute, 423 West 127 Street, New York, NY, 10027, USA
| |
Collapse
|
561
|
Jenkins TP, Formenti F, Castro C, Piubelli C, Perandin F, Buonfrate D, Otranto D, Griffin JL, Krause L, Bisoffi Z, Cantacessi C. A comprehensive analysis of the faecal microbiome and metabolome of Strongyloides stercoralis infected volunteers from a non-endemic area. Sci Rep 2018; 8:15651. [PMID: 30353019 PMCID: PMC6199319 DOI: 10.1038/s41598-018-33937-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/05/2018] [Indexed: 02/08/2023] Open
Abstract
Data from recent studies support the hypothesis that infections by human gastrointestinal (GI) helminths impact, directly and/or indirectly, on the composition of the host gut microbial flora. However, to the best of our knowledge, these studies have been conducted in helminth-endemic areas with multi-helminth infections and/or in volunteers with underlying gut disorders. Therefore, in this study, we explore the impact of natural mono-infections by the human parasite Strongyloides stercoralis on the faecal microbiota and metabolic profiles of a cohort of human volunteers from a non-endemic area of northern Italy (S+), pre- and post-anthelmintic treatment, and compare the findings with data obtained from a cohort of uninfected controls from the same geographical area (S-). Analyses of bacterial 16S rRNA high-throughput sequencing data revealed increased microbial alpha diversity and decreased beta diversity in the faecal microbial profiles of S+ subjects compared to S-. Furthermore, significant differences in the abundance of several bacterial taxa were observed between samples from S+ and S- subjects, and between S+ samples collected pre- and post-anthelmintic treatment. Faecal metabolite analysis detected marked increases in the abundance of selected amino acids in S+ subjects, and of short chain fatty acids in S- subjects. Overall, our work adds valuable knowledge to current understanding of parasite-microbiota associations and will assist future mechanistic studies aimed to unravel the causality of these relationships.
Collapse
Affiliation(s)
- Timothy P Jenkins
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Fabio Formenti
- Centre for Tropical Diseases, IRCCS Sacro Cuore-Don Calabria Hospital, Negrar, Verona, Italy
| | - Cecilia Castro
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Chiara Piubelli
- Centre for Tropical Diseases, IRCCS Sacro Cuore-Don Calabria Hospital, Negrar, Verona, Italy
| | - Francesca Perandin
- Centre for Tropical Diseases, IRCCS Sacro Cuore-Don Calabria Hospital, Negrar, Verona, Italy
| | - Dora Buonfrate
- Centre for Tropical Diseases, IRCCS Sacro Cuore-Don Calabria Hospital, Negrar, Verona, Italy
| | - Domenico Otranto
- Department of Veterinary Medicine, University of Bari, Valenzano, Italy
| | - Julian L Griffin
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Lutz Krause
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Zeno Bisoffi
- Centre for Tropical Diseases, IRCCS Sacro Cuore-Don Calabria Hospital, Negrar, Verona, Italy
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Cinzia Cantacessi
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
562
|
Abstract
PURPOSE OF REVIEW Obesity and diabetes are worldwide epidemics. There is also a growing body of evidence relating the gut microbiome composition to insulin resistance. The purpose of this review is to delineate the studies linking gut microbiota to obesity, metabolic syndrome, and diabetes. RECENT FINDINGS Animal studies as well as proof of concept studies using fecal transplantation demonstrate the pivotal role of the gut microbiota in regulating insulin resistance states and inflammation. While we still need to standardize methodologies to study the microbiome, there is an abundance of evidence pointing to the link between gut microbiome, inflammation, and insulin resistance, and future studies should be aimed at identifying unifying mechanisms.
Collapse
Affiliation(s)
- Xinpu Chen
- Department of Pathology & Immunology, Baylor College of Medicine and Texas Children's Hospital, 6621 Fannin Street, Houston, TX, 77030, USA
| | - Sridevi Devaraj
- Department of Pathology & Immunology, Baylor College of Medicine and Texas Children's Hospital, 6621 Fannin Street, Houston, TX, 77030, USA.
| |
Collapse
|
563
|
Impact of Diet-Modulated Butyrate Production on Intestinal Barrier Function and Inflammation. Nutrients 2018; 10:nu10101499. [PMID: 30322146 PMCID: PMC6213552 DOI: 10.3390/nu10101499] [Citation(s) in RCA: 342] [Impact Index Per Article: 48.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/01/2018] [Accepted: 10/11/2018] [Indexed: 02/07/2023] Open
Abstract
A major challenge in affluent societies is the increase in disorders related to gut and metabolic health. Chronic over nutrition by unhealthy foods high in energy, fat, and sugar, and low in dietary fibre is a key environmental factor responsible for this development, which may cause local and systemic inflammation. A low intake of dietary fibre is a limiting factor for maintaining a viable and diverse microbiota and production of short-chain fatty acids in the gut. A suppressed production of butyrate is crucial, as this short-chain fatty acid (SCFA) can play a key role not only in colonic health and function but also at the systemic level. At both sites, the mode of action is through mediation of signalling pathways involving nuclear NF-κB and inhibition of histone deacetylase. The intake and composition of dietary fibre modulate production of butyrate in the large intestine. While butyrate production is easily adjustable it is more variable how it influences gut barrier function and inflammatory markers in the gut and periphery. The effect of butyrate seems generally to be more consistent and positive on inflammatory markers related to the gut than on inflammatory markers in the peripheral tissue. This discrepancy may be explained by differences in butyrate concentrations in the gut compared with the much lower concentration at more remote sites.
Collapse
|
564
|
Microbiota and Derived Parameters in Fecal Samples of Infants with Non-IgE Cow's Milk Protein Allergy under a Restricted Diet. Nutrients 2018; 10:nu10101481. [PMID: 30314304 PMCID: PMC6213916 DOI: 10.3390/nu10101481] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/05/2018] [Accepted: 10/06/2018] [Indexed: 12/26/2022] Open
Abstract
Cow’s milk protein allergy (CMPA) is the most common food allergy in infancy. Non-IgE mediated (NIM) forms are little studied and the responsible mechanisms of tolerance acquisition remain obscure. Our aim was to study the intestinal microbiota and related parameters in the fecal samples of infants with NIM-CMPA, to establish potential links between type of formula substitutes, microbiota, and desensitization. Seventeen infants between one and two years old, diagnosed with NIM-CMPA, were recruited. They were all on an exclusion diet for six months, consuming different therapeutic protein hydrolysates. After this period, stool samples were obtained and tolerance development was evaluated by oral challenges. A control group of 10 age-matched healthy infants on an unrestricted diet were included in the study. Microbiota composition, short-chain fatty acids, calprotectin, and transforming growth factor (TGF)-β1 levels were determined in fecal samples from both groups. Infants with NIM-CMPA that consumed vegetable protein-based formulas presented microbiota colonization patterns different from those fed with an extensively hydrolyzed formula. Differences in microbiota composition and fecal parameters between NIM-CMPA and healthy infants were observed. Non-allergic infants showed a significantly higher proportion of Bacteroides compared to infants with NIM-CMPA. The type of protein hydrolysate was found to determine gut microbiota colonization and influence food allergy resolution in NIM-CMPA cases.
Collapse
|
565
|
Feng Y, Stams AJM, Sánchez-Andrea I, de Vos WM. Eubacterium maltosivorans sp. nov., a novel human intestinal acetogenic and butyrogenic bacterium with a versatile metabolism. Int J Syst Evol Microbiol 2018; 68:3546-3550. [PMID: 30285910 DOI: 10.1099/ijsem.0.003028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A novel anaerobic, non-spore-forming bacterium was isolated from a faecal sample of a healthy adult. The isolate, designated strain YIT, was cultured in a basal liquid medium under a gas phase of H2/CO2 supplemented with yeast extract (0.1 g l-1). Cells of strain YIT were short rods (0.4-0.7×2.0-2.5 µm), appearing singly or in pairs, and stained Gram-positive. Catalase activity and gelatin hydrolysis were positive while oxidase activity, indole formation, urease activity and aesculin hydrolysis were negative. Growth was observed within a temperature range of 20-45 °C (optimum, 35-37 °C), and a pH range of 5.0-8.0 (optimum pH 7.0-7.5). Doubling time was 2.3 h when grown with glucose at pH 7.2 and 37 °C. Besides acetogenic growth, the isolate was able to ferment a large range of monomeric sugars with acetate and butyrate as the main end products. Strain YIT did not show respiratory growth with sulfate, sulfite, thiosulfate or nitrate as electron acceptors. The major cellular fatty acids of the isolate were C16 : 0 and C18 : 0. The genomic DNA G+C content was 47.8 mol%. Strain YIT is affiliated to the genus Eubacterium, sharing highest levels of 16S rRNA gene similarity with Eubacterium limosum ATCC 8486T (97.3 %), Eubacterium callanderi DSM 3662T (97.5 %), Eubacterium aggregans DSM 12183T (94.4 %) and Eubacterium barkeri DSM 1223T (94.8 %). Considering its physiological and phylogenetic characteristics, strain YIT represents a novel species within the genus Eubacterium, for which the name Eubacterium maltosivorans sp. nov. is proposed. The type strain is YIT (=DSM 105863T=JCM 32297T).
Collapse
Affiliation(s)
- Yuan Feng
- 1Laboratory of Microbiology, Wageningen University, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Alfons J M Stams
- 2IBB - Institute for Biotechnology and Bioengineering, Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
- 1Laboratory of Microbiology, Wageningen University, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Irene Sánchez-Andrea
- 1Laboratory of Microbiology, Wageningen University, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Willem M de Vos
- 3RPU Immunobiology, Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
- 1Laboratory of Microbiology, Wageningen University, Stippeneng 4, 6708 WE Wageningen, The Netherlands
| |
Collapse
|
566
|
Trujillo-de Santiago G, Lobo-Zegers MJ, Montes-Fonseca SL, Zhang YS, Alvarez MM. Gut-microbiota-on-a-chip: an enabling field for physiological research. MICROPHYSIOLOGICAL SYSTEMS 2018; 2:7. [PMID: 33954286 PMCID: PMC8096182 DOI: 10.21037/mps.2018.09.01] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Overwhelming scientific evidence today confirms that the gut microbiota is a central player in human health. Knowledge about interactions between human gut microbiota and human health has evolved rapidly in the last decade, based on experimental work involving analysis of human fecal samples or animal models (mainly rodents). A more detailed and cost-effective description of this interplay is now being enabled by the use of in vitro systems (i.e., gut-microbiota-on-chip systems) that recapitulate key aspects of the interaction between microbiota and human cells. Here, we review recent examples of the design and use of pioneering on-chip platforms for the study of the cross-talk between representative members of human microbiota and human microtissues. In these systems, the combined use of state-of-the-art microfluidics, biomaterials, cell culture techniques, classical microbiology, and a touch of genetic expression profiling have converged for the development of gut-on-chip platforms capable of recreating key features of the interplay between human microbiota and host human tissues. We foresee that the integration of novel microfabrication techniques and stem cell technologies will further accelerate the development of more complex and physiologically relevant microbiota-on-chip platforms. In turn, this will foster the faster acquisition of knowledge regarding human microbiota and will enable important advances in the understanding of how to control or prevent disease.
Collapse
Affiliation(s)
- Grissel Trujillo-de Santiago
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey
- Departamento de Mecatrónica e Ingeniería Eléctrica, Campus Monterrey, CP 64849, Monterrey, Nuevo León, México
| | - Matías José Lobo-Zegers
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey
- Departamento de Mecatrónica e Ingeniería Eléctrica, Campus Monterrey, CP 64849, Monterrey, Nuevo León, México
| | - Silvia Lorena Montes-Fonseca
- Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, CP 31300, Chihuahua, México
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA, USA
| | | |
Collapse
|
567
|
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
|
568
|
On Controls in Ancient Microbiome Studies, and Microbial Resilience in Ancient Samples. Genes (Basel) 2018; 9:genes9100471. [PMID: 30262781 PMCID: PMC6210261 DOI: 10.3390/genes9100471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 11/28/2022] Open
Abstract
In the following comment, we reply to Eisenhofer and Weyrich’s letter “Proper authentication of ancient DNA is still essential” responding to the article “Gut Microbiome and Putative Resistome of Inca and Italian Nobility Mummies” by Santiago-Rodriguez et al. One of the concerns raised was the possibility that the patterns noted in the gut microbiome of pre-Inca/Inca and Italian nobility mummies were due to contamination of the blank control. When examining the blank controls and filtering the operational taxonomic units (OTUs) present in the blank controls, and further performing in-silico contamination analyses, we noticed very similar patterns as those previously reported. We also discuss controls in ancient microbiome studies, and aspects of microbial resilience in ancient samples.
Collapse
|
569
|
Braune A, Blaut M. Catenibacillus scindens gen. nov., sp. nov., a C-deglycosylating human intestinal representative of the Lachnospiraceae. Int J Syst Evol Microbiol 2018; 68:3356-3361. [PMID: 30179152 DOI: 10.1099/ijsem.0.003001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
An anaerobic Gram-stain-positive, non-spore-forming and non-motile bacterium isolated from the human gut, designated CG19-1T, capable of cleaving aromatic C-glucosides was characterized using a polyphasic taxonomic approach. Major fermentation products of this asaccharolytic organism were acetate and butyrate when grown on a complex medium. Growth of strain CG19-1T was stimulated by glucose or pyruvate. Growth inhibition was observed in the presence of several phenolic acids including ferulic acid, which nevertheless was reduced to dihydroferulic acid. Strain CG19-1T contained peptidoglycan type A4β l-Orn-d-Asp. The major cellular fatty acids were C16 : 0 and C18 : 1ω9c. The genomic DNA G+C content was 47.1 mol%. Based on its 16S rRNA gene sequence, strain CG19-1T is a member of the Lachnospiraceae. However, sequence identity to other Lachnospiraceae species with validly published names is approximately 93.0 % with Frisingicoccus caecimuris being the most closely related species according to phylogenetic analysis. Based on these findings, it is proposed to create a novel genus, Catenibacillus, and a novel species, Catenibacillus scindens, with the type strain CG19-1T (=DSM 106146T=CCUG 71490T).
Collapse
Affiliation(s)
- Annett Braune
- Department of Gastrointestinal Microbiology, German Institute of Human Nutrition Potsdam-Rehbruecke, D-14558 Nuthetal, Germany
| | - Michael Blaut
- Department of Gastrointestinal Microbiology, German Institute of Human Nutrition Potsdam-Rehbruecke, D-14558 Nuthetal, Germany
| |
Collapse
|
570
|
Teixeira C, Prykhodko O, Alminger M, Fåk Hållenius F, Nyman M. Barley Products of Different Fiber Composition Selectively Change Microbiota Composition in Rats. Mol Nutr Food Res 2018; 62:e1701023. [PMID: 30035373 PMCID: PMC6175208 DOI: 10.1002/mnfr.201701023] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 06/23/2018] [Indexed: 11/25/2022]
Abstract
Scope Several dietary fiber properties are suggested to be important for the profiling of the microbiota composition, but those characteristics are rather unclear. Whether different physico‐chemical properties of barley dietary fiber influence the gut microbiota composition is investigated. Methods and results Seven diets containing equal amounts of dietary fiber from barley malts, brewer's spent grain (BSG), and barley extracts, resulting in varying amounts of β‐glucan, soluble arabinoxylan, and insoluble arabinoxylan in the diets were given to conventional rats. Malts increased microbiota alpha diversity more than BSG and the extracts. The intake of soluble arabinoxylan was related to Akkermansia and propionic acid formation in the cecum of rats, whereas β‐glucan and/or insoluble arabinoxylan were attributed to some potentially butyrate‐producing bacteria (e.g., Lactobacillus, Blautia, and Allobaculum). Conclusion This study demonstrates that there is a potential to stimulate butyrate‐ and propionate‐producing bacteria in the cecum of rats with malt products of specific fiber properties. Moreover, BSG, a by product from beer production, added to malt can possibly be used to further modulate the microbiota composition, toward a higher butyric acid formation. A complex mixture of fiber as in the malts is of greater importance for microbiota diversity than purer fiber extracts.
Collapse
Affiliation(s)
- Cristina Teixeira
- Food for Health Science Centre, Kemicentrum, Lund University, SE-221 00, Lund, Sweden.,Food and Nutrition Science, Biology and Biological Engineering, Chalmers University of Technology, SE-412 96, Göteborg, Sweden
| | - Olena Prykhodko
- Food for Health Science Centre, Kemicentrum, Lund University, SE-221 00, Lund, Sweden.,Food Technology, Engineering and Nutrition, Kemicentrum, Lund University, SE-221 00, Lund, Sweden
| | - Marie Alminger
- Food and Nutrition Science, Biology and Biological Engineering, Chalmers University of Technology, SE-412 96, Göteborg, Sweden
| | - Frida Fåk Hållenius
- Food for Health Science Centre, Kemicentrum, Lund University, SE-221 00, Lund, Sweden.,Food Technology, Engineering and Nutrition, Kemicentrum, Lund University, SE-221 00, Lund, Sweden
| | - Margareta Nyman
- Food for Health Science Centre, Kemicentrum, Lund University, SE-221 00, Lund, Sweden.,Food Technology, Engineering and Nutrition, Kemicentrum, Lund University, SE-221 00, Lund, Sweden
| |
Collapse
|
571
|
Korpela K, de Vos WM. Early life colonization of the human gut: microbes matter everywhere. Curr Opin Microbiol 2018; 44:70-78. [DOI: 10.1016/j.mib.2018.06.003] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/27/2018] [Accepted: 06/27/2018] [Indexed: 12/15/2022]
|
572
|
Wasimuddin, Brändel SD, Tschapka M, Page R, Rasche A, Corman VM, Drosten C, Sommer S. Astrovirus infections induce age-dependent dysbiosis in gut microbiomes of bats. ISME JOURNAL 2018; 12:2883-2893. [PMID: 30061706 DOI: 10.1038/s41396-018-0239-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/05/2018] [Accepted: 06/30/2018] [Indexed: 02/08/2023]
Abstract
Astroviruses (AstV) are a major cause of diarrhoea in children. Interestingly, some wildlife species, including bats, remain phenotypically asymptomatic after infection. Disease symptoms, however, may only be less visible in bats and enteric viruses may indeed perturb their gut microbial communities. Gut microbiomes represent an important driver of immune defence mechanisms but potential effects of enteric virus-host microbiome interactions are largely unexplored. Using bats as a natural model system, we show that AstV-infections affect the gut microbiome, with the strength of the effect depending on host age. The gut microbial α- and β-diversity and the predicted microbial functional orthologs decreased in young bats but surprisingly increased in adult AstV + bats. The abundance of bacterial taxa characteristic for healthy microbiomes was strongly reduced in young AstV+ bats, possibly attributable to their immature immune system. Regardless of age, pathogen-containing genera exhibited negative interactions with several commensal taxa and increased after AstV-infection, leading to pathobiont-like shifts in the gut microbiome of all infected bats. Thus, in apparently healthy bats, AstV-infections disturb gut bacterial homeostasis, possibly increasing previously suppressed health risks by promoting co-infections. If similar processes are present in humans, the effects of enteric virus infections might have longer-term impacts extending beyond the directly observed symptoms.
Collapse
Affiliation(s)
- Wasimuddin
- Institute of Evolutionary Ecology and Conservation Genomics, University of Ulm, Albert-Einstein Allee 11, Ulm, D-89069, Germany
| | - Stefan Dominik Brändel
- Institute of Evolutionary Ecology and Conservation Genomics, University of Ulm, Albert-Einstein Allee 11, Ulm, D-89069, Germany.,Smithsonian Tropical Research Institute, Ancon, Apartado, Balboa, Panama, 0843-03092, Republic of Panama
| | - Marco Tschapka
- Institute of Evolutionary Ecology and Conservation Genomics, University of Ulm, Albert-Einstein Allee 11, Ulm, D-89069, Germany.,Smithsonian Tropical Research Institute, Ancon, Apartado, Balboa, Panama, 0843-03092, Republic of Panama
| | - Rachel Page
- Smithsonian Tropical Research Institute, Ancon, Apartado, Balboa, Panama, 0843-03092, Republic of Panama
| | - Andrea Rasche
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate member of Free University, Humboldt-University and Berlin Institute of Health, Berlin, Germany.,German Centre for Infection Research (DZIF), Berlin, Germany
| | - Victor M Corman
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate member of Free University, Humboldt-University and Berlin Institute of Health, Berlin, Germany.,German Centre for Infection Research (DZIF), Berlin, Germany
| | - Christian Drosten
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate member of Free University, Humboldt-University and Berlin Institute of Health, Berlin, Germany.,German Centre for Infection Research (DZIF), Berlin, Germany
| | - Simone Sommer
- Institute of Evolutionary Ecology and Conservation Genomics, University of Ulm, Albert-Einstein Allee 11, Ulm, D-89069, Germany.
| |
Collapse
|
573
|
Hiippala K, Jouhten H, Ronkainen A, Hartikainen A, Kainulainen V, Jalanka J, Satokari R. The Potential of Gut Commensals in Reinforcing Intestinal Barrier Function and Alleviating Inflammation. Nutrients 2018; 10:nu10080988. [PMID: 30060606 PMCID: PMC6116138 DOI: 10.3390/nu10080988] [Citation(s) in RCA: 394] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 07/19/2018] [Accepted: 07/27/2018] [Indexed: 02/06/2023] Open
Abstract
The intestinal microbiota, composed of pro- and anti-inflammatory microbes, has an essential role in maintaining gut homeostasis and functionality. An overly hygienic lifestyle, consumption of processed and fiber-poor foods, or antibiotics are major factors modulating the microbiota and possibly leading to longstanding dysbiosis. Dysbiotic microbiota is characterized to have altered composition, reduced diversity and stability, as well as increased levels of lipopolysaccharide-containing, proinflammatory bacteria. Specific commensal species as novel probiotics, so-called next-generation probiotics, could restore the intestinal health by means of attenuating inflammation and strengthening the epithelial barrier. In this review we summarize the latest findings considering the beneficial effects of the promising commensals across all major intestinal phyla. These include the already well-known bifidobacteria, which use extracellular structures or secreted substances to promote intestinal health. Faecalibacterium prausnitzii, Roseburia intestinalis, and Eubacterium hallii metabolize dietary fibers as major short-chain fatty acid producers providing energy sources for enterocytes and achieving anti-inflammatory effects in the gut. Akkermansia muciniphila exerts beneficial action in metabolic diseases and fortifies the barrier function. The health-promoting effects of Bacteroides species are relatively recently discovered with the findings of excreted immunomodulatory molecules. These promising, unconventional probiotics could be a part of biotherapeutic strategies in the future.
Collapse
Affiliation(s)
- Kaisa Hiippala
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Hanne Jouhten
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Aki Ronkainen
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Anna Hartikainen
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Veera Kainulainen
- Pharmacology, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Jonna Jalanka
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Reetta Satokari
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| |
Collapse
|
574
|
Klychnikov OI, Shamorkina TM, Weeks SD, van Leeuwen HC, Corver J, Drijfhout JW, van Veelen PA, Sluchanko NN, Strelkov SV, Hensbergen PJ. Discovery of a new Pro-Pro endopeptidase, PPEP-2, provides mechanistic insights into the differences in substrate specificity within the PPEP family. J Biol Chem 2018; 293:11154-11165. [PMID: 29794027 DOI: 10.1074/jbc.ra118.003244] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/22/2018] [Indexed: 12/20/2022] Open
Abstract
Pro-Pro endopeptidases (PPEPs) belong to a recently discovered family of proteases capable of hydrolyzing a Pro-Pro bond. The first member from the bacterial pathogen Clostridium difficile (PPEP-1) cleaves two C. difficile cell-surface proteins involved in adhesion, one of which is encoded by the gene adjacent to the ppep-1 gene. However, related PPEPs may exist in other bacteria and may shed light on substrate specificity in this enzyme family. Here, we report on the homolog of PPEP-1 in Paenibacillus alvei, which we denoted PPEP-2. We found that PPEP-2 is a secreted metalloprotease, which likewise cleaved a cell-surface protein encoded by an adjacent gene. However, the cleavage motif of PPEP-2, PLP↓PVP, is distinct from that of PPEP-1 (VNP↓PVP). As a result, an optimal substrate peptide for PPEP-2 was not cleaved by PPEP-1 and vice versa. To gain insight into the specificity mechanism of PPEP-2, we determined its crystal structure at 1.75 Å resolution and further confirmed the structure in solution using small-angle X-ray scattering (SAXS). We show that a four-amino-acid loop, which is distinct in PPEP-1 and -2 (GGST in PPEP-1 and SERV in PPEP-2), plays a crucial role in substrate specificity. A PPEP-2 variant, in which the four loop residues had been swapped for those from PPEP-1, displayed a shift in substrate specificity toward PPEP-1 substrates. Our results provide detailed insights into the PPEP-2 structure and the structural determinants of substrate specificity in this new family of PPEP proteases.
Collapse
Affiliation(s)
- Oleg I Klychnikov
- From the Laboratory for Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | | | - Stephen D Weeks
- From the Laboratory for Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | | | | | - Jan W Drijfhout
- Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 Leiden, The Netherlands
| | | | - Nikolai N Sluchanko
- the A. N. Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, 119071 Moscow, Russia, and.,the Department of Biophysics, Faculty of Biology, M. V. Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Sergei V Strelkov
- From the Laboratory for Biocrystallography, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | | |
Collapse
|
575
|
Hillemacher T, Bachmann O, Kahl KG, Frieling H. Alcohol, microbiome, and their effect on psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2018; 85:105-115. [PMID: 29705711 DOI: 10.1016/j.pnpbp.2018.04.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 04/05/2018] [Accepted: 04/25/2018] [Indexed: 12/12/2022]
Abstract
There is accumulating evidence that alcohol consumption and especially alcohol withdrawal increase brain levels of known innate immune signaling molecules and cause neuroinflammation. It has been shown that microbiota play a pivotal role in this process and affect central neurochemistry and behavior. Disruption of or alterations in the intimate cross-talk between microbiome and brain may be a significant factor in many psychiatric disorders. Alterations in the composition of the microbiome, so called dysbiosis, may result in detrimental distortion of microbe-host homeostasis modulating the hypothalamic-pituitary-adrenal axis. A variety of pathologies are associated with changes in the community structure and function of the gut microbiota, suggesting a link between dysbiosis and disease etiology, including irritable bowel syndrome depression, anxiety disorders, schizophrenia, and alcoholism. Despite a paucity of clinical studies in alcohol-dependent humans, emerging data suggests that alcohol induced alterations of the microbiome may explain reward-seeking behaviors as well as anxiety, depression, and craving in withdrawal and increase the risk of developing psychiatric disorders.
Collapse
Affiliation(s)
- Thomas Hillemacher
- Department of Psychiatry and Psychotherapy, Paracelsus Medical University Nuremberg, Germany; Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School (MHH), Germany
| | - Oliver Bachmann
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School (MHH), Germany; German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany
| | - Kai G Kahl
- Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School (MHH), Germany
| | - Helge Frieling
- Department of Psychiatry, Social Psychiatry, and Psychotherapy, Hannover Medical School (MHH), Germany.
| |
Collapse
|
576
|
Salem I, Ramser A, Isham N, Ghannoum MA. The Gut Microbiome as a Major Regulator of the Gut-Skin Axis. Front Microbiol 2018; 9:1459. [PMID: 30042740 PMCID: PMC6048199 DOI: 10.3389/fmicb.2018.01459] [Citation(s) in RCA: 349] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022] Open
Abstract
The adult intestine hosts a myriad of diverse bacterial species that reside mostly in the lower gut maintaining a symbiosis with the human habitat. In the current review, we describe the neoteric advancement in our comprehension of how the gut microbiota communicates with the skin as one of the main regulators in the gut-skin axis. We attempted to explore how this potential link affects skin differentiation and keratinization, its influence on modulating the cutaneous immune response in various diseases, and finally how to take advantage of this communication in the control of different skin conditions.
Collapse
Affiliation(s)
- Iman Salem
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
| | - Amy Ramser
- Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Nancy Isham
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
| | - Mahmoud A. Ghannoum
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
- Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
577
|
Gut microbial diversity in HIV infection post combined antiretroviral therapy: a key target for prevention of cardiovascular disease. Curr Opin HIV AIDS 2018; 13:38-44. [PMID: 29045253 PMCID: PMC5718258 DOI: 10.1097/coh.0000000000000426] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Although the HIV-infected population is living longer and getting older under current treatment regimens, significant challenges arise for health management as the infection is associated with various premature aging phenotypes, particularly increased incidence of cardiovascular diseases (CVDs). Here we review the current understanding of HIV-related gut dysbiosis in association with CVD and advances in clinical trials aiming to restore gut microbial diversity. RECENT FINDING Identification of a unique signature for gut dysbiosis in HIV infection between different cohorts remains challenging. However, low diversity of microbiota combined with the outgrowth of pathogenic bacterial species together with dysregulated metabolic pathways have been linked to compromised gut immunity, bacterial translocation and systemic inflammation, hence higher CVD risk among different cohorts. Data from recent clinical trials aiming to evaluate the tolerability and efficacy of probiotics in treated HIV+ patients are promising and support a significant increase in microbiota diversity and reduction of systemic inflammation. However, the impact of these microbial and immunological corrections on the prevalence of CVD in HIV+ patients remains unclear. SUMMARY Positive immunological outcomes following enrichment of the gut microbial diversity have been documented, and further trials are in progress to evaluate the range of patients, with different immunological backgrounds, who might benefit from these treatments.
Collapse
|
578
|
Barr T, Sureshchandra S, Ruegger P, Zhang J, Ma W, Borneman J, Grant K, Messaoudi I. Concurrent gut transcriptome and microbiota profiling following chronic ethanol consumption in nonhuman primates. Gut Microbes 2018; 9:338-356. [PMID: 29517944 PMCID: PMC6219653 DOI: 10.1080/19490976.2018.1441663] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/28/2017] [Accepted: 02/13/2018] [Indexed: 02/07/2023] Open
Abstract
Alcohol use disorder (AUD) results in increased intestinal permeability, nutrient malabsorption, and increased risk of colorectal cancer (CRC). Our understanding of the mechanisms underlying these morbidities remains limited because studies to date have relied almost exclusively on short-term heavy/binge drinking rodent models and colonic biopsies/fecal samples collected from AUD subjects with alcoholic liver disease (ALD). Consequently, the dose- and site-dependent impact of chronic alcohol consumption in the absence of overt liver disease remains poorly understood. In this study, we addressed this knowledge gap using a nonhuman primate model of voluntary ethanol self-administration where rhesus macaques consume varying amounts of 4% ethanol in water for 12 months. Specifically, we performed RNA-Seq and 16S rRNA gene sequencing on duodenum, jejunum, ileum, and colon biopsies collected from 4 controls and 8 ethanol-consuming male macaques. Our analysis revealed that chronic ethanol consumption leads to changes in the expression of genes involved in protein trafficking, metabolism, inflammation, and CRC development. Additionally, we observed differences in the relative abundance of putatively beneficial bacteria as well as those associated with inflammation and CRC. Given that the animals studied in this manuscript did not exhibit signs of ALD or CRC, our data suggest that alterations in gene expression and bacterial communities precede clinical disease and could serve as biomarkers as well as facilitate future studies aimed at developing interventions to restore gut homeostasis.
Collapse
Affiliation(s)
- Tasha Barr
- Division of Biomedical Sciences, University of California-Riverside, Riverside, CA, USA
| | - Suhas Sureshchandra
- Department of Molecular Biology and Biochemistry, University of California-Irvine, Irvine, CA, USA
| | - Paul Ruegger
- Department of Plant Pathology and Microbiology, University of California-Riverside, Riverside, CA, USA
| | - Jingfei Zhang
- Department of Statistics, University of California-Riverside, Riverside, CA, USA
| | - Wenxiu Ma
- Department of Statistics, University of California-Riverside, Riverside, CA, USA
| | - James Borneman
- Department of Plant Pathology and Microbiology, University of California-Riverside, Riverside, CA, USA
| | - Kathleen Grant
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
579
|
Hamilton AL, Kamm MA, Ng SC, Morrison M. Proteus spp. as Putative Gastrointestinal Pathogens. Clin Microbiol Rev 2018; 31:e00085-17. [PMID: 29899011 PMCID: PMC6056842 DOI: 10.1128/cmr.00085-17] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Proteus species, members of the Enterobacteriaceae family, are usually considered commensals in the gut and are most commonly recognized clinically as a cause of urinary tract infections. However, the recent identification of Proteus spp. as potential pathogens in Crohn's disease recurrence after intestinal resection serves as a stimulus to examine their potential role as gut pathogens. Proteus species possess many virulence factors potentially relevant to gastrointestinal pathogenicity, including motility; adherence; the production of urease, hemolysins, and IgA proteases; and the ability to acquire antibiotic resistance. Gastrointestinal conditions that have been linked to Proteus include gastroenteritis (spontaneous and foodborne), nosocomial infections, appendicitis, colonization of devices such as nasogastric tubes, and Crohn's disease. The association of Proteus species with Crohn's disease was particularly strong. Proteus species are low-abundance commensals of the human gut that harbor significant pathogenic potential; further investigation is needed.
Collapse
Affiliation(s)
- Amy L Hamilton
- Department of Gastroenterology, St Vincent's Hospital, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Australia
| | - Michael A Kamm
- Department of Gastroenterology, St Vincent's Hospital, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Australia
| | - Siew C Ng
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Mark Morrison
- The University of Queensland Diamantina Institute, Faculty of Medicine, Translational Research Institute, Brisbane, Australia
| |
Collapse
|
580
|
Samuel P, Ayoob KT, Magnuson BA, Wölwer-Rieck U, Jeppesen PB, Rogers PJ, Rowland I, Mathews R. Stevia Leaf to Stevia Sweetener: Exploring Its Science, Benefits, and Future Potential. J Nutr 2018; 148:1186S-1205S. [PMID: 29982648 DOI: 10.1093/jn/nxy102] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 04/19/2018] [Indexed: 12/13/2022] Open
Abstract
Steviol glycoside sweeteners are extracted and purified from the Stevia rebaudiana Bertoni plant, a member of the Asteraceae (Compositae) family that is native to South America, where it has been used for its sweet properties for hundreds of years. With continued increasing rates of obesity, diabetes, and other related comorbidities, in conjunction with global public policies calling for reductions in sugar intake as a means to help curb these issues, low- and no-calorie sweeteners (LNCSs, also known as high-potency sweeteners) such as stevia are gaining interest among consumers and food manufacturers. This appeal is related to stevia being plant-based, zero calorie and with a sweet taste that is 50-350 times sweeter than sugar, making it an excellent choice for use in sugar- and calorie-reduced food and beverage products. Despite the fact that the safety of stevia has been affirmed by several food regulatory and safety authorities around the world, insufficient education about stevia's safety and benefits, including continuing concern with regard to the safety of LNCSs in general, deters health professionals and consumers from recommending or using stevia. Therefore, the aim of this review and the stevia symposium that preceded this review at the ASN's annual conference in 2017 was to examine, in a comprehensive manner, the state of the science for stevia, its safety and potential health benefits, and future research and application. Topics covered included metabolism, safety and acceptable intake, dietary exposure, impact on blood glucose and insulin concentrations, energy intake and weight management, blood pressure, dental caries, naturality and processing, taste and sensory properties, regulatory status, consumer insights, and market trends. Data for stevia are limited in the case of energy intake and weight management as well as for the gut microbiome; therefore, the broader literature on LNCSs was reviewed at the symposium and therefore is also included in this review.
Collapse
Affiliation(s)
| | - Keith T Ayoob
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY
| | | | - Ursula Wölwer-Rieck
- Department of Bioanalytics/Food Chemistry, University of Bonn, Bonn, Germany
| | | | - Peter J Rogers
- School of Experimental Psychology, University of Bristol, Bristol, United Kingdom
| | - Ian Rowland
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | | |
Collapse
|
581
|
Abstract
PURPOSE OF REVIEW The objective of this review is to critically assess the contributing role of the gut microbiota in human obesity and type 2 diabetes (T2D). RECENT FINDINGS Experiments in animal and human studies have produced growing evidence for the causality of the gut microbiome in developing obesity and T2D. The introduction of high-throughput sequencing technologies has provided novel insight into the interpersonal differences in microbiome composition and function. The intestinal microbiota is known to be associated with metabolic syndrome and related comorbidities. Associated diseases including obesity, T2D, and fatty liver disease (NAFLD/NASH) all seem to be linked to altered microbial composition; however, causality has not been proven yet. Elucidating the potential causal and personalized role of the human gut microbiota in obesity and T2D is highly prioritized.
Collapse
Affiliation(s)
- Ömrüm Aydin
- Department of Internal Medicine, MC Slotervaart, Amsterdam, The Netherlands
- Department of Internal Medicine, AMC-UVA, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Internal Medicine, AMC-UVA, Amsterdam, The Netherlands
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
- Wallenberg Laboratory, University of Gothenberg, Gothenberg, Sweden
| | - Victor Gerdes
- Department of Internal Medicine, MC Slotervaart, Amsterdam, The Netherlands.
- Department of Internal Medicine, AMC-UVA, Amsterdam, The Netherlands.
| |
Collapse
|
582
|
Bermingham EN, Young W, Butowski CF, Moon CD, Maclean PH, Rosendale D, Cave NJ, Thomas DG. The Fecal Microbiota in the Domestic Cat ( Felis catus) Is Influenced by Interactions Between Age and Diet; A Five Year Longitudinal Study. Front Microbiol 2018; 9:1231. [PMID: 29971046 PMCID: PMC6018416 DOI: 10.3389/fmicb.2018.01231] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/22/2018] [Indexed: 01/14/2023] Open
Abstract
In humans, aging is associated with changes in the gastrointestinal microbiota; these changes may contribute to the age-related increase in incidence of many chronic diseases, including Type 2 diabetes. The life expectancies of cats are increasing, and they are also exhibiting the same types of diseases. While there are some studies investigating the impacts of diets on gastrointestinal microbiota in young cats, the impacts of aging in older cats has not been explored. We followed a cohort of related kittens, maintained on two commercial diets (kibbled and canned) from weaning (8 weeks) to 5 years of age (260 weeks). We hypothesized that the long-term feeding of specific diet formats would (a) lead to microbial composition changes due to aging, (b) impact body composition, and (c) affect insulin sensitivity in the aging cat. We observed that both diet and age affected fecal microbial composition, and while age correlated with changes in body composition, diet had no effect on body composition. Similarly insulin sensitivity was not affected by age nor diet. 16S rRNA sequencing found unclassified Peptostreptococcaceae were prominent across all ages averaging 21.3% of gene sequence reads and were higher in cats fed canned diets (average of 25.7% of gene sequence reads, vs. 17.0% for kibble-fed cats). Age-related effects on body composition and insulin sensitivity may become apparent as the cats grow older; this study will continue to assess these parameters.
Collapse
Affiliation(s)
- Emma N Bermingham
- Food Nutrition and Health Team, AgResearch, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Wayne Young
- Food Nutrition and Health Team, AgResearch, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand.,Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Christina F Butowski
- Food Nutrition and Health Team, AgResearch, Palmerston North, New Zealand.,School of Agriculture and Environment, Massey University, Palmerston North, New Zealand
| | - Christina D Moon
- Rumen Microbiology Team, AgResearch, Palmerston North, New Zealand
| | - Paul H Maclean
- Bioinformatics and Statistics Team, AgResearch, Lincoln, New Zealand
| | - Douglas Rosendale
- Food Nutrition and Health Group, The New Zealand Institute for Plant and Food Research Ltd, Food Industry Science Centre, Palmerston North, New Zealand
| | - Nicholas J Cave
- School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - David G Thomas
- School of Agriculture and Environment, Massey University, Palmerston North, New Zealand
| |
Collapse
|
583
|
Clinical intervention using Bifidobacterium strains in celiac disease children reveals novel microbial modulators of TNF-α and short-chain fatty acids. Clin Nutr 2018; 38:1373-1381. [PMID: 29960810 DOI: 10.1016/j.clnu.2018.06.931] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 05/30/2018] [Accepted: 06/06/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Celiac disease (CD) is an immune-mediated systemic disease, caused by ingestion of gluten in genetically predisposed individuals. Gut microbiota dysbiosis might play a significant role in pathogenesis of chronic enteropathies and its modulation can be used as an intervention strategy in CD as well. In this study, we aimed to identify correlations between fecal microbiota, serum tumor necrosis factor alpha (TNF-α) and fecal short-chain fatty acids (SCFAs) in healthy children and children with CD after administration of probiotic Bifidobacterium breve BR03 and B632. METHODS A double-blind placebo-controlled study enrolled 40 children with CD (CD) and 16 healthy children (HC). CD children were randomly allocated into two groups, of which 20 belonged to the placebo (PL) group and 20 to the Probiotic (PR) group. The PR group received a probiotic formulation containing a mixture of 2 strains, B. breve BR03 (DSM 16604) and B. breve B632 (DSM 24706) in 1:1 ratio for 3 months. Subsequently, for statistical analysis, blood and fecal samples from CD children (on enrolment - T0 and after 3 months, at the end of intervention with probiotic/placebo - T1) and HC children were used. The HC group was sampled only once (T0). RESULTS Verrucomicrobia, Parcubacteria and some yet unknown phyla of Bacteria and Archaea may be involved in the disease, indicated by a strong correlation to TNF-α. Likewise, Proteobacteria strongly correlated with fecal SCFAs concentration. The effect of probiotic administration has disclosed a negative correlation between Verrucomicrobia, some unknown phyla of Bacteria, Synergistetes, Euryarchaeota and some SCFAs, turning them into an important target in microbiome restoration process. Synergistetes and Euryarchaeota may have a role in the anti-inflammatory process in healthy human gut. CONCLUSIONS Our results highlight new phyla, which may have an important relation to disease-related parameters, CD itself and health.
Collapse
|
584
|
Bang SJ, Kim G, Lim MY, Song EJ, Jung DH, Kum JS, Nam YD, Park CS, Seo DH. The influence of in vitro pectin fermentation on the human fecal microbiome. AMB Express 2018; 8:98. [PMID: 29909506 PMCID: PMC6004267 DOI: 10.1186/s13568-018-0629-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 06/12/2018] [Indexed: 12/21/2022] Open
Abstract
Pectin is a complex dietary fiber and a prebiotic. To investigate pectin-induced changes in the gut microbiome and their effects on the short chain fatty acids (SCFAs) production, we performed in vitro pectin fermentation using the feces of three Korean donors. The pectin degradations in all three donors were observed. While the donors displayed differences in baseline gut microbiota composition, commonly increased bacteria after pectin fermentation included Lachnospira, Dorea, Clostridium, and Sutterella. Regarding SCFAs, acetate levels rapidly increased with incubation with pectin, and butyrate levels also increased after 6 h of incubation. The results suggest that pectin fermentation increases bacterial species belonging to Clostridium cluster XIV (Lachnospira, Dorea, and Clostridium), with Lachnospira displaying the greatest increase. The results also confirm that pectin fermentation leads to the production of acetate and butyrate.
Collapse
|
585
|
Bandara HMHN, Panduwawala CP, Samaranayake LP. Biodiversity of the human oral mycobiome in health and disease. Oral Dis 2018; 25:363-371. [PMID: 29786923 DOI: 10.1111/odi.12899] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 05/14/2018] [Accepted: 05/15/2018] [Indexed: 01/01/2023]
Abstract
The organisms that colonize the human body over a lifetime are diverse, extensive and gargantuan. A fair proportion of the microbiota that constitutes this human microbiome live within our oral cavities mostly as harmonious associates causing only sporadic disease. An important core constituent of the microbiome is the mycobiome, representing various fungal genera. Up until recently, only a few species of fungi, mainly Candida species, were thought to constitute the human oral mycobiome. The reasons for this are manifold, although the uncultivable nature of many fungi in conventional laboratory media, and their complex genetic composition seem to be the major factors which eluded their detection over the years. Nevertheless, recent advances in computing and high-throughput sequencing such as next-generation sequencing (NGS) platforms have provided us a panoramic view of a totally new world of fungi that are human oral cohabitués. Their diversity is perplexing, and functionality yet to be deciphered. Here, we provide a glimpse of what is currently known of the oral mycobiome, in health and disease, with some future perspectives.
Collapse
Affiliation(s)
| | - Chamila P Panduwawala
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, Sharjah, UAE
| | - Lakshman Perera Samaranayake
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, Sharjah, UAE
| |
Collapse
|
586
|
Abstract
PURPOSE OF REVIEW The review aims to discuss emerging evidence in the field of microbiome-dependent roles in host defense during critical illness with a focus on lung, kidney, and brain inflammation. RECENT FINDINGS The gut microbiota of critical ill patients is characterized by lower diversity, lower abundances of key commensal genera, and in some cases overgrowth by one bacterial genera, a state otherwise known as dysbiosis. Increasing evidence suggests that microbiota-derived components can reach the circulatory system from the gut and modulate immune homeostasis. Dysbiosis might have greater consequences for the critically ill than previously imagined and could contribute to poor outcome. Preclinical studies suggest that impaired communication across the gut - organ axes is associated with brain, lung - and kidney failure. SUMMARY In health, a diverse microbiome might enhance host defense, while during critical illness, the dysbiotic microbiome might contribute to comorbidity and organ dysfunction. Future research should be aimed at further establishing the causes and consequences of dysbiosis seen in the critically ill, which will provide perspective for developing new strategies of intervention.
Collapse
|
587
|
Sherman SB, Sarsour N, Salehi M, Schroering A, Mell B, Joe B, Hill JW. Prenatal androgen exposure causes hypertension and gut microbiota dysbiosis. Gut Microbes 2018; 9:400-421. [PMID: 29469650 PMCID: PMC6219642 DOI: 10.1080/19490976.2018.1441664] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/24/2017] [Accepted: 02/08/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Conditions of excess androgen in women, such as polycystic ovary syndrome (PCOS), often exhibit intergenerational transmission. One way in which the risk for PCOS may be increased in daughters of affected women is through exposure to elevated androgens in utero. Hyperandrogenemic conditions have serious health consequences, including increased risk for hypertension and cardiovascular disease. Recently, gut dysbiosis has been found to induce hypertension in rats, such that blood pressure can be normalized through fecal microbial transplant. Therefore, we hypothesized that the hypertension seen in PCOS has early origins in gut dysbiosis caused by in utero exposure to excess androgen. We investigated this hypothesis with a model of prenatal androgen (PNA) exposure and maternal hyperandrogenemia by single-injection of testosterone cypionate or sesame oil vehicle (VEH) to pregnant dams in late gestation. We then completed a gut microbiota and cardiometabolic profile of the adult female offspring. RESULTS The metabolic assessment revealed that adult PNA rats had increased body weight and increased mRNA expression of adipokines: adipocyte binding protein 2, adiponectin, and leptin in inguinal white adipose tissue. Radiotelemetry analysis revealed hypertension with decreased heart rate in PNA animals. The fecal microbiota profile of PNA animals contained higher relative abundance of bacteria associated with steroid hormone synthesis, Nocardiaceae and Clostridiaceae, and lower abundance of Akkermansia, Bacteroides, Lactobacillus, Clostridium. The PNA animals also had an increased relative abundance of bacteria associated with biosynthesis and elongation of unsaturated short chain fatty acids (SCFAs). CONCLUSIONS We found that prenatal exposure to excess androgen negatively impacted cardiovascular function by increasing systolic and diastolic blood pressure and decreasing heart rate. Prenatal androgen was also associated with gut microbial dysbiosis and altered abundance of bacteria involved in metabolite production of short chain fatty acids. These results suggest that early-life exposure to hyperandrogenemia in daughters of women with PCOS may lead to long-term alterations in gut microbiota and cardiometabolic function.
Collapse
Affiliation(s)
- Shermel B. Sherman
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Nadeen Sarsour
- Department of Biological Sciences, University of Toledo, Toledo, OH
| | - Marziyeh Salehi
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Allen Schroering
- Department of Neurosciences and Neurological Disorders, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Blair Mell
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
- Center for Hypertension and Personalized Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Bina Joe
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
- Center for Hypertension and Personalized Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Jennifer W. Hill
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
- Center for Diabetes and Endocrine Research, The University of Toledo College of Medicine and Life Sciences, Toledo, OH
| |
Collapse
|
588
|
Abstract
PURPOSE OF REVIEW The composition and diversity of the microbiota of the human gut, skin, and several other sites is severely deranged in critically ill patients on the ICU, and it is likely that these disruptions can negatively affect outcome. We here review new and ongoing studies that investigate the use of microbiota-targeted therapeutics in the ICU, and provide recommendations for future research. RECENT FINDINGS Practically every intervention in the ICU as well as the physiological effects of critical illness itself can have a profound impact on the gut microbiota. Therapeutic modulation of the microbiota, aimed at restoring the balance between 'pathogenic' and 'health-promoting' microbes is therefore of significant interest. Probiotics have shown to be effective in the treatment of ventilator-associated pneumonia, and the first fecal microbiota transplantations have recently been safely and successfully performed in the ICU. However, all-encompassing data in this vulnerable patient group remain sparse, and only a handful of novel studies that study microbiota-targeted therapies in the ICU are currently ongoing. SUMMARY Enormous strides have been made in characterizing the gut microbiome of critically ill patients in the ICU, and an increasing amount of preclinical data reveals the huge potential of microbiota-targeted therapies. Further understanding of the causes and consequences of dysbiosis on ICU-related outcomes are warranted to push the field forward.
Collapse
|
589
|
Iebba V, Guerrieri F, Di Gregorio V, Levrero M, Gagliardi A, Santangelo F, Sobolev AP, Circi S, Giannelli V, Mannina L, Schippa S, Merli M. Combining amplicon sequencing and metabolomics in cirrhotic patients highlights distinctive microbiota features involved in bacterial translocation, systemic inflammation and hepatic encephalopathy. Sci Rep 2018; 8:8210. [PMID: 29844325 PMCID: PMC5974022 DOI: 10.1038/s41598-018-26509-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 05/09/2018] [Indexed: 12/13/2022] Open
Abstract
In liver cirrhosis (LC), impaired intestinal functions lead to dysbiosis and possible bacterial translocation (BT). Bacteria or their byproducts within the bloodstream can thus play a role in systemic inflammation and hepatic encephalopathy (HE). We combined 16S sequencing, NMR metabolomics and network analysis to describe the interrelationships of members of the microbiota in LC biopsies, faeces, peripheral/portal blood and faecal metabolites with clinical parameters. LC faeces and biopsies showed marked dysbiosis with a heightened proportion of Enterobacteriaceae. Our approach showed impaired faecal bacterial metabolism of short-chain fatty acids (SCFAs) and carbon/methane sources in LC, along with an enhanced stress-related response. Sixteen species, mainly belonging to the Proteobacteria phylum, were shared between LC peripheral and portal blood and were functionally linked to iron metabolism. Faecal Enterobacteriaceae and trimethylamine were positively correlated with blood proinflammatory cytokines, while Ruminococcaceae and SCFAs played a protective role. Within the peripheral blood and faeces, certain species (Stenotrophomonas pavanii, Methylobacterium extorquens) and metabolites (methanol, threonine) were positively related to HE. Cirrhotic patients thus harbour a 'functional dysbiosis' in the faeces and peripheral/portal blood, with specific keystone species and metabolites related to clinical markers of systemic inflammation and HE.
Collapse
Affiliation(s)
- Valerio Iebba
- Istituto Pasteur Cenci Bolognetti Foundation, Public Health and Infectious Diseases Department, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Francesca Guerrieri
- Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Vincenza Di Gregorio
- Gastroenterology, Department of Clinical Medicine, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy
| | - Massimo Levrero
- Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
- INSERM, U1052, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL1), Centre Léon Bérard, Lyon, France
| | - Antonella Gagliardi
- Public Health and Infectious Diseases Department, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Floriana Santangelo
- Public Health and Infectious Diseases Department, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Anatoly P Sobolev
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185, Rome, Italy
- Magnetic Resonance Laboratory "Annalaura Segre", Institute of Chemical Methodologies, CNR, via Salaria km 29.300, 00015, Monterotondo, (RM), Italy
| | - Simone Circi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185, Rome, Italy
| | - Valerio Giannelli
- Gastroenterology, Department of Clinical Medicine, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy
| | - Luisa Mannina
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185, Rome, Italy
- Magnetic Resonance Laboratory "Annalaura Segre", Institute of Chemical Methodologies, CNR, via Salaria km 29.300, 00015, Monterotondo, (RM), Italy
| | - Serena Schippa
- Public Health and Infectious Diseases Department, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Manuela Merli
- Gastroenterology, Department of Clinical Medicine, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy.
| |
Collapse
|
590
|
Mohamadkhani A. On the potential role of intestinal microbial community in hepatocarcinogenesis in chronic hepatitis B. Cancer Med 2018; 7:3095-3100. [PMID: 29761927 PMCID: PMC6051233 DOI: 10.1002/cam4.1550] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/17/2018] [Accepted: 04/19/2018] [Indexed: 12/12/2022] Open
Abstract
The chronic infection of hepatitis B virus (HBV) is the most potent risk factor for the development of cirrhosis and hepatocellular carcinoma (HCC). The association of intestinal microbiota alteration with progressive liver disease has been investigated in recent studies. Overgrowth of potentially pathogenic bacteria of gram‐negative species and, in particular, a significant increase in the fecal count of Escherichia coli (E. coli) are characterized in the presence of HCC. This study was conducted to describe the characteristics of the intestinal microbiota related to the presence of HCC in HBV‐carrier patients. The available literature indicates the colonization of E. coli as principal source of portal vein lipopolysaccharide (LPS), in the gut may contribute to the carcinogenesis process by inducing chronic inflammation. This understanding could help to predict the clinical outcomes in HBV‐carrier patients and innovative strategies to reduce the virulence of liver disease from intestinal dysbiosis.
Collapse
Affiliation(s)
- Ashraf Mohamadkhani
- Liver and Pancreatobiliary Disease Research Center, Digestive Disease Research institute, Shariati Hospital, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
591
|
Zhang J, Shi H, Wang Y, Cao Z, Yang H, Li S. Effect of Limit-Fed Diets With Different Forage to Concentrate Ratios on Fecal Bacterial and Archaeal Community Composition in Holstein Heifers. Front Microbiol 2018; 9:976. [PMID: 29867879 PMCID: PMC5962747 DOI: 10.3389/fmicb.2018.00976] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/25/2018] [Indexed: 11/25/2022] Open
Abstract
Limit-feeding of a high concentrate diet has been proposed as an effective method for improving feed efficiency and reducing total manure output of dairy heifers; meanwhile the effects of this method on hindgut microbiota are still unclear. This study investigated the effects of a wide range of dietary forage:concentrate ratios (F:C) on the fecal composition of bacteria and archaea in heifers using next-generation sequencing. Four diets with different F:C (80:20, 60:40, 40:60, and 20:80) were limit-fed to 24 Holstein heifers, and the fecal fermentation parameters and bacterial and archaeal communities were investigated. With increasing dietary concentrate levels, the fecal dry matter output, neutral detergent fiber (NDF) content, and proportion of acetate decreased linearly (P < 0.01), while the fecal starch content and proportions of propionate, butyrate, and total branched-chain volatile fatty acids (TBCVFAs) were increased (P ≤ 0.05). An increased concentrate level linearly increased (P = 0.02) the relative abundance of Proteobacteria, and linearly decreased (P = 0.02) the relative abundance of Bacteroidetes in feces. At the genus level, the relative abundance of unclassified Ruminococcaceae and Paludibacter which may have the potential to degrade forage decreased linearly (q ≤ 0.02) with increasing dietary concentrate levels, while the relative abundance of Roseburia and Succinivibrio which may be non-fibrous carbohydrate degrading bacteria increased linearly (q ≤ 0.05). Some core microbiota operational taxonomic units (OTUs) also showed significant association with fecal VFAs, NDF, and/or acid detergent fiber (ADF) content. Meanwhile, the relative abundance of most detected taxa in archaea were similar across different F:C, and only Methanosphaera showed a linear decrease (P = 0.01) in high concentrate diets. Our study provides a better understanding of fecal fermentation parameters and microbiota under a wide range of dietary F:C. These findings support the potential for microbial manipulation by diet, which could enhance feed digestibility and relieve environmental problems associated with heifer rearing.
Collapse
Affiliation(s)
- Jun Zhang
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Haitao Shi
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China.,Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK, Canada
| | - Yajing Wang
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhijun Cao
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Hongjian Yang
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shengli Li
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
592
|
van Belkum A, Welker M, Pincus D, Charrier JP, Girard V. Matrix-Assisted Laser Desorption Ionization Time-of-Flight Mass Spectrometry in Clinical Microbiology: What Are the Current Issues? Ann Lab Med 2018; 37:475-483. [PMID: 28840984 PMCID: PMC5587819 DOI: 10.3343/alm.2017.37.6.475] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/20/2017] [Accepted: 07/25/2017] [Indexed: 12/12/2022] Open
Abstract
Matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) has revolutionized the identification of microbial species in clinical microbiology laboratories. MALDI-TOF-MS has swiftly become the new gold-standard method owing to its key advantages of simplicity and robustness. However, as with all new methods, adoption of the MALDI-TOF MS approach is still not widespread. Optimal sample preparation has not yet been achieved for several applications, and there are continuing discussions on the need for improved database quality and the inclusion of additional microbial species. New applications such as in the field of antimicrobial susceptibility testing have been proposed but not yet translated to the level of ease and reproducibility that one should expect in routine diagnostic systems. Finally, during routine identification testing, unexpected results are regularly obtained, and the best methods for transmitting these results into clinical care are still evolving. We here discuss the success of MALDI-TOF MS in clinical microbiology and highlight fields of application that are still amenable to improvement.
Collapse
Affiliation(s)
- Alex van Belkum
- Scientific Office, bioMérieux, La Balme Les Grottes, France.
| | - Martin Welker
- Scientific Office, bioMérieux, La Balme Les Grottes, France
| | - David Pincus
- Scientific Office, bioMérieux, La Balme Les Grottes, France
| | | | | |
Collapse
|
593
|
Microbiome Responses to an Uncontrolled Short-Term Diet Intervention in the Frame of the Citizen Science Project. Nutrients 2018; 10:nu10050576. [PMID: 29738477 PMCID: PMC5986456 DOI: 10.3390/nu10050576] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 04/28/2018] [Accepted: 05/04/2018] [Indexed: 12/14/2022] Open
Abstract
Personalized nutrition is of increasing interest to individuals actively monitoring their health. The relations between the duration of diet intervention and the effects on gut microbiota have yet to be elucidated. Here we examined the associations of short-term dietary changes, long-term dietary habits and lifestyle with gut microbiota. Stool samples from 248 citizen-science volunteers were collected before and after a self-reported 2-week personalized diet intervention, then analyzed using 16S rRNA sequencing. Considerable correlations between long-term dietary habits and gut community structure were detected. A higher intake of vegetables and fruits was associated with increased levels of butyrate-producing Clostridiales and higher community richness. A paired comparison of the metagenomes before and after the 2-week intervention showed that even a brief, uncontrolled intervention produced profound changes in community structure: resulting in decreased levels of Bacteroidaceae, Porphyromonadaceae and Rikenellaceae families and decreased alpha-diversity coupled with an increase of Methanobrevibacter, Bifidobacterium, Clostridium and butyrate-producing Lachnospiraceae- as well as the prevalence of a permatype (a bootstrapping-based variation of enterotype) associated with a higher diversity of diet. The response of microbiota to the intervention was dependent on the initial microbiota state. These findings pave the way for the development of an individualized diet.
Collapse
|
594
|
Garcia-Mantrana I, Selma-Royo M, Alcantara C, Collado MC. Shifts on Gut Microbiota Associated to Mediterranean Diet Adherence and Specific Dietary Intakes on General Adult Population. Front Microbiol 2018; 9:890. [PMID: 29867803 PMCID: PMC5949328 DOI: 10.3389/fmicb.2018.00890] [Citation(s) in RCA: 376] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 04/18/2018] [Indexed: 12/26/2022] Open
Abstract
There is increasing evidence for the interaction between gut microbiome, diet, and health. It is known that dysbiosis is related to disease and that most of the times this imbalances in gut microbial populations can be promoted through diet. Western dietary habits, which are characterized by high intakes of calories, animal proteins, saturated fats, and simple sugars have been linked with higher risk of obesity, diabetes, cancer, and cardiovascular disease. However, little is known about the impact of dietary patterns, dietary components, and nutrients on gut microbiota in healthy people. The aim of our study is to determine the effect of nutrient compounds as well as adherence to a dietary pattern, as the Mediterranean diet (MD) on the gut microbiome of healthy adults. Consequently, gut microbiota composition in healthy individuals, may be used as a potential biomarker to identify nutritional habits as well as risk of disease related to these habits. Dietary information from healthy volunteers (n = 27) was recorded using the Food Frequency Questionnaire. Adherence to the MD was measured using the PREDIMED test. Microbiota composition and diversity were obtained by 16S rRNA gene sequencing and specific quantitative polymerase chain reaction. Microbial metabolic activity was determined by quantification of short chain fatty acids (SCFA) on high performance liquid chromatography (HPLC). The results indicated that a higher ratio of Firmicutes–Bacteroidetes was related to lower adherence to the MD, and greater presence of Bacteroidetes was associated with lower animal protein intake. High consumption of animal protein, saturated fats, and sugars affected gut microbiota diversity. A significant higher presence of Christensenellaceae was found in normal-weight individuals compared to those who were overweight. This was also the case in volunteers with greater adherence to the MD compared to those with lower adherence. Butyricimonas, Desulfovibrio, and Oscillospira genera were associated with a BMI <25 and the genus Catenibacterium with a higher PREDIMED score. Higher bifidobacterial counts, and higher total SCFA were related to greater consumption of plant-based nutrients, such as vegetable proteins and polysaccharides. Better adherence to the MD was associated with significantly higher levels of total SCFA. Consequently, diet and specific dietary components could affect microbiota composition, diversity, and activity, which may have an effect on host metabolism by increasing the risk of Western diseases.
Collapse
Affiliation(s)
- Izaskun Garcia-Mantrana
- Institute of Agrochemistry and Food Technology, Spanish National Research Council, Valencia, Spain
| | - Marta Selma-Royo
- Institute of Agrochemistry and Food Technology, Spanish National Research Council, Valencia, Spain
| | - Cristina Alcantara
- Institute of Agrochemistry and Food Technology, Spanish National Research Council, Valencia, Spain
| | - María C Collado
- Institute of Agrochemistry and Food Technology, Spanish National Research Council, Valencia, Spain
| |
Collapse
|
595
|
Hylemon PB, Harris SC, Ridlon JM. Metabolism of hydrogen gases and bile acids in the gut microbiome. FEBS Lett 2018; 592:2070-2082. [PMID: 29683480 DOI: 10.1002/1873-3468.13064] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 12/26/2022]
Abstract
The human gut microbiome refers to a highly diverse microbial ecosystem, which has a symbiotic relationship with the host. Molecular hydrogen (H2 ) and carbon dioxide (CO2 ) are generated by fermentative metabolism in anaerobic ecosystems. H2 generation and oxidation coupled to CO2 reduction to methane or acetate help maintain the structure of the gut microbiome. Bile acids are synthesized by hepatocytes from cholesterol in the liver and are important regulators of host metabolism. In this Review, we discuss how gut bacteria metabolize hydrogen gases and bile acids in the intestinal tract and the consequences on host physiology. Finally, we focus on bile acid metabolism by the Actinobacterium Eggerthella lenta. Eggerthella lenta appears to couple hydroxyl group oxidations to reductive acetogenesis under a CO2 or N2 atmosphere, but not under H2 . Hence, at low H2 levels, E. lenta is proposed to use NADH from bile acid hydroxyl group oxidations to reduce CO2 to acetate.
Collapse
Affiliation(s)
- Phillip B Hylemon
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA.,McGuire Veterans Hospital, Richmond, VA, USA
| | - Spencer C Harris
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA.,McGuire Veterans Hospital, Richmond, VA, USA
| | - Jason M Ridlon
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, IL, USA
| |
Collapse
|
596
|
Kaur CP, Vadivelu J, Chandramathi S. Impact of Klebsiella pneumoniae in lower gastrointestinal tract diseases. J Dig Dis 2018; 19:262-271. [PMID: 29573336 DOI: 10.1111/1751-2980.12595] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/26/2018] [Accepted: 03/19/2018] [Indexed: 12/11/2022]
Abstract
The 2016 Global Burden of Disease report by WHO revealed that diseases of the gastrointestinal tract (GIT) had one of the highest incidence rates worldwide. The plethora of factors that contribute to the development of GIT-related illnesses can be divided into genetic, environmental and lifestyle factors. Apart from that, the role that infectious agents play in the development of GIT diseases has piqued the interest of researchers worldwide. The human gut harbors approximately 1014 bacteria in it with increasing concentration toward the lower GIT. Among the various microbiota that colonize the human gut, Gram-negative bacteria have been most notoriously linked to GIT-related diseases such as inflammatory bowel disease (IBD) including Crohn's disease and ulcerative colitis and colorectal cancer (CRC). Some of the notable culprits that have been attributed to these diseases are Bacteroides fragilis, Fusobacterium nucleatum, Escherichia coli and Helicobacter pylori. However, studies in recent years are beginning to recognize a new player, Klebsiella pneumoniae (K. pneumoniae) in the causation and progression of GIT diseases. Once synonymous with infections and diseases of the upper respiratory tract, K. pneumoniae has now emerged as one of the pathogens commonly isolated from patients with GIT diseases. However, extensive studies attributing K. pneumoniae to GIT diseases, particularly that of CRC are scanty. Therefore, this review intends to shed light on the association of K. pneumoniae in gastrointestinal diseases such as Crohn's disease, ulcerative colitis as well as CRC.
Collapse
Affiliation(s)
- Christina Parvinder Kaur
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Jamuna Vadivelu
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Samudi Chandramathi
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
597
|
Abstract
Allogeneic haematopoietic stem cell transplantation (allo-HSCT) is considered to be the strongest curative immunotherapy for various malignancies (primarily, but not limited to, haematologic malignancies). However, application of allo-HSCT is limited owing to its life-threatening major complications, such as graft-versus-host disease (GVHD), relapse and infections. Recent advances in large-scale DNA sequencing technology have facilitated rapid identification of the microorganisms that make up the microbiota and evaluation of their interactions with host immunity in various diseases, including cancer. This has resulted in renewed interest regarding the role of the intestinal flora in patients with haematopoietic malignancies who have received an allo-HSCT and in whether the microbiota affects clinical outcomes, including GVHD, relapse, infections and transplant-related mortality. In this Review, we discuss the potential role of intestinal microbiota in these major complications after allo-HSCT, summarize clinical trials evaluating the microbiota in patients who have received allo-HSCT and discuss how further studies of the microbiota could inform the development of strategies that improve outcomes of allo-HSCT.
Collapse
Affiliation(s)
- Yusuke Shono
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, New York, USA
| | - Marcel R. M. van den Brink
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Medical College of Cornell University, New York, New York, USA
- Adult BMT Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
598
|
Joch M, Mrázek J, Skřivanová E, Čermák L, Marounek M. Effects of pure plant secondary metabolites on methane production, rumen fermentation and rumen bacteria populations in vitro. J Anim Physiol Anim Nutr (Berl) 2018; 102:869-881. [PMID: 29707819 DOI: 10.1111/jpn.12910] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 03/23/2018] [Indexed: 12/25/2022]
Abstract
In this study, the effects of seven pure plant secondary metabolites (PSMs) on rumen fermentation, methane (CH4 ) production and rumen bacterial community composition were determined. Two in vitro trials were conducted. In trial 1, nine concentrations of 8-hydroxyquinoline, α-terpineol, camphor, bornyl acetate, α-pinene, thymoquinone and thymol were incubated on separate days using in vitro 24-hr batch incubations. All compounds tested demonstrated the ability to alter rumen fermentation parameters and decrease CH4 production. However, effective concentrations differed among individual PSMs. The lowest concentrations that reduced (p < .05) CH4 production were as follows: 8 mg/L of 8-hydroxyquinoline, 120 mg/L of thymoquinone, 240 mg/L of thymol and 480 mg/L of α-terpineol, camphor, bornyl acetate and α-pinene. These concentrations were selected for use in trial 2. In trial 2, PSMs were incubated in one run. Methane was decreased (p < .05) by all PSMs at selected concentrations. However, only 8-hydroxyquinoline, bornyl acetate and thymoquinone decreased (p < .05) CH4 relative to volatile fatty acids (VFAs). Based on denaturing gradient gel electrophoresis analysis, different PSMs changed the composition of bacterial communities to different extents. As revealed by Ion Torrent sequencing, the effects of PSMs on relative abundance were most pronounced in the predominant families, especially in Lachnospiraceae, Succinivibrionaceae, Prevotellaceae, unclassified Clostridiales and Ruminococcaceae. The CH4 production was correlated negatively (-.72; p < .05) with relative abundance of Succinivibrionaceae and positively with relative abundance of Ruminococcaceae (.86; p < .05). In summary, this study identified three pure PSMs (8hydroxyquinoline, bornyl acetate and thymoquinone) with potentially promising effects on rumen CH4 production. The PSMs tested in this study demonstrated considerable impact on rumen bacterial communities even at the lowest concentrations that decreased CH4 production. The findings from this study may help to elucidate how PSMs affect rumen bacterial fermentation.
Collapse
Affiliation(s)
- M Joch
- Nutrition and Feeding of Farm Animals, Institute of Animal Science, Prague, Czech Republic.,Department of Microbiology, Nutrition and Dietetics, Czech University of Life Sciences, Prague, Czech Republic
| | - J Mrázek
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Prague, Czech Republic
| | - E Skřivanová
- Nutrition and Feeding of Farm Animals, Institute of Animal Science, Prague, Czech Republic.,Department of Microbiology, Nutrition and Dietetics, Czech University of Life Sciences, Prague, Czech Republic
| | - L Čermák
- Nutrition and Feeding of Farm Animals, Institute of Animal Science, Prague, Czech Republic
| | - M Marounek
- Nutrition and Feeding of Farm Animals, Institute of Animal Science, Prague, Czech Republic.,Department of Microbiology, Nutrition and Dietetics, Czech University of Life Sciences, Prague, Czech Republic
| |
Collapse
|
599
|
Kumar J, Verma MK, Kumar T, Gupta S, Pandey R, Yadav M, Chauhan NS. S9A Serine Protease Engender Antigenic Gluten Catabolic Competence to the Human Gut Microbe. Indian J Microbiol 2018; 58:294-300. [PMID: 30013273 DOI: 10.1007/s12088-018-0732-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 04/22/2018] [Indexed: 12/13/2022] Open
Abstract
The human gut microbiome has a significant role in host physiology; however its role in gluten catabolism is debatable. Present study explores the role of human gut microbes in gluten catabolism and a native human gut microbe Cellulomonas sp. HM71 was identified. SSU rDNA analysis has described human gut microbiome structure and also confirmed the permanent residentship of Cellulomonas sp. HM71. Catabolic potential of Cellulomonas sp. HM71 to cleave antigenic gluten peptides indicates presence of candidate gene encoding biocatalytic machinery. Genome analysis has identified the presence of gene encoding S9A serine protease family-prolyl endopeptidase, with Ser591, Asp664 and His685 signature residues. Cellulomonas sp. HM71 prolyl endopeptidase activity was found optimal at pH 7.0 and 37 °C with a KM of 35.53 μmol and specifically cleaves at proline residue. Current study describes the gluten catabolism potential of Cellulomonas sp. HM71 depicting possible role of human gut microbes in gluten catabolism to confer resistance mechanisms for the onset of celiac diseases in populations with gluten diet.
Collapse
Affiliation(s)
- Jitendra Kumar
- 1Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana 124001 India
| | - Manoj Kumar Verma
- 1Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana 124001 India
| | - Tarun Kumar
- 1Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana 124001 India
| | - Shashank Gupta
- 1Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana 124001 India
| | - Rajesh Pandey
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD UK
| | - Monika Yadav
- 1Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana 124001 India
| | - Nar Singh Chauhan
- 1Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana 124001 India
| |
Collapse
|
600
|
Lucke A, Böhm J, Zebeli Q, Metzler-Zebeli BU. Dietary Deoxynivalenol Contamination and Oral Lipopolysaccharide Challenge Alters the Cecal Microbiota of Broiler Chickens. Front Microbiol 2018; 9:804. [PMID: 29922239 PMCID: PMC5996912 DOI: 10.3389/fmicb.2018.00804] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/10/2018] [Indexed: 12/18/2022] Open
Abstract
Dietary deoxynivalenol (DON) impairs the intestinal functions and performance in broiler chickens, whereas little is known about the effect of DON on the gastrointestinal microbiota. This study evaluated the impact of graded levels of dietary DON contamination on the cecal bacterial microbiota, their predicted metabolic abilities and short-chain fatty acid (SCFA) profiles in chickens. In using a single oral lipopolysaccharide (LPS) challenge we further assessed whether an additional intestinal stressor would potentiate DON-related effects on the cecal microbiota. Eighty 1-day-old chicks were fed diets with increasing DON concentrations (0, 2.5, 5, and 10 mg DON per kg diet) for 5 weeks and were sampled after half of the chickens received an oral LPS challenge (1 mg LPS/kg bodyweight) 1 day before sampling. The bacterial composition was investigated by Illumina MiSeq sequencing of the V3–5 region of the 16S rRNA gene. DON-feeding decreased (p < 0.05) the cecal species richness (Chao1) and evenness (Shannon) compared to the non-contaminated diet. The phyla Firmicutes and Proteobacteria tended to linearly increase and decrease with increasing DON-concentrations, respectively. Within the Firmicutes, DON decreased the relative abundance of Oscillospira, Clostridiaceae genus, Clostridium, and Ruminococcaceae genus 2 (p < 0.05), whereas it increased Clostridiales genus 2 (p < 0.05). Moreover, increasing DON levels linearly decreased a high-abundance Enterobacteriaceae genus and an Escherichia/Shigella-OTU (p < 0.05). Changes in the bacterial composition and their imputed metagenomic capabilities may be explained by DON-related changes in host physiology and cecal nutrient availability. The oral LPS challenge only decreased the abundance of an unassigned Clostridiales genus 2 (p = 0.03). Increasing dietary concentrations of DON quadratically increased the cecal total SCFA and butyrate concentration (p < 0.05), whereas a DON × LPS interaction indicated that LPS mainly increased cecal total SCFA, butyrate, and acetate concentrations in chickens fed the diets that were not contaminated with DON. The present findings showed that even the lowest level of dietary DON contamination had modulatory effects on chicken's cecal bacterial microbiota composition and diversity, whereas the additional oral challenge with LPS did not potentiate DON effects on the cecal bacterial composition.
Collapse
Affiliation(s)
- Annegret Lucke
- Department for Farm Animals and Veterinary Public Health, Institute of Animal Nutrition and Functional Plant Compounds, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Josef Böhm
- Department for Farm Animals and Veterinary Public Health, Institute of Animal Nutrition and Functional Plant Compounds, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Qendrim Zebeli
- Department for Farm Animals and Veterinary Public Health, Institute of Animal Nutrition and Functional Plant Compounds, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Barbara U Metzler-Zebeli
- Department for Farm Animals and Veterinary Public Health, Institute of Animal Nutrition and Functional Plant Compounds, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|