201
|
Syeda T, Sanchez-Tapia M, Pinedo-Vargas L, Granados O, Cuervo-Zanatta D, Rojas-Santiago E, Díaz-Cintra SA, Torres N, Perez-Cruz C. Bioactive Food Abates Metabolic and Synaptic Alterations by Modulation of Gut Microbiota in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2019; 66:1657-1682. [PMID: 30475761 DOI: 10.3233/jad-180556] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent investigations have demonstrated an important role of gut microbiota (GM) in the pathogenesis of Alzheimer's disease (AD). GM modulates a host's health and disease by production of several substances, including lipopolysaccharides (LPS) and short-chain fatty acids (SCFAs), among others. Diet can modify the composition and diversity of GM, and ingestion of a healthy diet has been suggested to lower the risk to develop AD. We have previously shown that bioactive food (BF) ingestion can abate neuroinflammation and oxidative stress and improve cognition in obese rats, effects associated with GM composition. Therefore, BF can impact the gut-brain axis and improved behavior. In this study, we aim to explore if inclusion of BF in the diet may impact central pathological markers of AD by modulation of the GM. Triple transgenic 3xTg-AD (TG) female mice were fed a combination of dried nopal, soy, chia oil, and turmeric for 7 months. We found that BF ingestion improved cognition and reduced Aβ aggregates and tau hyperphosphorylation. In addition, BF decreased MDA levels, astrocyte and microglial activation, PSD-95, synaptophysin, GluR1 and ARC protein levels in TG mice. Furthermore, TG mice fed BF showed increased levels of pGSK-3β. GM analysis revealed that pro-inflammatory bacteria were more abundant in TG mice compared to wild-type, while BF ingestion was able to restore the GM's composition, LPS, and propionate levels to control values. Therefore, the neuroprotective effects of BF may be mediated, in part, by modulation of GM and the release of neurotoxic substances that alter brain function.
Collapse
Affiliation(s)
- Tauqeerunnisa Syeda
- Departmento de Farmacología, Centro de Investigación y de Estudios Avanzados del I.P.N. 2508, Mexico City, Mexico
| | - Mónica Sanchez-Tapia
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Laura Pinedo-Vargas
- Instituto Nacional de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla-Querétaro, Mexico
| | - Omar Granados
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Daniel Cuervo-Zanatta
- Departmento de Farmacología, Centro de Investigación y de Estudios Avanzados del I.P.N. 2508, Mexico City, Mexico
| | | | - Sof A Díaz-Cintra
- Instituto Nacional de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla-Querétaro, Mexico
| | - Nimbe Torres
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Claudia Perez-Cruz
- Departmento de Farmacología, Centro de Investigación y de Estudios Avanzados del I.P.N. 2508, Mexico City, Mexico
| |
Collapse
|
202
|
Ueyama J, Oda M, Hirayama M, Sugitate K, Sakui N, Hamada R, Ito M, Saito I, Ohno K. Freeze-drying enables homogeneous and stable sample preparation for determination of fecal short-chain fatty acids. Anal Biochem 2019; 589:113508. [PMID: 31751532 DOI: 10.1016/j.ab.2019.113508] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/15/2019] [Accepted: 11/15/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND The analysis methods for fecal short-chain fatty acids (SCFAs) have evolved considerably. Recently, the role of SCFAs in gastrointestinal physiology and their association with intestinal microbiota and disease were reported. However, the intra-fecal variability and storage stability of SCFAs have not been extensively investigated. The aim of this study was to understand the limitations of the measurement of SCFAs in crude feces and develop a useful pre-examination procedure using the freeze-drying technique. METHODS SCFAs in crude feces, obtained from healthy volunteers, and freeze-dried feces were determined by derivatization with isobutyl chloroformate, followed by liquid-liquid extraction with hexane, and separation and analysis using gas chromatography-mass spectrometry. RESULTS Among the SCFAS, the maximum intra-fecal variability was observed for iso-butyrate (coefficient of variation of 37.7%), but the freeze-drying procedure reduced this variability (coefficient of variation of 7.9%). Similar improvements were also observed for other SCFAs. Furthermore, significant decreases in the SCFA amounts were observed with storage at 4 °C for 24 h. CONCLUSIONS The freeze-drying procedure affords fecal SCFA stability, even with storage at room temperature for 3 d. The freeze-drying procedure allows reliable SCFA measurements without labour-intensive processes. Therefore, the freeze-drying procedure can be applied in basic, clinical, and epidemiological studies.
Collapse
Affiliation(s)
- Jun Ueyama
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daikominami, Higashi-ku, Nagoya, 461-8673, Japan.
| | - Masaya Oda
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daikominami, Higashi-ku, Nagoya, 461-8673, Japan
| | - Masaaki Hirayama
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daikominami, Higashi-ku, Nagoya, 461-8673, Japan
| | - Kuniyo Sugitate
- Agilent Technologies Japan, Ltd, 9-1 Takakura-cho, Hachioji, Tokyo, 192-8510, Japan
| | - Norihiro Sakui
- Agilent Technologies Japan, Ltd, 9-1 Takakura-cho, Hachioji, Tokyo, 192-8510, Japan
| | - Risa Hamada
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daikominami, Higashi-ku, Nagoya, 461-8673, Japan
| | - Mikako Ito
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Isao Saito
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, 1-1-20 Daikominami, Higashi-ku, Nagoya, 461-8673, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
203
|
Combination of cannabinoids, delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD), mitigates experimental autoimmune encephalomyelitis (EAE) by altering the gut microbiome. Brain Behav Immun 2019; 82:25-35. [PMID: 31356922 PMCID: PMC6866665 DOI: 10.1016/j.bbi.2019.07.028] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/17/2019] [Accepted: 07/25/2019] [Indexed: 02/07/2023] Open
Abstract
Currently, a combination of marijuana cannabinoids including delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD) is used as a drug to treat muscle spasticity in patients with Multiple Sclerosis (MS). Because these cannabinoids can also suppress inflammation, it is unclear whether such patients benefit from suppression of neuroinflammation and if so, what is the mechanism through which cannabinoids act. In the currently study, we used a murine model of MS, experimental autoimmune encephalomyelitis (EAE), to study the role of gut microbiota in the attenuation of clinical signs of paralysis and inflammation caused by cannabinoids. THC + CBD treatment attenuated EAE and caused significant decrease in inflammatory cytokines such as IL-17 and IFN-γ while promoting the induction of anti-inflammatory cytokines such as IL-10 and TGF-β. Use of 16S rRNA sequencing on bacterial DNA extracted from the gut revealed that EAE mice showed high abundance of mucin degrading bacterial species, such as Akkermansia muciniphila (A. muc), which was significantly reduced after THC + CBD treatment. Fecal Material Transfer (FMT) experiments confirmed that THC + CBD-mediated changes in the microbiome play a critical role in attenuating EAE. In silico computational metabolomics revealed that LPS biosynthesis, a key component in gram-negative bacteria such as A. muc, was found to be elevated in EAE mice which was confirmed by demonstrating higher levels of LPS in the brain, while treatment with THC + CBD reversed this trend. EAE mice treated with THC + CBD also had significantly higher levels of short chain fatty acids such as butyric, isovaleric, and valeric acids compared to naïve or disease controls. Collectively, our data suggest that cannabinoids may attenuate EAE and suppress neuroinflammation by preventing microbial dysbiosis seen during EAE and promoting healthy gut microbiota.
Collapse
|
204
|
Zhang C, Fan L, Zhao H. Rapid Detection of Short-Chain Fatty Acids in Biological Samples. Chromatographia 2019. [DOI: 10.1007/s10337-019-03824-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
205
|
Mohammed AD, Khan MAW, Chatzistamou I, Chamseddine D, Williams-Kang K, Perry M, Enos R, Murphy A, Gomez G, Aladhami A, Oskeritzian CA, Jolly A, Chang Y, He S, Pan Z, Kubinak JL. Gut Antibody Deficiency in a Mouse Model of CVID Results in Spontaneous Development of a Gluten-Sensitive Enteropathy. Front Immunol 2019; 10:2484. [PMID: 31708923 PMCID: PMC6820504 DOI: 10.3389/fimmu.2019.02484] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 10/04/2019] [Indexed: 12/14/2022] Open
Abstract
Primary immunodeficiencies are heritable disorders of immune function. CD19 is a B cell co-receptor important for B cell development, and CD19 deficiency is a known genetic risk factor for a rare form of primary immunodeficiency known as “common variable immunodeficiency” (CVID); an antibody deficiency resulting in low levels of serum IgG and IgA. Enteropathies are commonly observed in CVID patients but the underlying reason for this is undefined. Here, we utilize CD19−/− mice as a model of CVID to test the hypothesis that antibody deficiency negatively impacts gut physiology under steady-state conditions. As anticipated, immune phenotyping experiments demonstrate that CD19−/− mice develop a severe B cell deficiency in gut-associated lymphoid tissues that result in significant reductions to antibody concentrations in the gut lumen. Antibody deficiency was associated with defective anti-commensal IgA responses and the outgrowth of anaerobic bacteria in the gut. Expansion of anaerobic bacteria coincides with the development of a chronic inflammatory condition in the gut of CD19−/− mice that results in an intestinal malabsorption characterized by defects in lipid metabolism and transport. Administration of the antibiotic metronidazole to target anaerobic members of the microbiota rescues mice from disease indicating that intestinal malabsorption is a microbiota-dependent phenomenon. Finally, intestinal malabsorption in CD19−/− mice is a gluten-sensitive enteropathy as exposure to a gluten-free diet also significantly reduces disease severity in CD19−/− mice. Collectively, these results support an effect of antibody deficiency on steady-state gut physiology that compliment emerging data from human studies linking IgA deficiency with non-infectious complications associated with CVID. They also demonstrate that CD19−/− mice are a useful model for studying the role of B cell deficiency and gut dysbiosis on gluten-sensitive enteropathies; a rapidly emerging group of diseases in humans with an unknown etiology.
Collapse
Affiliation(s)
- Ahmed Dawood Mohammed
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States.,School of Veterinary Medicine, University of Baghdad, Baghdad, Iraq
| | - Md A Wadud Khan
- Biology Department, University of Texas at Arlington, Arlington, TX, United States
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Douja Chamseddine
- Biology Department, University of Texas at Arlington, Arlington, TX, United States
| | - Katie Williams-Kang
- Biology Department, University of Texas at Arlington, Arlington, TX, United States
| | - Mason Perry
- Biology Department, University of Texas at Arlington, Arlington, TX, United States
| | - Reilly Enos
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Angela Murphy
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Gregorio Gomez
- Department of Biomedical Sciences, College of Medicine, University of Houston, Houston, TX, United States
| | - Ahmed Aladhami
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Carole A Oskeritzian
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Amy Jolly
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Yan Chang
- College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX, United States
| | - Shuqian He
- College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX, United States
| | - Zui Pan
- College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX, United States
| | - Jason L Kubinak
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| |
Collapse
|
206
|
Cires MJ, Navarrete P, Pastene E, Carrasco-Pozo C, Valenzuela R, Medina DA, Andriamihaja M, Beaumont M, Blachier F, Gotteland M. Protective Effect of an Avocado Peel Polyphenolic Extract Rich in Proanthocyanidins on the Alterations of Colonic Homeostasis Induced by a High-Protein Diet. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:11616-11626. [PMID: 31542929 DOI: 10.1021/acs.jafc.9b03905] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Avocado peel, a byproduct from the avocado pulp industry, is a promising source of polyphenolic compounds. We evaluated the effect of a proanthocyanidin-rich avocado peel polyphenol extract (AvPPE) on the composition and metabolic activity of human fecal microbiota cultured for 24 h in a bioreactor in the presence of high protein (HP) amounts and the effect of the resulting culture supernatants (CSs) on HT-29Glc-/+ and Caco-2 cells. AvPPE decreased the HP-induced production of ammonia, H2S, propionate, and isovalerate and increased that of indole and butyrate. Microbiota composition was marginally affected by HP, whileAvPPE increased the microorganisms/abundance of phylum Actinobacteria, families Coriobacteriaceae and Ruminococcaceae, and genus Faecalibacterium. AvPPE failed to prevent the HP-induced decrease of HT-29Glc-/+ cell viability and energy efficiency but prevented the HP-induced alterations of barrier function in Caco-2 cells. Additionally, the genotoxic effect of the CSs upon HT-29Glc-/+ was attenuated by AvPPE. Therefore, AvPPE may be considered as a promising product for improving colonic homeostasis.
Collapse
Affiliation(s)
- María José Cires
- Department of Nutrition, Faculty of Medicine , University of Chile , Santiago 8380453 , Chile
| | - Paola Navarrete
- Laboratory of Microbiology and Probiotics, Institute of Nutrition and Food Technology (INTA) , University of Chile , Santiago 7830490 , Chile
- Millennium Nucleus in the Biology of Intestinal Microbiota , Santiago , Chile
| | - Edgar Pastene
- Laboratory of Pharmacognosy, Faculty of Pharmacy , University of Concepción , Concepción 4030000 , Chile
- Laboratorio de Sı́ntesis y Biotransformación de Productos Naturales, Departamento de Ciencias Básicas, Facultad de Ciencias , Universidad del Bı́o-Bı́o , Chillán 3780000 , Chile
| | - Catalina Carrasco-Pozo
- Department of Nutrition, Faculty of Medicine , University of Chile , Santiago 8380453 , Chile
- Discovery Biology, Griffith Institute for Drug Discovery , Griffith University , Nathan , Queensland 4111 , Australia
| | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine , University of Chile , Santiago 8380453 , Chile
| | - Daniel A Medina
- Laboratorio de Biotecnología Aplicada, Facultad de Medicina Veterinaria , Universidad San Sebastián , Lago Panguipulli 1390 , Puerto Montt 5480000 , Chile
| | | | - Martin Beaumont
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay , Paris 75005 , France
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT , Toulouse 31326 , France
| | - François Blachier
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay , Paris 75005 , France
| | - Martin Gotteland
- Department of Nutrition, Faculty of Medicine , University of Chile , Santiago 8380453 , Chile
- Laboratory of Microbiology and Probiotics, Institute of Nutrition and Food Technology (INTA) , University of Chile , Santiago 7830490 , Chile
- Millennium Nucleus in the Biology of Intestinal Microbiota , Santiago , Chile
| |
Collapse
|
207
|
Tornero-Martínez A, Cruz-Ortiz R, Jaramillo-Flores ME, Osorio-Díaz P, Ávila-Reyes SV, Alvarado-Jasso GM, Mora-Escobedo R. In vitro Fermentation of Polysaccharides from Aloe vera and the Evaluation of Antioxidant Activity and Production of Short Chain Fatty Acids. Molecules 2019; 24:E3605. [PMID: 31591306 PMCID: PMC6803901 DOI: 10.3390/molecules24193605] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023] Open
Abstract
Soluble or fermentable fibre has prebiotic effects that can be used in the food industry to modify the composition of microbiota species to benefit human health. Prebiotics mostly target Bifidobacterium and Lactobacillus strains, among others, which can fight against chronic diseases since colonic fermentation produces short chain fatty acids (SCFAs). The present work studied the changes produced in the fibre and polyphenolic compounds during in vitro digestion of gel (AV) and a polysaccharide extract (AP) from Aloe vera, after which, these fractions were subjected to in vitro colonic fermentation to evaluate the changes in antioxidant capacity and SCFAs production during the fermentation. The results showed that the phenolic compounds increased during digestion, but were reduced in fermentation, as a consequence, the antioxidant activity increased significantly in AV and AP after the digestion. On the other hand, during in vitro colon fermentation, the unfermented fibre of AV and AP responded as lactulose and the total volume of gas produced, which indicates the possible use of Aloe vera and polysaccharide extract as prebiotics.
Collapse
Affiliation(s)
- Antonio Tornero-Martínez
- Instituto Politécnico Nacional, ENCB, Campus Zacatenco. Miguel Othón de Mendizábal 699, Alcaldía G.A. Madero, Ciudad de México C.P. 07360, CDMX, Mexico.
| | - Rubén Cruz-Ortiz
- Instituto Politécnico Nacional, ENCB, Campus Zacatenco. Miguel Othón de Mendizábal 699, Alcaldía G.A. Madero, Ciudad de México C.P. 07360, CDMX, Mexico.
| | - María Eugenia Jaramillo-Flores
- Instituto Politécnico Nacional, ENCB, Campus Zacatenco. Miguel Othón de Mendizábal 699, Alcaldía G.A. Madero, Ciudad de México C.P. 07360, CDMX, Mexico.
| | - Perla Osorio-Díaz
- Instituto Politécnico Nacional, CEPROBI, Carretera Yautepec-Jojutla, Km. 6, Yautepec C.P. 62731, Morelos, Mexico.
| | - Sandra Victoria Ávila-Reyes
- Instituto Politécnico Nacional, CEPROBI, Carretera Yautepec-Jojutla, Km. 6, Yautepec C.P. 62731, Morelos, Mexico.
- CONACYT-Instituto Politécnico Nacional, CEPROBI, Carretera Yautepec-Jojutla, Km. 6, Yautepec C.P. 62731, Morelos, Mexico.
| | | | - Rosalva Mora-Escobedo
- Instituto Politécnico Nacional, ENCB, Campus Zacatenco. Miguel Othón de Mendizábal 699, Alcaldía G.A. Madero, Ciudad de México C.P. 07360, CDMX, Mexico.
| |
Collapse
|
208
|
Optimized germinated soybean/cornstarch extrudate and its in vitro fermentation with human inoculum. Journal of Food Science and Technology 2019; 57:848-857. [PMID: 32123405 DOI: 10.1007/s13197-019-04116-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 06/03/2019] [Accepted: 09/26/2019] [Indexed: 10/25/2022]
Abstract
The aim of this work was to optimise a soybean/cornstarch extrudate by adjusting a central composite design and to maximise a product with a high protein and resistant starch (RS) content by evaluating the indigestible fractions through in vitro colonic fermentation and production of short-chain fatty acids (SCFAs) with potential health benefits. According to the response surface analysis and RS maximisation results, an optimisation of the independent variables was obtained as follows: 32.5% feed moisture, 144 °C extrusion temperature and a proportion of 44% germinated soybean flour and 56% cornstarch. A product with a 2.11% expansion index, 6.25 N hardness, a glycaemic index of 49 and 12% resistant starch was obtained. The optimised extrudate showed a 36% indigestible fraction and high fermentability with respect to that of the lactulose control. Furthermore, the decrease in pH was inversely proportional to the production of SCFAs and the volume of gas generated. Acetic, propionic, and butyric acids were produced at a molar ratio of 62:27:11, while the highest SCFA concentrations were found 48 h after incubation. The RS of the optimised extruder was a viable substrate for in vitro colonic fermentation, suggesting that it is a good food source to produce SCFAs, which could exert an effect on the regulation of lipid and glucose metabolism.
Collapse
|
209
|
Nan H, Kuroda K, Takahashi K, Anderson JL. Examining the unique retention behavior of volatile carboxylic acids in gas chromatography using zwitterionic liquid stationary phases. J Chromatogr A 2019; 1603:288-296. [DOI: 10.1016/j.chroma.2019.06.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/27/2019] [Accepted: 06/07/2019] [Indexed: 10/26/2022]
|
210
|
Oral Administration of Compound Probiotics Ameliorates HFD-Induced Gut Microbe Dysbiosis and Chronic Metabolic Inflammation via the G Protein-Coupled Receptor 43 in Non-alcoholic Fatty Liver Disease Rats. Probiotics Antimicrob Proteins 2019; 11:175-185. [PMID: 29353414 DOI: 10.1007/s12602-017-9378-3] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aim of this study was to investigate how the effects of compound probiotics modulate the gut microbiota, short-chain fatty acid (SCFA), body composition, serum and liver lipids, and inflammatory markers in non-alcoholic fatty liver disease (NAFLD) rats. Twenty-four male SD rats were randomly divided into 3 groups: normal control group (standard feed), high-fat diet (HFD) feeding group (83% standard feed + 10% lard oil + 1.5% cholesterol + 0.5% cholate + 5% sucrose), and compound probiotics intervention group (HFD + 0.6 g × kg-1 × d-1 compound probiotics). The microbial population was assessed by 16S rDNA amplification and sequence analysis. Body composition, serum and liver lipids, serum inflammatory markers, colonic SCFAs, and relative proteins were assessed. The results showed that compound probiotics significantly reduced body weight, visceral and total fat mass, and the levels of hepatic TC and TG and serum TG, FFA, ALT, LPS, IL-1β, and IL-18 (P < 0.05). The proportions of TM7 phylum (0.06 vs 1.57%, P < 0.05) clearly increased, while that of Verrucomicrobia phylum (5.69 vs 2.61%, P < 0.05) clearly decreased. Compound probiotics also increased the representation of Ruminococcus genus (0.95 vs 1.83%, P < 0.05), while the proportion of Veillonella genus decreased (0.10 vs 0.03%, P < 0.05). The levels of colonic SCFAs and GPR43, NLRP3, ASC, and CASPASE-1 proteins also changed significantly (P < 0.05). Compound probiotics modulated gut microbiota, SCFAs, and their receptor GPR43 in NAFLD rats. These changes might inhibit lipid deposition and chronic metabolic inflammation in response to the insult of HFD.
Collapse
|
211
|
Wang Y, Mortimer EK, Katundu KGH, Kalanga N, Leong LEX, Gopalsamy GL, Christophersen CT, Richard AC, Shivasami A, Abell GCJ, Young GP, Rogers GB. The Capacity of the Fecal Microbiota From Malawian Infants to Ferment Resistant Starch. Front Microbiol 2019; 10:1459. [PMID: 31316490 PMCID: PMC6611432 DOI: 10.3389/fmicb.2019.01459] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/11/2019] [Indexed: 01/10/2023] Open
Abstract
In Low and Middle-Income Countries (LMIC), weaning is associated with environmentally acquired and inflammation-associated enteric disorders. Dietary intake of high amylose maize starch (HAMS) can promote commensal fermentative bacteria and drive the production of short chain fatty acids (SCFAs). By stabilizing commensal gut microbiology, and stimulating the production of anti-inflammatory metabolites, HAMS supplementation might therefore influence enteric health. However, the extent to which the gut microbiota of LMIC infants are capable of fermenting HAMS is unclear. We assessed the capacity of the fecal microbiota from pre-weaning and weaning Malawian infants to ferment HAMS and produce SCFAs using an in vitro fermentation model. Fecal microbiota from both pre-weaning and weaning infants were able to ferment HAMS, as indicated by an increase in bacterial load and total SCFA concentration, and a reduction in pH. All of these changes were more substantial in the weaning group. Acetate production was observed with both pre-weaning and weaning groups, while propionate production was only observed in the weaning group. HAMS fermentation resulted in significant alterations to the fecal microbial community in the weaning group, with significant increases in levels of Prevotella, Veillonella, and Collinsella associated with propionate production. In conclusion, fecal microbiota from Malawian infants before and during weaning has the capacity to produce acetate through HAMS fermentation, with propionate biosynthetic capability appearing only at weaning. Our results suggest that HAMS supplementation might provide benefit to infants during weaning.
Collapse
Affiliation(s)
- Yanan Wang
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - Elissa K. Mortimer
- Flinders University Global GI Health Unit, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Kondwani G. H. Katundu
- Division of Physiology, Biomedical Sciences Department, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Noel Kalanga
- Department of Health Systems and Policy, School of Public Health, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Lex E. X. Leong
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - Geetha L. Gopalsamy
- Flinders University Global GI Health Unit, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Claus T. Christophersen
- School of Medical & Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- School of Molecular & Life Sciences, Curtin University, Perth, WA, Australia
| | - Alyson C. Richard
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - Aravind Shivasami
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - Guy C. J. Abell
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Graeme P. Young
- Flinders University Global GI Health Unit, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Geraint B. Rogers
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
212
|
Liu HY, Walden TB, Cai D, Ahl D, Bertilsson S, Phillipson M, Nyman M, Holm L. Dietary Fiber in Bilberry Ameliorates Pre-Obesity Events in Rats by Regulating Lipid Depot, Cecal Short-Chain Fatty Acid Formation and Microbiota Composition. Nutrients 2019; 11:nu11061350. [PMID: 31208043 PMCID: PMC6627426 DOI: 10.3390/nu11061350] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/05/2019] [Accepted: 06/11/2019] [Indexed: 12/19/2022] Open
Abstract
Obesity is linked to non-alcoholic fatty liver disease and risk factors associated to metabolic syndrome. Bilberry (Vaccinium myrtillus) that contains easily fermentable fiber may strengthen the intestinal barrier function, attenuate inflammation and modulate gut microbiota composition, thereby prevent obesity development. In the current study, liver lipid metabolism, fat depot, cecal and serum short-chain fatty acids (SCFAs) and gut microbiome were evaluated in rats fed bilberries in a high-fat (HFD + BB) or low-fat (LFD + BB) setting for 8 weeks and compared with diets containing equal amount of fiber resistant to fermentation (cellulose, HFD and LFD). HFD fed rats did not obtain an obese phenotype but underwent pre-obesity events including increased liver index, lipid accumulation and increased serum cholesterol levels. This was linked to shifts of cecal bacterial community and reduction of major SCFAs. Bilberry inclusion improved liver metabolism and serum lipid levels. Bilberry inclusion under either LFD or HFD, maintained microbiota homeostasis, stimulated interscapular-brown adipose tissue depot associated with increased mRNA expression of uncoupling protein-1; enhanced SCFAs in the cecum and circulation; and promoted butyric acid and butyrate-producing bacteria. These findings suggest that bilberry may serve as a preventative dietary measure to optimize microbiome and associated lipid metabolism during or prior to HFD.
Collapse
Affiliation(s)
- Hao-Yu Liu
- Department of Medical Cell Biology, Uppsala University, 75123 Uppsala, Sweden.
| | - Tomas B Walden
- Department of Medical Cell Biology, Uppsala University, 75123 Uppsala, Sweden.
| | - Demin Cai
- Department of Biochemistry and Molecular Medicine, University of California at Davis, Sacramento, CA 95817, USA.
| | - David Ahl
- Department of Medical Cell Biology, Uppsala University, 75123 Uppsala, Sweden.
| | - Stefan Bertilsson
- Department of Ecology and Genetics, Limnology and Science for Life Laboratory, Uppsala University, 75236 Uppsala, Sweden.
| | - Mia Phillipson
- Department of Medical Cell Biology, Uppsala University, 75123 Uppsala, Sweden.
| | - Margareta Nyman
- Department of Food Technology, Engineering and Nutrition, Lund University, 22100 Lund, Sweden.
| | - Lena Holm
- Department of Medical Cell Biology, Uppsala University, 75123 Uppsala, Sweden.
| |
Collapse
|
213
|
Monovalerin and trivalerin increase brain acetic acid, decrease liver succinic acid, and alter gut microbiota in rats fed high-fat diets. Eur J Nutr 2019. [DOI: 10.1007/s00394-018-1688-z and 21=21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
|
214
|
Effect of a polyphenol-rich plant matrix on colonic digestion and plasma antioxidant capacity in a porcine model. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
215
|
Maurer LH, Cazarin CBB, Quatrin A, Minuzzi NM, Costa EL, Morari J, Velloso LA, Leal RF, Rodrigues E, Bochi VC, Júnior MRM, Emanuelli T. Grape peel powder promotes intestinal barrier homeostasis in acute TNBS-colitis: A major role for dietary fiber and fiber-bound polyphenols. Food Res Int 2019; 123:425-439. [PMID: 31284994 DOI: 10.1016/j.foodres.2019.04.068] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 04/28/2019] [Accepted: 04/29/2019] [Indexed: 02/07/2023]
Abstract
Inflammatory bowel diseases are characterized by impaired intestinal barrier function. This study aimed to evaluate the effects of grape peel powder (GPP) and its bioactive rich-fractions on the barrier function and colonic injury in a model of colitis induced by 2,4,6 trinitrobenzene sulfonic acid (TNBS). Wistar rats received diets supplemented with either GPP (8%), extractable polyphenols (EP), non-extractable polyphenols-rich fraction (NEP-F), or polyphenols-poor, fiber-rich fraction (F) from grapes at amounts equivalent to the GPP group during 15 days before and for 7 days after colitis induction. NEP-F has decreased the extension of colonic lesion but the other grape peel bioactive fractions did not protect against macroscopic or microscopic colonic damage, EP diet increased macroscopic colonic damage. GPP, EP, and NEP-F reduced claudin-2 mRNA expression, whereas GPP and F fraction increased occludin and ZO-1 mRNA expression. All experimental diets reduced the colitis-triggered increase of MMP-9 mRNA expression. Colitis reduced by 30% the production of cecal short-chain fatty acids (SCFA). GPP and NEP-F completely protected against this effect, whereas F fraction was ineffective. Only GPP and NEP-F were able to decrease the upregulation of GRP94 mRNA triggered by colitis. Dietary fiber seems to reestablish the intestinal barrier function, whereas fiber-bound phenolics were able to restore cecal metabolism to produce beneficial metabolites like SCFA and to reduce the activation of the unfolded protein response.
Collapse
Affiliation(s)
- Luana Haselein Maurer
- Graduate Program on Food Science and Technology, Center of Rural Sciences, Federal University of Santa Maria, 97105-900 Santa Maria, Rio Grande do Sul, Brazil; Federal Institute of Education, Science, and Technology Farroupilha, 97555-000 Alegrete, Rio Grande do Sul, Brazil
| | - Cinthia Baú Betim Cazarin
- School of Food Engineering, Department of Food and Nutrition, University of Campinas, Campinas, São Paulo 13083-862, Brazil
| | - Andréia Quatrin
- Federal Institute of Education, Science, and Technology Farroupilha, 97555-000 Alegrete, Rio Grande do Sul, Brazil
| | - Natália Machado Minuzzi
- Integrated Center for Laboratory Analysis Development (NIDAL), Department of Food Technology and Science, Center of Rural Sciences, Federal University of Santa Maria, 97105-900 Santa Maria, Rio Grande do Sul, Brazil
| | - Eduarda Lasch Costa
- Integrated Center for Laboratory Analysis Development (NIDAL), Department of Food Technology and Science, Center of Rural Sciences, Federal University of Santa Maria, 97105-900 Santa Maria, Rio Grande do Sul, Brazil
| | - Joseane Morari
- School of Medical Sciences, Laboratory of Cell Signaling, University of Campinas, Campinas, São Paulo 13084-970, Brazil
| | - Lício Augusto Velloso
- School of Medical Sciences, Laboratory of Cell Signaling, University of Campinas, Campinas, São Paulo 13084-970, Brazil
| | - Raquel Franco Leal
- School of Medical Sciences, Department of Surgery, University of Campinas, Campinas, São Paulo 13083-887, Brazil
| | - Eliseu Rodrigues
- Federal University of Rio Grande do Sul, Institute of Food Science and Technology, 91501-970 Porto Alegre, RS, Brazil
| | - Vivian Caetano Bochi
- Federal University of Health Sciences of Porto Alegre, Department of Nutrition, 90050-170 Porto Alegre, RS, Brazil
| | - Mário Roberto Maróstica Júnior
- School of Food Engineering, Department of Food and Nutrition, University of Campinas, Campinas, São Paulo 13083-862, Brazil
| | - Tatiana Emanuelli
- Graduate Program on Food Science and Technology, Center of Rural Sciences, Federal University of Santa Maria, 97105-900 Santa Maria, Rio Grande do Sul, Brazil; Integrated Center for Laboratory Analysis Development (NIDAL), Department of Food Technology and Science, Center of Rural Sciences, Federal University of Santa Maria, 97105-900 Santa Maria, Rio Grande do Sul, Brazil.
| |
Collapse
|
216
|
Hsu YL, Chen CC, Lin YT, Wu WK, Chang LC, Lai CH, Wu MS, Kuo CH. Evaluation and Optimization of Sample Handling Methods for Quantification of Short-Chain Fatty Acids in Human Fecal Samples by GC–MS. J Proteome Res 2019; 18:1948-1957. [DOI: 10.1021/acs.jproteome.8b00536] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Ya-Lin Hsu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei City, Taiwan
- The Metabolomics Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei City, Taiwan
| | - Chieh-Chang Chen
- Departments of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Ya-Ting Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei City, Taiwan
- The Metabolomics Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei City, Taiwan
| | - Wei-Kai Wu
- Department of Internal Medicine, National Taiwan University Hospital Bei-Hu Branch, Taipei City, Taiwan
- Institute of Food Science and Technology, National Taiwan University, Taipei City, Taiwan
| | - Lin-Chau Chang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Chang-Hao Lai
- Departments of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Ming-Shiang Wu
- Departments of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Ching-Hua Kuo
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei City, Taiwan
- The Metabolomics Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei City, Taiwan
- Department of Pharmacy, National Taiwan University Hospital, Taipei City, Taiwan
| |
Collapse
|
217
|
Dobranowski PA, Tang C, Sauvé JP, Menzies SC, Sly LM. Compositional changes to the ileal microbiome precede the onset of spontaneous ileitis in SHIP deficient mice. Gut Microbes 2019; 10:578-598. [PMID: 30760087 PMCID: PMC6748580 DOI: 10.1080/19490976.2018.1560767] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Inflammatory bowel disease, encompassing both ulcerative colitis and Crohn's disease, is characterized by chronic, relapsing-remitting gastrointestinal inflammation of unknown etiology. SHIP deficient mice develop fully penetrant, spontaneous ileitis at 6 weeks of age, and thus offer a tractable model of Crohn's disease-like inflammation. Since disruptions to the microbiome are implicated in the pathogenesis of Crohn's disease, we conducted a 16S rRNA gene survey of the ileum, cecum, colon, and stool contents of SHIP+/+ and SHIP-/- mice. We predicted that diversity and compositional changes would occur after, and possibly prior to, the onset of overt disease. No differences were found in alpha diversity, but significant changes in beta diversity and specific commensal populations were observed in the ileal compartment of SHIP deficient mice after the onset of overt disease. Specifically, reductions in the Bacteroidales taxa, Muribaculum intestinale, and an expansion in Lactobacillus were most notable. In contrast, expansions to bacterial taxa previously associated with inflammation, including Bacteroides, Parabacteroides, and Prevotella were observed in the ilea of SHIP deficient mice prior to the onset of overt disease. Finally, antibiotic treatment reduced the development of intestinal inflammation in SHIP-/- mice. Thus, our findings indicate that SHIP is involved in maintaining ileal microbial homeostasis. These results have broader implications for humans, since reduced SHIP protein levels have been reported in people with Crohn's disease.
Collapse
Affiliation(s)
| | | | | | | | - Laura May Sly
- University of British Columbia,BC Children’s Hospital research institute,CONTACT Laura May Sly BC Children’s Hospital research institute, 950 West 28th Avenue, A5-142TRB, Vancouver, British Columbia V5Z 4H4, Canada
| |
Collapse
|
218
|
de Almeida Bianchini Campos RC, Martins EMF, de Andrade Pires B, do Carmo Gouveia Peluzio M, da Rocha Campos AN, Ramos AM, de Castro Leite Júnior BR, de Oliveira Martins AD, da Silva RR, Martins ML. In vitro and in vivo resistance of Lactobacillus rhamnosus GG carried by a mixed pineapple (Ananas comosus L. Merril) and jussara (Euterpe edulis Martius) juice to the gastrointestinal tract. Food Res Int 2019; 116:1247-1257. [DOI: 10.1016/j.foodres.2018.10.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 09/22/2018] [Accepted: 10/02/2018] [Indexed: 12/12/2022]
|
219
|
Monobutyrin Reduces Liver Cholesterol and Improves Intestinal Barrier Function in Rats Fed High-Fat Diets. Nutrients 2019; 11:nu11020308. [PMID: 30717248 PMCID: PMC6412756 DOI: 10.3390/nu11020308] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/15/2019] [Accepted: 01/23/2019] [Indexed: 01/14/2023] Open
Abstract
Butyric acid has been shown to reduce high-fat diet-related metabolic disturbances and to improve intestinal barrier function due to its potent anti-inflammatory capacity. This study investigates whether a butyric acid ester, monobutyrin (MB) affects lipid profiles and gut barrier function in a dose-response manner in rats fed butter- or lard-based high-fat diets. Four-week-old male Wistar rats were fed butter-based diets containing 0, 0.25, 0.75 and 1.5 MB g/100 g (dry weight basis) or 0.5 glycerol g/100 g, and diets with lard (La) containing 0 and 0.5 MB g/100 g or a low-fat control diet for 3⁻4 weeks. Lipid profiles in blood and liver tissue, intestinal permeability and cecal short-chain fatty acids were examined. The results showed a dose-dependent decrease in liver total cholesterol for 1.5 MB (p < 0.05) and liver triglycerides for 0.75 MB (p < 0.05) and 1.5 MB (p = 0.08) groups compared to the high-fat control group. Furthermore, a lower excretion of mannitol in urine in the 1.5 MB group indicated improved intestinal barrier function. When MB was supplemented in the lard-based diet, serum total cholesterol levels decreased, and total amount of liver high-density lipoprotein-cholesterol increased. Thus, MB dietary supplementation can be effective in counteracting lipid metabolism disturbances and impaired gut barrier function induced by high-fat diets.
Collapse
|
220
|
Description and genomic characterization of Massiliimalia massiliensis gen. nov., sp. nov., and Massiliimalia timonensis gen. nov., sp. nov., two new members of the family Ruminococcaceae isolated from the human gut. Antonie van Leeuwenhoek 2019; 112:905-918. [DOI: 10.1007/s10482-018-01223-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/29/2018] [Indexed: 12/16/2022]
|
221
|
Phoenicibacter congonensis gen. nov., sp. nov., a new genus isolated from the human gut and its description using a taxonogenomic approach. Antonie van Leeuwenhoek 2019; 112:775-784. [PMID: 30666529 DOI: 10.1007/s10482-018-01211-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/01/2018] [Indexed: 10/27/2022]
Abstract
Culturomics has recently allowed the isolation and description of previously uncultured bacteria from the human microbiome at different body sites. As part of a project aiming to describe the human gut microbiota by culturomics, Phoenicibacter congonensis strain Marseille-P3241T was isolated from the gut of a 45 years old Pygmy female. In the present work, we aim to describe this strain via the taxonogenomics approach. The major phenotypic, genomic and biochemical characteristics of this strain were analysed. Strain Marseille-P3241T is an anaerobic, Gram-positive and motile coccobacillus that grows optimally at 37 °C. The genome of strain Marseille-P3241T is 1,447,956 bp long with 43.44% GC content and its 16S rRNA gene sequence exhibited 89% sequence similarity with that of Denitrobacterium detoxificans strain NPOH1T, the phylogenetically closest related species with current standing in nomenclature. After performing a phylogenetic and genomic analysis, we conclude that strain Marseille-P3241T (= CCUG 70681T = CSUR P3241T) represents the type species of a new genus, for which we propose the name Phoenicibacter congonensis gen. nov., sp. nov.
Collapse
|
222
|
Dysgonomonas massiliensis sp. nov., a new species isolated from the human gut and its taxonogenomic description. Antonie van Leeuwenhoek 2019; 112:935-945. [DOI: 10.1007/s10482-019-01227-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/05/2019] [Indexed: 12/22/2022]
|
223
|
Liu H, Hou C, Li N, Zhang X, Zhang G, Yang F, Zeng X, Liu Z, Qiao S. Microbial and metabolic alterations in gut microbiota of sows during pregnancy and lactation. FASEB J 2019; 33:4490-4501. [PMID: 30653349 DOI: 10.1096/fj.201801221rr] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The gut microbiota plays a critical role in various physiologic processes; however, maternal microbial and metabolic changes during pregnancy and lactation remain elusive. Using pigs as an animal model, we conducted comparative analyses of gut microbiota and short-chain fatty acid (SCFA) profiles across different stages of gestation, lactation, and the empty (nonpregnancy) phase in 2 distinct breeds of sow, Rongchang (RS) and Landrace (LS). Coriobacteriaceae were found to gradually increase over gestational time irrespective of breed, which was further validated in an independent cohort of sows, indicating that Coriobacteriaceae are likely associated with the progression of pregnancy. Escherichia increased as well. Relative to empty and gestation, lactation was associated with an increase in SCFA producers and a concomitant augmentation in SCFA production in both breeds. A comparison between the 2 breeds revealed that Ruminococcaceae were more abundant in RSs than in LSs, consistent with the strong ability of Rongchang pigs to digest highly fibrous feedstuffs. Taken together, we revealed characteristic structural and metabolic changes in maternal gut microbiota throughout pregnancy, lactation, and the empty phase, which could potentially help improve the pregnancy and lactation outcomes for both animals and humans.-Liu, H., Hou, C., Li, N., Zhang, X., Zhang, G., Yang, F., Zeng, X., Liu, Z., Qiao, S. Microbial and metabolic alterations in gut microbiota of sows during pregnancy and lactation.
Collapse
Affiliation(s)
- Hongbin Liu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing Key Laboratory of Bio-Feed Additives, Beijing, China
| | - Chengli Hou
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Ning Li
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing Key Laboratory of Bio-Feed Additives, Beijing, China
| | - Xiaoya Zhang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing Key Laboratory of Bio-Feed Additives, Beijing, China
| | - Guolong Zhang
- Department of Animal Science, Oklahoma State University, Stillwater, Oklahoma, USA; and
| | - Feiyun Yang
- Chongqing Academy of Animal Science, Chongqing, China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing Key Laboratory of Bio-Feed Additives, Beijing, China
| | - Zuohua Liu
- Chongqing Academy of Animal Science, Chongqing, China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing Key Laboratory of Bio-Feed Additives, Beijing, China
| |
Collapse
|
224
|
Luan H, Wang X, Cai Z. Mass spectrometry-based metabolomics: Targeting the crosstalk between gut microbiota and brain in neurodegenerative disorders. MASS SPECTROMETRY REVIEWS 2019; 38:22-33. [PMID: 29130504 DOI: 10.1002/mas.21553] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/12/2017] [Indexed: 05/10/2023]
Abstract
Metabolomics seeks to take a "snapshot" in a time of the levels, activities, regulation and interactions of all small molecule metabolites in response to a biological system with genetic or environmental changes. The emerging development in mass spectrometry technologies has shown promise in the discovery and quantitation of neuroactive small molecule metabolites associated with gut microbiota and brain. Significant progress has been made recently in the characterization of intermediate role of small molecule metabolites linked to neural development and neurodegenerative disorder, showing its potential in understanding the crosstalk between gut microbiota and the host brain. More evidence reveals that small molecule metabolites may play a critical role in mediating microbial effects on neurotransmission and disease development. Mass spectrometry-based metabolomics is uniquely suitable for obtaining the metabolic signals in bidirectional communication between gut microbiota and brain. In this review, we summarized major mass spectrometry technologies including liquid chromatography-mass spectrometry, gas chromatography-mass spectrometry, and imaging mass spectrometry for metabolomics studies of neurodegenerative disorders. We also reviewed the recent advances in the identification of new metabolites by mass spectrometry and metabolic pathways involved in the connection of intestinal microbiota and brain. These metabolic pathways allowed the microbiota to impact the regular function of the brain, which can in turn affect the composition of microbiota via the neurotransmitter substances. The dysfunctional interaction of this crosstalk connects neurodegenerative diseases, including Parkinson's disease, Alzheimer's disease and Huntington's disease. The mass spectrometry-based metabolomics analysis provides information for targeting dysfunctional pathways of small molecule metabolites in the development of the neurodegenerative diseases, which may be valuable for the investigation of underlying mechanism of therapeutic strategies.
Collapse
Affiliation(s)
- Hemi Luan
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
| | - Xian Wang
- Key Laboratory of Analytical Chemistry of State Ethnic Affairs Commission, College of Chemistry and Materials Science, South-Central University for Nationalities, Wuhan, Hubei, China
| | - Zongwei Cai
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
| |
Collapse
|
225
|
Li X, Wang H, Wang T, Zheng F, Wang H, Wang C. Dietary wood pulp-derived sterols modulation of cholesterol metabolism and gut microbiota in high-fat-diet-fed hamsters. Food Funct 2019; 10:775-785. [DOI: 10.1039/c8fo02271b] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Wood pulp-derived sterols (WS) supplementation ameliorated HFD-associated metabolic disorder; WS supplementation increased the amounts of fecal sterols excretion and SCFAs content; WS supplementation modulated gut microbiota composition.
Collapse
Affiliation(s)
- Xiang Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health
- Beijing Technology & Business University (BTBU)
- Beijing 100048
- China
- Beijing Laboratory of Food Quality and Safety
| | - Huali Wang
- State Key Laboratory of Food Nutrition and Safety
- Tianjin University of Science and Technology (TUST)
- Tianjin 300457
- China
| | - Tianxin Wang
- State Key Laboratory of Food Nutrition and Safety
- Tianjin University of Science and Technology (TUST)
- Tianjin 300457
- China
| | - Fuping Zheng
- Beijing Laboratory of Food Quality and Safety
- Beijing Technology and Business University
- Beijing 100048
- China
| | - Hao Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health
- Beijing Technology & Business University (BTBU)
- Beijing 100048
- China
- State Key Laboratory of Food Nutrition and Safety
| | - Chengtao Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health
- Beijing Technology & Business University (BTBU)
- Beijing 100048
- China
| |
Collapse
|
226
|
Gong L, Wang T, Sun C, Wang J, Sun B. Whole barley prevents obesity and dyslipidemia without the involvement of the gut microbiota in germ free C57BL/6J obese mice. Food Funct 2019; 10:7498-7508. [DOI: 10.1039/c9fo01268k] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Whole barley (WB) consumption is the subject of renewed interest because of its health benefits.
Collapse
Affiliation(s)
- Lingxiao Gong
- China-Canada Joint Lab of Food Nutrition and Health (Beijing)
- Beijing Advanced Innovation Center for Food Nutrition and Human Health (BTBU)
- Beijing Engineering and Technology Research Center of Food Additives
- Beijing Technology & Business University (BTBU)
- Beijing 100048
| | - Tianxi Wang
- China-Canada Joint Lab of Food Nutrition and Health (Beijing)
- Beijing Advanced Innovation Center for Food Nutrition and Human Health (BTBU)
- Beijing Engineering and Technology Research Center of Food Additives
- Beijing Technology & Business University (BTBU)
- Beijing 100048
| | - Cong Sun
- China-Canada Joint Lab of Food Nutrition and Health (Beijing)
- Beijing Advanced Innovation Center for Food Nutrition and Human Health (BTBU)
- Beijing Engineering and Technology Research Center of Food Additives
- Beijing Technology & Business University (BTBU)
- Beijing 100048
| | - Jing Wang
- China-Canada Joint Lab of Food Nutrition and Health (Beijing)
- Beijing Advanced Innovation Center for Food Nutrition and Human Health (BTBU)
- Beijing Engineering and Technology Research Center of Food Additives
- Beijing Technology & Business University (BTBU)
- Beijing 100048
| | - Baoguo Sun
- China-Canada Joint Lab of Food Nutrition and Health (Beijing)
- Beijing Advanced Innovation Center for Food Nutrition and Human Health (BTBU)
- Beijing Engineering and Technology Research Center of Food Additives
- Beijing Technology & Business University (BTBU)
- Beijing 100048
| |
Collapse
|
227
|
Gong L, Chi H, Wang J, Zhang H, Sun B. In vitro fermentabilities of whole wheat as compared with refined wheat in different cultivars. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.11.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
228
|
Chen M, Hui S, Lang H, Zhou M, Zhang Y, Kang C, Zeng X, Zhang Q, Yi L, Mi M. SIRT3 Deficiency Promotes High-Fat Diet-Induced Nonalcoholic Fatty Liver Disease in Correlation with Impaired Intestinal Permeability through Gut Microbial Dysbiosis. Mol Nutr Food Res 2018; 63:e1800612. [PMID: 30525304 DOI: 10.1002/mnfr.201800612] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 11/15/2018] [Indexed: 12/11/2022]
Abstract
SCOPE Sirtuin 3 (SIRT3) plays a protective role against nonalcoholic fatty liver disease (NAFLD) by improving hepatic mitochondrial dysfunction. Gut microbiota imbalance contributes to the pathogenesis of NAFLD, yet the underlying mechanism linking SIRT3 with gut microbiota in NAFLD progression remains obscure. METHODS AND RESULTS Wild-type 129 mice and SIRT3 knockout (SIRT3KO) mice are placed under a chow diet or high-fat diet (HFD) treatment for 18 weeks. HFD resulted in a significantly increased hepatic steatosis and inflammation, which are exacerbated in SIRT3KO mice. The gut microbiota by 16s rRNA gene sequencing and phylogenetic reconstruction of unobserved states analysis are characterized. Lack of SIRT3 facilitates gut microbial dysbiosis in mice following HFD, with increased Desulfovibrio, Oscillibacter, and decreased Alloprevotella. SIRT3 deficiency resulted in an impaired intestinal permeability and inflammation in HFD-fed mice, which can be attenuated by sodium butyrate (NaB). SIRT3KO HFD-fed mice is followed by an increased lipopolysaccharide into the circulation and dysregulated expressions of cannabinoid receptor 1 and 2 in colon and liver, which are significantly associated with the alterations of intestinal microbiota. CONCLUSIONS SIRT3 deficiency promotes NAFLD progression in correlation with impaired intestinal permeability through gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Mengting Chen
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Suocheng Hui
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Hedong Lang
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Min Zhou
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Yong Zhang
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Chao Kang
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Xianglong Zeng
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Qianyong Zhang
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Long Yi
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Mantian Mi
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, 400038, P. R. China
| |
Collapse
|
229
|
Zhang S, Wang H, Zhu MJ. A sensitive GC/MS detection method for analyzing microbial metabolites short chain fatty acids in fecal and serum samples. Talanta 2018; 196:249-254. [PMID: 30683360 DOI: 10.1016/j.talanta.2018.12.049] [Citation(s) in RCA: 235] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/14/2018] [Accepted: 12/15/2018] [Indexed: 01/23/2023]
Abstract
Gut microbiota and their major metabolites, short-chain fatty acids (SCFAs), are recognized as important players in gut homeostasis and metabolic disease occurance. A convenient and sensitive detection method is needed to profile SCFAs in limited and complex biological samples. The gas chromatography/mass spectrometry (GC/MS) is the most common method for SCFAs profiling in biological samples. Trimethylsilyl (TMS) derivatization reagents such as N, O-bis(trimethyl-silyl)-trifluoroacetamide (BSTFA) are commonly used in GC/MS analysis to improve sensitivity and accuracy, but they were barely used in SCFA analysis due to their sensitivity to moisture and the volatility of SCFAs. Here, we developed a rapid, convenient and reliable method for SCFAs profiling in small amounts of fecal and serum samples by GC/MS using BSTFA in combination with sodium sulfate dehydration pretreatment. SCFAs were extracted with anhydrous ether from acidified fecal water extract or serum samples, followed by dehydration with sodium sulfate and BSTFA derivatization at a reduced temperature. Select ion monitoring mode was used for highly sensitive quantification of SCFAs by GC/MS. The derivation with BSTFA at 37 °C or 22 °C showed an excellent linearity (R2 > 0.999), good recoveries (81.27-128.42%), high repeatability (RSD < 2%) and low limit of detections (LODs) of different SCFAs ranging from 0.064 to 0.067 µM. All major SCFAs including acetic acid, propionic acid, isobutyric acid, butyric acid, isovaleric acid and valeric acid were identified and quantified accurately in fecal and serum samples. In conclusions, a reliable, convenient and sensitive method wasdeveloped for the measurement of SCFA and other volatile compounds in small biological samples using sodium sulfate dehydration pretreatment and BSTFA derivatization-based GC/MS analyses.
Collapse
Affiliation(s)
- Shuming Zhang
- School of Food Science, Washington State University, Pullman, WA 99164, USA
| | - Hongbin Wang
- School of Food Science, Washington State University, Pullman, WA 99164, USA
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
230
|
da Silva-Maia JK, Batista AG, Correa LC, Lima GC, Bogusz Junior S, Maróstica Junior MR. Aqueous extract of berry (Plinia jaboticaba) byproduct modulates gut microbiota and maintains the balance on antioxidant defense system in rats. J Food Biochem 2018; 43:e12705. [PMID: 31353652 DOI: 10.1111/jfbc.12705] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 08/16/2018] [Accepted: 09/13/2018] [Indexed: 02/02/2023]
Abstract
Previous studies have assessed the properties of aqueous extracts, using byproducts such as jaboticaba peel. We have assessed potential antioxidant effects of jaboticaba extract (Plinia jaboticaba) (JAE = 50 g/L) in vitro and in vivo. Healthy Wistar rats received ad libitum JAE for either 15 or 49 days in vivo. Cyanidin-3-O-glucoside, delphinidin-3-O-glucoside, gallic acid, rutin, myricetin, and quercetin were identified as the main polyphenols in JAE. Lipid peroxidation values in the serum and colon were similar throughout the groups. In addition, JAE did not disturb the antioxidant systems. JAE also altered gut microbiota, increasing since Lactobacillus, Bifidobacterium and Enterobacteriaceae counts. Bacterial metabolites were higher in the colon content of rats fed with JAE than in the control group. Given these results, under healthy conditions, JAE dietary supplementation could perform in vivo modulation of gut microbiota, without disturbing the antioxidant system. PRACTICAL APPLICATION: Jaboticaba (Plinia jaboticaba) peel is a rich and often-wasted source of bioactive compounds, such as polyphenols. Previous studies have shown that physiological benefits of this berry. The jaboticaba peel could contribute to antioxidant defense systems; it may also have an effect over gut microbiota related to polyphenols contents. Aqueous extraction may be a practical way of employing the bioactive compounds of jaboticaba peel; these compounds can be consumed daily and safely, and thus have attracted particular attention. This work showed positive impacts of jaboticaba peel treatments on microbiota and antioxidant defense systems, and could guide future clinical studies.
Collapse
Affiliation(s)
- Juliana Kelly da Silva-Maia
- Faculty of Food Engineering, Department of Food and Nutrition, University of Campinas (UNICAMP), Campinas, Brazil
| | - Angela Giovana Batista
- Faculty of Food Engineering, Department of Food and Nutrition, University of Campinas (UNICAMP), Campinas, Brazil.,Department of Food and Nutrition, Federal University of Santa Maria (UFSM, Campus Palmeira das Missões), Palmeira das Missões, Brazil
| | - Luiz Claudio Correa
- Brazilian Agricultural Research Corporation, Embrapa Tropical Semi-arid, Petrolina, Brazil
| | - Glaucia Carielo Lima
- Faculty of Food Engineering, Department of Food and Nutrition, University of Campinas (UNICAMP), Campinas, Brazil.,Faculty of Nutrition, Federal University of Goiás (UFG), Goiânia, Brazil
| | | | | |
Collapse
|
231
|
Qiu D, Xia Z, Jiao X, Deng J, Zhang L, Li J. Altered Gut Microbiota in Myasthenia Gravis. Front Microbiol 2018; 9:2627. [PMID: 30483222 PMCID: PMC6241162 DOI: 10.3389/fmicb.2018.02627] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 10/15/2018] [Indexed: 01/01/2023] Open
Abstract
Myasthenia gravis (MG) is an autoimmune-mediated disorder, the etiology of which involves both environmental factors and genetics. While the exact factors responsible for predisposition to MG remain elusive, it is hypothesized that gut microbiota play a critical role in the pathogenesis of MG. This study investigated whether gut microbiota are altered in MG patients by comparing the fecal microbiota profiles of MG patients to those of age- and sex-matched healthy controls. Phylotype profiles of gut microbial populations were generated using hypervariable tag sequencing of the V4 region of the 16S ribosomal RNA gene. Fecal short-chain fatty acids (SCFAs) were assessed by gas chromatographic analyses. The results demonstrated that, compared to the healthy cohort, the gut microbiota of the MG group was changed in terms of the relative abundances of bacterial taxa, with sharply reduced microbial richness, particularly in the genus Clostridium. The fecal SCFA content was significantly lower in the MG group. Furthermore, microbial dysbiosis was closely related to the levels of inflammatory biomarkers in the sera of MG patients.
Collapse
Affiliation(s)
- Dongxu Qiu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Neurology of Hunan Province, Changsha, China
| | - Zhiwei Xia
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Neurology of Hunan Province, Changsha, China
| | - Xiao Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Neurology of Hunan Province, Changsha, China
| | - Jun Deng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Neurology of Hunan Province, Changsha, China
| | - Lei Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Neurology of Hunan Province, Changsha, China
| |
Collapse
|
232
|
Konkol Y, Keskitalo A, Vuorikoski H, Pietilä S, Elo LL, Munukka E, Bernoulli J, Tuomela J. Chronic nonbacterial prostate inflammation in a rat model is associated with changes of gut microbiota that can be modified with a galactoglucomannan-rich hemicellulose extract in the diet. BJU Int 2018; 123:899-908. [PMID: 30256506 DOI: 10.1111/bju.14553] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES To investigate dietary effects on the gut microbiota composition in a rat model of nonbacterial chronic prostate inflammation (CPI). MATERIALS AND METHODS Nonbacterial CPI was induced in the Wistar rat strain with subcutaneous testosterone and 17β-oestradiol (E2 ) hormone pellets for 18 weeks. Rats with placebo pellets served as healthy controls. Rats with CPI were stratified into two groups, which drank either plain tap water (control group) or tap water supplemented with 2% galactoglucomannan-rich hemicellulose extract (GGM group) from Norway spruce (Picea abies) for 5 weeks. Faecal samples were collected at the end of the study, total DNA was extracted, and the bacterial composition was analysed by 16S rRNA gene sequencing. In addition, faecal samples were assayed for short-chain fatty acid (SCFA) concentrations using gas chromatography. Lipopolysaccharide-binding protein (LBP) was measured in serum samples, as an indirect indicator for bacterial lipopolysaccharide (LPS) load in blood. RESULTS The microbial biodiversity was significantly different between the treatment groups. In the rats with CPI, there was a significant increase in gut microbial populations Rikenellaceae, Odoribacter, Clostridiaceae, Allobaculum and Peptococcaceae compared with healthy rats. Conversely, levels of Bacteroides uniformis, Lactobacillus and Lachnospiraceae were decreased in rats with CPI. SCFA butyric-, valeric- and caproic-acid concentrations were also decreased in the faecal samples of the rats with CPI. In contrast, acetic acid concentrations and serum LBP were significantly elevated in CPI rats compared with healthy ones. Amongst rats with CPI, treatment with the GGM extract significantly reduced the abundance of Odoribacter and Clostridiaceae levels, and increased the B. uniformis levels compared with CPI rats drinking tap water only. In addition, GGM significantly increased the levels of butyric acid and caproic acid, and reduced the levels of LBP in serum. CONCLUSIONS Hormone-induced nonbacterial CPI in rats is associated with specific changes in gut microbiota and secondary changes in SCFAs and LPS due to gut microbiota alteration. Our results further suggest that fermentable compounds may have a beneficial effect on CPI.
Collapse
Affiliation(s)
- Yvonne Konkol
- Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
| | - Anniina Keskitalo
- Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
| | | | - Sami Pietilä
- Bioinformatics Unit, Turku Centre of Biotechnology, University of Turku, Åbo Akademi University, Turku, Finland
| | - Laura L Elo
- Bioinformatics Unit, Turku Centre of Biotechnology, University of Turku, Åbo Akademi University, Turku, Finland
| | - Eveliina Munukka
- Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
| | | | - Johanna Tuomela
- Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
| |
Collapse
|
233
|
Han X, Guo J, You Y, Yin M, Ren C, Zhan J, Huang W. A fast and accurate way to determine short chain fatty acids in mouse feces based on GC–MS. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1099:73-82. [DOI: 10.1016/j.jchromb.2018.09.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 08/24/2018] [Accepted: 09/11/2018] [Indexed: 12/20/2022]
|
234
|
Kang Y, Yang G, Zhang S, Ross CF, Zhu MJ. Goji Berry Modulates Gut Microbiota and Alleviates Colitis in IL-10-Deficient Mice. Mol Nutr Food Res 2018; 62:e1800535. [PMID: 30243032 DOI: 10.1002/mnfr.201800535] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/04/2018] [Indexed: 12/17/2022]
Abstract
SCOPE This study examines the beneficial effects of Goji berry against spontaneous colitis and its prebiotic role in IL-10-deficient mice. METHODS IL-10-deficient mice are assigned to a standard rodent diet (control) or a control diet supplemented with Goji (1% of dry feed weight) for 10 weeks, at which point colonic tissues and fecal contents are collected. RESULTS Goji supplementation decreases colonic pathobiological scores and mRNA expression of Il17a and Tgfb1, while it enhances Muc1 expression and fecal IgA content. Illumina MiSeq sequencing reveals that Goji supplementation increases Actinobacteria phylum, resulting in a bloom of Bifidobacteria in gut microbiota. Additionally, dietary Goji promotes butyrate-producing bacteria including Lachnospiraceae-Ruminococcaceae family and Roseburia spp. under Clostridium cluster XIVa. Furthermore, butyrate-producers Clostridium leptum and its dominant constituent Fecalibacterium prazusnitzii are markedly increased in the Goji group. Moreover, the gene-encoding butyryl-coenzyme A CoA transferase, a key enzyme responsible for butyrate synthesis in butyrate-producing bacteria, is increased sixfold in the fecal samples of Goji group associated with increased fecal butyrate content. CONCLUSION Data collectively show that dietary Goji results in the blooming of Bifidobacteria and butyrate-producing bacteria. These bacteria may cross-feed each other, conferring preventative effects against colitis in IL-10-deficient mice.
Collapse
Affiliation(s)
- Yifei Kang
- School of Food Science, Washington State University, Pullman, WA, 99164, USA
| | - Guan Yang
- School of Food Science, Washington State University, Pullman, WA, 99164, USA
| | - Shuming Zhang
- School of Food Science, Washington State University, Pullman, WA, 99164, USA
| | - Carolyn F Ross
- School of Food Science, Washington State University, Pullman, WA, 99164, USA
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA, 99164, USA
| |
Collapse
|
235
|
Al Hinai EA, Kullamethee P, Rowland IR, Swann J, Walton GE, Commane DM. Modelling the role of microbial p-cresol in colorectal genotoxicity. Gut Microbes 2018; 10:398-411. [PMID: 30359553 PMCID: PMC6546321 DOI: 10.1080/19490976.2018.1534514] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Background: A greater understanding of mechanisms explaining the interactions between diet and the gut microbiota in colorectal cancer is desirable. Genotoxic microbial metabolites present in the colon may be implicated in carcinogenesis and potentially influenced by diet. Aims: We hypothesised that microbial p-cresol is a colonic genotoxin and set out to model potential exposures in the colon and the effects of these exposures on colonic cells. Methods: Batch culture fermentations with human faecal inoculate were used to determine the synthesis of p-cresol and other metabolites in response to various substrates. The fermentation supernatants were evaluated for genotoxicity and the independent effects of p-cresol on colonic cells were studied in vitro. Results: In batch culture fermentation, supplementary protein increased the synthesis of phenols, indoles and p-cresol, whereas supplementary fructoligosaccharide (FOS) increased the synthesis of short chain fatty acids. The p-cresol was the greatest predictor of genotoxicity against colonocytes in the fermentation supernatants. Spiking fermentation supernatants with exogenous p-cresol further increased DNA damage, and independently p-cresol induced DNA damage in a dose-dependent manner against HT29 and Caco-2 cells and influenced cell cycle kinetics. Conclusions: In the colon p-cresol may reach physiologically significant concentrations which contribute to genotoxic exposures in the intestinal lumen, p-cresol production may be attenuated by substrate, and therefore diet, making it a potential modifiable biomarker of genotoxicity in the colon.
Collapse
Affiliation(s)
- Eiman Abdulla Al Hinai
- Department of Food and Nutritional Sciences, University of Reading, Reading, UK,Dietetics Department, Al Nahdha Hospital, Ministry of Health, Muscat, Sultanate of Oman
| | - Piyarach Kullamethee
- Department of innovation and technology of product development, Faculty of Agro-industry, King Mongkut’s University of Technology North Bangkok, Prachinburi, Thailand
| | - Ian R. Rowland
- Department of Food and Nutritional Sciences, University of Reading, Reading, UK
| | - Jonathan Swann
- Faculty of Medicine, Department of Surgery & Cancer, Imperial College London, London, UK
| | - Gemma E. Walton
- Department of Food and Nutritional Sciences, University of Reading, Reading, UK
| | - Daniel M. Commane
- Department of Applied and Health Sciences, University of Northumbria, Newcastle Upon Tyne, UK,CONTACT Daniel M. Commane Department of Applied and Health Sciences, University of Northumbria, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
236
|
Bilen M, Mbogning Fonkou MD, Nguyen TT, Richez M, Daoud Z, Fournier PE, Raoult D, Cadoret F. Miniphocibacter massiliensis gen. nov., sp. nov., a new species isolated from the human gut and its taxono-genomics description. Microbiologyopen 2018; 8:e00735. [PMID: 30280501 PMCID: PMC6528615 DOI: 10.1002/mbo3.735] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/11/2018] [Accepted: 08/20/2018] [Indexed: 12/23/2022] Open
Abstract
With the aim of describing the human microbiota by the means of culture methods, culturomics was developed in order to target previously un‐isolated bacterial species and describe it via the taxono‐genomics approach. While performing a descriptive study of the human gut microbiota of the pygmy people, strain Marseille‐P4678T has been isolated from a stool sample of a healthy 39‐year‐old pygmy male. Cells of this strain were Gram‐positive cocci, spore‐forming, non‐motile, catalase‐positive and oxidase‐negative, and grow optimally at 37°C under anaerobic conditions. Its 16S rRNA gene sequence exhibited 89.69% of sequence similarity with Parvimonas micra strain 3119BT (NR 036934.1), its phylogenetically closest species with standing in nomenclature. The genome of strain Marseille‐P4678T is 2,083,161 long with 28.26 mol% of G+C content. Based on its phenotypic, biochemical, genotypic and proteomic profile, this bacterium was classified as a new bacterial genus and species Miniphocibacter massiliensis gen. nov., sp. nov. with the type strain Marseille‐P4678T.
Collapse
Affiliation(s)
- Melhem Bilen
- Aix Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France.,Clinical Microbiology Department, Faculty of Medicine and Medical Sciences, University of Balamand, Amioun, Lebanon
| | | | - Thi T Nguyen
- Aix Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| | - Magali Richez
- Aix Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| | - Ziad Daoud
- Clinical Microbiology Department, Faculty of Medicine and Medical Sciences, University of Balamand, Amioun, Lebanon
| | - Pierre E Fournier
- Aix Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| | - Didier Raoult
- Aix Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France.,Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Frédéric Cadoret
- Aix Marseille Univ, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Marseille, France
| |
Collapse
|
237
|
Lu HY, Zeng H, Zhang L, Porres JM, Cheng WH. Fecal fermentation products of common bean-derived fiber inhibit C/EBPα and PPARγ expression and lipid accumulation but stimulate PPARδ and UCP2 expression in the adipogenesis of 3T3-L1 cells. J Nutr Biochem 2018; 60:9-15. [DOI: 10.1016/j.jnutbio.2018.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 05/22/2018] [Accepted: 06/06/2018] [Indexed: 12/19/2022]
|
238
|
Antibiotic treatment of rat dams affects bacterial colonization and causes decreased weight gain in pups. Commun Biol 2018; 1:145. [PMID: 30272021 PMCID: PMC6137057 DOI: 10.1038/s42003-018-0140-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022] Open
Abstract
Intergenerational transmission of bacteria during birth initiates the natural successional development of the intestinal microbiota in mammals. This process can be disrupted by antibiotic exposure, potentially affecting early-life microbiota-dependent metabolic programming. In the present study, we specifically investigate the metabolic consequences of exposing neonate Wistar rats to an antibiotic-perturbed low-diversity microbiota from birth until weaning, without exposing the pups directly to antibiotics. Here, we show that pups born from both amoxicillin and vancomycin-treated dams gain less weight than controls. This was concordant with lower feed intake as well as increased colonic expression of the PYY satiety hormone gene at weaning. The weight difference persists into adulthood even though the initial differences in gut microbiota subsided. Our results demonstrate that early-life exposure to an antibiotic-perturbed low-diversity microbiota is sufficient to cause changes in body weight persisting into adulthood.
Collapse
|
239
|
Jaboticaba berry peel intake increases short chain fatty acids production and prevent hepatic steatosis in mice fed high-fat diet. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.07.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
240
|
van de Wouw M, Boehme M, Lyte JM, Wiley N, Strain C, O'Sullivan O, Clarke G, Stanton C, Dinan TG, Cryan JF. Short-chain fatty acids: microbial metabolites that alleviate stress-induced brain-gut axis alterations. J Physiol 2018; 596:4923-4944. [PMID: 30066368 DOI: 10.1113/jp276431] [Citation(s) in RCA: 499] [Impact Index Per Article: 71.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 06/22/2018] [Indexed: 02/06/2023] Open
Abstract
KEY POINTS Chronic (psychosocial) stress changes gut microbiota composition, as well as inducing behavioural and physiological deficits. The microbial metabolites short-chain fatty acids (SCFAs) have been implicated in gastrointestinal functional, (neuro)immune regulation and host metabolism, but their role in stress-induced behavioural and physiological alterations is poorly understood. Administration of SCFAs to mice undergoing psychosocial stress alleviates enduring alterations in anhedonia and heightened stress-responsiveness, as well as stress-induced increases in intestinal permeability. In contrast, chronic stress-induced alterations in body weight gain, faecal SCFAs and the gene expression of the SCFA receptors FFAR2 and FFAR3 remained unaffected by SCFA supplementation. These results present novel insights into mechanisms underpinning the influence of the gut microbiota on brain homeostasis, behaviour and host metabolism, informing the development of microbiota-targeted therapies for stress-related disorders. ABSTRACT There is a growing recognition of the involvement of the gastrointestinal microbiota in the regulation of physiology and behaviour. Microbiota-derived metabolites play a central role in the communication between microbes and their host, with short-chain fatty acids (SCFAs) being perhaps the most studied. SCFAs are primarily derived from fermentation of dietary fibres and play a pivotal role in host gut, metabolic and immune function. All these factors have previously been demonstrated to be adversely affected by stress. Therefore, we sought to assess whether SCFA supplementation could counteract the enduring effects of chronic psychosocial stress. C57BL/6J male mice received oral supplementation of a mixture of the three principle SCFAs (acetate, propionate and butyrate). One week later, mice underwent 3 weeks of repeated psychosocial stress, followed by a comprehensive behavioural analysis. Finally, plasma corticosterone, faecal SCFAs and caecal microbiota composition were assessed. SCFA treatment alleviated psychosocial stress-induced alterations in reward-seeking behaviour, and increased responsiveness to an acute stressor and in vivo intestinal permeability. In addition, SCFAs exhibited behavioural test-specific antidepressant and anxiolytic effects, which were not present when mice had also undergone psychosocial stress. Stress-induced increases in body weight gain, faecal SCFAs and the colonic gene expression of the SCFA receptors free fatty acid receptors 2 and 3 remained unaffected by SCFA supplementation. Moreover, there were no collateral effects on caecal microbiota composition. Taken together, these data show that SCFA supplementation alleviates selective and enduring alterations induced by repeated psychosocial stress and these data may inform future research into microbiota-targeted therapies for stress-related disorders.
Collapse
Affiliation(s)
- Marcel van de Wouw
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Marcus Boehme
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Joshua M Lyte
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Niamh Wiley
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Conall Strain
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Orla O'Sullivan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
| |
Collapse
|
241
|
He L, Prodhan MAI, Yuan F, Yin X, Lorkiewicz PK, Wei X, Feng W, McClain C, Zhang X. Simultaneous quantification of straight-chain and branched-chain short chain fatty acids by gas chromatography mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1092:359-367. [PMID: 29936372 PMCID: PMC6190712 DOI: 10.1016/j.jchromb.2018.06.028] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 06/12/2018] [Accepted: 06/13/2018] [Indexed: 12/29/2022]
Abstract
Biomedical research in areas such as metabolic disorders, neuromodulatory, and immunomodulatory conditions involves lipid metabolism and demands a reliable and inexpensive method for quantification of short chain fatty acids (SCFAs). We report a GC-MS method for analysis of all straight-chain and branched-chain SCFAs using pentafluorobenzyl bromide (PFBBr) as derivatization reagent. We optimized the derivatization and GC-MS conditions using a mixture containing all eight SCFA standards, i.e., five straight-chain and three branched-chain SCFAs. The optimal derivatization conditions were derivatization time 90 min, temperature 60 °C, pH 7, and (CH3)2CO:H2O ratio 2:1 (v:v). Comparing the performance of different GC column configurations, a 30 m DB-225ms hyphenated with a 30 m DB-5ms column in tandem showed the best separation of SCFAs. Using the optimized experiment conditions, we simultaneously detected all SCFAs with much improved detection limit, 0.244-0.977 μM. We further applied the developed method to measure the SCFAs in mouse feces and all SCFAs were successfully quantified. The recovery rates of the eight SCFAs ranged from 55.7% to 97.9%.
Collapse
Affiliation(s)
- Liqing He
- Department of Chemistry, University of Louisville, Louisville, KY 40208, USA; University of Louisville Alcohol Research Center, University of Louisville, Louisville, KY 40208, USA; University of Louisville Hepatobiology & Toxicology Program, University of Louisville, Louisville, KY 40208, USA; Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY 40208, USA.
| | - Md Aminul Islam Prodhan
- Department of Chemistry, University of Louisville, Louisville, KY 40208, USA; University of Louisville Alcohol Research Center, University of Louisville, Louisville, KY 40208, USA; University of Louisville Hepatobiology & Toxicology Program, University of Louisville, Louisville, KY 40208, USA; Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY 40208, USA
| | - Fang Yuan
- Department of Chemistry, University of Louisville, Louisville, KY 40208, USA; University of Louisville Alcohol Research Center, University of Louisville, Louisville, KY 40208, USA; University of Louisville Hepatobiology & Toxicology Program, University of Louisville, Louisville, KY 40208, USA; Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY 40208, USA
| | - Xinmin Yin
- Department of Chemistry, University of Louisville, Louisville, KY 40208, USA; University of Louisville Alcohol Research Center, University of Louisville, Louisville, KY 40208, USA; University of Louisville Hepatobiology & Toxicology Program, University of Louisville, Louisville, KY 40208, USA; Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY 40208, USA
| | - Pawel K Lorkiewicz
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY 40208, USA; Diabetes and Obesity Center, University of Louisville, Louisville, KY 40208, USA
| | - Xiaoli Wei
- Department of Chemistry, University of Louisville, Louisville, KY 40208, USA; University of Louisville Alcohol Research Center, University of Louisville, Louisville, KY 40208, USA; University of Louisville Hepatobiology & Toxicology Program, University of Louisville, Louisville, KY 40208, USA; Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY 40208, USA
| | - Wenke Feng
- University of Louisville Hepatobiology & Toxicology Program, University of Louisville, Louisville, KY 40208, USA; Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY 40208, USA; Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40208, USA
| | - Craig McClain
- University of Louisville Hepatobiology & Toxicology Program, University of Louisville, Louisville, KY 40208, USA; Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY 40208, USA; Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40208, USA; Department of Medicine, University of Louisville, Louisville, KY 40208, USA; Robley Rex Louisville VAMC, Louisville, KY 40292, USA
| | - Xiang Zhang
- Department of Chemistry, University of Louisville, Louisville, KY 40208, USA; University of Louisville Alcohol Research Center, University of Louisville, Louisville, KY 40208, USA; University of Louisville Hepatobiology & Toxicology Program, University of Louisville, Louisville, KY 40208, USA; Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY 40208, USA; Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40208, USA
| |
Collapse
|
242
|
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: 32] [Impact Index Per Article: 4.6] [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
|
243
|
Fu YP, Feng B, Zhu ZK, Feng X, Chen SF, Li LX, Yin ZQ, Huang C, Chen XF, Zhang BZ, Jia RY, Song X, Lv C, Yue GZ, Ye G, Liang XX, He CL, Yin LZ, Zou YF. The Polysaccharides from Codonopsis pilosula Modulates the Immunity and Intestinal Microbiota of Cyclophosphamide-Treated Immunosuppressed Mice. Molecules 2018; 23:1801. [PMID: 30037030 PMCID: PMC6100181 DOI: 10.3390/molecules23071801] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/11/2018] [Accepted: 07/19/2018] [Indexed: 01/28/2023] Open
Abstract
Based on previous studies about microflora regulation and immunity enhancement activities of polysaccharides from Codonopsis pilosula Nannf. var. modesta (Nannf.) L. T. Shen (CPP), there is little study on intestinal mucosal immunity, which is a possible medium for contacting microflora and immunity. In the present study, the BALB/c mice were divided into five groups (eight mice in each group), including a normal group (Con), a model control group (Model), and model groups that were administered CPP (50, 100, 200 mg/kg/d) orally each day for seven days after intraperitoneal injection of 60 mg/kg BW/d cyclophosphamide (CP) for three days. CPP recovered the spleen index and restored the levels of IFN-γ, IL-2, IL-10, as well as serum IgG. In addition, it elevated ileum secretory immunoglobulin A (sIgA), the number of Lactobacillus and acetic acid content in cecum. These results indicated that CPP plays an important role in the protection against immunosuppression, especially mucosa immune damage, and the inhibition of pathogenic bacteria colonization, which could be considered a potential natural source of immunoregulator.
Collapse
Affiliation(s)
- Yu-Ping Fu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China.
| | - Zhong-Kai Zhu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Xin Feng
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Shu-Fan Chen
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Li-Xia Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Zhong-Qiong Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Chao Huang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Xing-Fu Chen
- Key Laboratory of Crop Ecophysiology and Farming System in Southwest China, Ministry of Agriculture, College of Agronomy, Sichuan Agricultural University, Chengdu 611130, China.
| | - Bing-Zhao Zhang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, China.
| | - Ren-Yong Jia
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Xu Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Cheng Lv
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Gui-Zhou Yue
- Department of Applied Chemistry, College of Science, Sichuan Agricultural University, Chengdu 611130, China.
| | - Gang Ye
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Xiao-Xia Liang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Chang-Liang He
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Li-Zi Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Yuan-Feng Zou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
244
|
Yacoubi N, Saulnier L, Bonnin E, Devillard E, Eeckhaut V, Rhayat L, Ducatelle R, Van Immerseel F. Short-chain arabinoxylans prepared from enzymatically treated wheat grain exert prebiotic effects during the broiler starter period. Poult Sci 2018; 97:412-424. [PMID: 29140465 DOI: 10.3382/ps/pex297] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Indexed: 11/20/2022] Open
Abstract
Carbohydrate-degrading multi-enzyme preparations (MEP) are used to improve broiler performances. Their mode of action is complex and not fully understood. In this study, we compared the effect of water-soluble fractions isolated at the pilot scale from wheat grain incubated with (WE) and without (WC) MEP. The fractions were incorporated in a wheat-based diet (0.1% w/w) to feed Ross PM3 broilers and compared with a non-supplemented control group (NC). The body weight gain (BWG), feed intake (FI), and feed conversion ratio (FCR) until d 14 were determined. At d 14, ileal and cecal contents and tissue samples were collected from euthanized animals. The intestinal contents were used to measure the short-chain fatty acids (SCFA) concentration using gas chromatography and to determine the abundance and composition of microbiota using 16S sequencing. Villi length of ileal samples was measured, while L-cell and T-cell densities were determined using immuno-histochemistry. The MEP treatment increased the amount of water-soluble arabinoxylans (AX) and reduced their molecular weight while retaining their polymer behavior. The WE fraction significantly (P < 0.05) increased FI by 13.8% and BWG by 14.7% during the first wk post hatch when compared to NC. No significant effect on FCR was recorded during the trial. The WE increased the abundance of Enterococcus durans and Candidatus arthromitus in the ileum and of bacteria within the Lachnospiraceae and Ruminococcaceae families, containing abundant butyrate-producing bacteria, in the ceca. It also increased the concentration of SCFA in the ceca, decreased the T-lymphocyte infiltration in the intestinal mucosa, and increased the glucagon-like-peptide-2 (GLP-2)-producing L-cell density in the ileal epithelium compared with WC and NC. No significant effects were observed on villi length. These results showed that AX present in the WE fraction altered the microbiota composition towards butyrate producers in the ceca. Butyrate may be responsible for the reduction of inflammation, as suggested by the decrease in T-lymphocyte infiltration, which may explain the higher feed intake leading to improved animal growth.
Collapse
Affiliation(s)
- N Yacoubi
- INRA, UR1268 Biopolymers Interactions Assemblies, BP 71627, F-44316 Nantes, France.,Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium.,Adisseo France SAS, Center of Expertise and Research in Nutrition, F-03600 Commentry, France
| | - L Saulnier
- INRA, UR1268 Biopolymers Interactions Assemblies, BP 71627, F-44316 Nantes, France
| | - E Bonnin
- INRA, UR1268 Biopolymers Interactions Assemblies, BP 71627, F-44316 Nantes, France
| | - E Devillard
- Adisseo France SAS, Center of Expertise and Research in Nutrition, F-03600 Commentry, France
| | - V Eeckhaut
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - L Rhayat
- Adisseo France SAS, Center of Expertise and Research in Nutrition, F-03600 Commentry, France
| | - R Ducatelle
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | - F Van Immerseel
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| |
Collapse
|
245
|
|
246
|
Han F, Wang Y, Han Y, Zhao J, Han F, Song G, Jiang P, Miao H. Effects of Whole-Grain Rice and Wheat on Composition of Gut Microbiota and Short-Chain Fatty Acids in Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:6326-6335. [PMID: 29766722 DOI: 10.1021/acs.jafc.8b01891] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Diets rich in whole grain (WG) cereals bring lower disease risks compared with refined grain-based diets. We investigated the effects of polished rice (PR), refined wheat (RW), unpolished rice (UPR), and whole wheat (WW) on short-chain fatty acids (SCFAs) and gut microbiota in ileal, cecal, and colonic digesta of normal rats. Animals fed with UPR and WW diets exhibited higher total SCFA in cecal and colonic digesta compared with those fed with PR and RW diets. Wheat diets contributed higher total SCFA than rice diets. In cecal and colonic digesta, animals fed with UPR and WW diets demonstrated higher acetate and butyrate contents than those given PR and RW. Firmicutes were the dominant eumycota in rat ileum digesta (>92% abundance). Cecal and colonic digesta were dominated by Firmicutes, Verrucomicrobia, and Bacteroidetes. UPR and WW affected gut microbiota, decreasing the proportion of Firmicutes to Bacteroidetes. SMB53, Lactobacillus, and Faecalibacterium were the main bacterial genera in ileal digesta. Akkermansia was highest in cecal and colonic digesta. In the colonic digesta of rats, the relative abundance of Akkermansia in rats on wheat diets was higher than that in rats on rice diets ( P < 0.05). Thus, UPR and WW could modulate gut microbiota composition and increase the SCFA concentration. Wheat diet was superior to rice diet in terms of intestinal microbiota adjustment.
Collapse
Affiliation(s)
- Fei Han
- Academy of State Administration of Grain , Beijing 100037 , People's Republic of China
| | - Yong Wang
- Academy of State Administration of Grain , Beijing 100037 , People's Republic of China
| | - Yangyang Han
- Academy of State Administration of Grain , Beijing 100037 , People's Republic of China
- School of Food Science & Technology , Jiangnan University , Wuxi , Jiangsu 214122 , People's Republic of China
| | - Jianxin Zhao
- School of Food Science & Technology , Jiangnan University , Wuxi , Jiangsu 214122 , People's Republic of China
| | - Fenli Han
- Academy of State Administration of Grain , Beijing 100037 , People's Republic of China
- School of Food Science & Technology , Jiangnan University , Wuxi , Jiangsu 214122 , People's Republic of China
| | - Ge Song
- Academy of State Administration of Grain , Beijing 100037 , People's Republic of China
| | - Ping Jiang
- Academy of State Administration of Grain , Beijing 100037 , People's Republic of China
| | - Haijiang Miao
- Academy of State Administration of Grain , Beijing 100037 , People's Republic of China
| |
Collapse
|
247
|
Intake of Polydextrose Alters Hematology and the Profile of Short Chain Fatty Acids in Partially Gastrectomized Rats. Nutrients 2018; 10:nu10060792. [PMID: 29925762 PMCID: PMC6024616 DOI: 10.3390/nu10060792] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/05/2018] [Accepted: 06/06/2018] [Indexed: 02/06/2023] Open
Abstract
Polydextrose (PDX) ingestion may increase the intestinal absorption of iron. This study evaluated the effects of 7.5% polydextrose supplementation on markers of iron uptake, transport and storage in partially gastrectomized rats. Half of a batch of 40 male Wistar rats (250 g) underwent Billroth II partial gastrectomy with anterior truncal vagotomy (GXT), while the other half underwent sham gastrectomy (SHAM). At 7 postoperative days, the animals were subdivided into four groups (n = 10): Sham Control and GXT Control (no polydextrose); Sham PDX and GXT PDX (with 7.5% PDX). The animals were euthanized after 60 day of PDX treatment. Organ weight, cecal pH, the characterization and quantification of short-chain fatty acids (SCFA), hematological parameters, hepatic iron content and the expression of ferroportin (FPT) in the jejunum, cecum, colon and liver were evaluated. PDX caused changes in the cecum of the supplemented animals, where there was a decrease in pH, increase in cecal wall and marked production of SCFA, especially acetic and propionic acids (p < 0.05). Hepatic iron levels were lower in GXT animals. PDX increased hemoglobin (HGB) values by 29.2% and hematocrit (HCT) by 55.8% in the GXT PDX group compared to the GXT Control group. The GXT PDX group had lower hepatic FPT expression (p < 0.05). PDX led to increased SCFA concentration in the supplemented animals. Considering that SCFAs play a central role in the increasing nutrients uptake, this mechanism may be involved in altering the hematology profile observed in these animals but not enough to reverse iron deficiency anemia in post-gastrectomy rats.
Collapse
|
248
|
Marungruang N, Arévalo Sureda E, Lefrançoise A, Weström B, Nyman M, Prykhodko O, Fåk Hållenius F. Impact of dietary induced precocious gut maturation on cecal microbiota and its relation to the blood-brain barrier during the postnatal period in rats. Neurogastroenterol Motil 2018; 30:e13285. [PMID: 29327435 DOI: 10.1111/nmo.13285] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 12/06/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Precocious maturation of the gastrointestinal barrier (GIB) in newborn mammals can be induced by dietary provocation, but how this affects the gut microbiota and the gut-brain axis remains unknown. The objective of this study was to investigate effects of induced GIB maturation on gut microbiota composition and blood-brain barrier (BBB) permeability. METHODS Suckling rats were studied at 72 h after gavage with phytohemagglutinin (PHA) or microbial protease (PT) to induce maturation of GIB. For comparison, untreated suckling and weaned rats were included (n = 10). Human serum albumin (HSA) was administered orally and analyzed in blood to assess permeability of the GIB, while intraperitoneally injected bovine serum albumin (BSA) was measured in the brain tissue for BBB permeability. The cecal microbial composition, plasma lipopolysaccharide-binding protein (LBP) levels and short-chain fatty acids in serum and brain were analyzed. KEY RESULTS Cessation of HSA passage to blood after PHA or PT treatment was similar to that seen in weaned rats. Interestingly, concomitant increases in cecal Bacteroidetes and plasma LBP levels were observed after both PHA and PT treatments. The BBB passage of BSA was surprisingly elevated after weaning, coinciding with lower plasma LBP levels and specific microbial taxa and increased acetate uptake into the brain. CONCLUSIONS & INFERENCES This study provides evidence that the gut microbiota alteration following induced precocious GIB maturation may induce low-grade systemic inflammation and alter SCFAs utilization in the brain which may also play a potential role in GIB-BBB dysfunction disorders in neonates.
Collapse
Affiliation(s)
- N Marungruang
- Food for Health Science Center, Lund University Kemicentrum, Lund, Sweden
| | | | - A Lefrançoise
- Food for Health Science Center, Lund University Kemicentrum, Lund, Sweden
| | - B Weström
- Department of Biology, Lund University, Lund, Sweden
| | - M Nyman
- Food for Health Science Center, Lund University Kemicentrum, Lund, Sweden
| | - O Prykhodko
- Food for Health Science Center, Lund University Kemicentrum, Lund, Sweden
| | - F Fåk Hållenius
- Food for Health Science Center, Lund University Kemicentrum, Lund, Sweden
| |
Collapse
|
249
|
Description of Mediterraneibacter massiliensis, gen. nov., sp. nov., a new genus isolated from the gut microbiota of an obese patient and reclassification of Ruminococcus faecis, Ruminococcus lactaris, Ruminococcus torques, Ruminococcus gnavus and Clostridium glycyrrhizinilyticum as Mediterraneibacter faecis comb. nov., Mediterraneibacter lactaris comb. nov., Mediterraneibacter torques comb. nov., Mediterraneibacter gnavus comb. nov. and Mediterraneibacter glycyrrhizinilyticus comb. nov. Antonie van Leeuwenhoek 2018; 111:2107-2128. [PMID: 29855844 DOI: 10.1007/s10482-018-1104-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/20/2018] [Indexed: 12/20/2022]
Abstract
An anaerobic isolate, strain AT7T, was cultivated from a stool sample of a morbidly obese French woman using a microbial culturomics approach. The 16S rRNA gene sequence analysis showed that strain AT7T exhibited 96% nucleotide sequence similarity with Ruminococcus torques strain JCM 6553T (= ATCC 27756T = VPI B2-51T), currently the closest related species with a validly published name. The strain was observed to be a Gram-stain positive, non-motile, asporogenous and coccobacillary-shaped bacterium. It was found to be catalase positive and oxidase negative. Its major fatty acids were identified as C16:0 (54%) and C18:1n9 (30%). The draft genome of strain AT7T is 3,069,882 bp long with 42.4% G+C content. 2925 genes were predicted, including 2867 protein-coding genes and 58 RNAs. Based on phenotypic, biochemical, phylogenetic and genomic evidence, we propose the creation of the new genus Mediterraneibacter and species, Mediterraneibacter massiliensis, that contains strain AT7T (= CSUR P2086T = DSM 100837T), and the reclassification of Ruminococcus faecis, Ruminococcus lactaris, Ruminococcus torques, Ruminococcus gnavus, Clostridium glycyrrhizinilyticum as Mediterraneibacter faecis comb. nov., with type strain Eg2T (= KCTC 5757T = JCM15917T), Mediterraneibacter lactaris comb. nov., with type strain ATCC 29176T (= VPI X6-29T), Mediterraneibacter torques comb. nov., with type strain ATCC 27756T (= VPI B2-51T), Mediterraneibacter gnavus comb. nov., with type strain ATCC 29149T (= VPI C7-9T) and Mediterraneibacter glycyrrhizinilyticus comb. nov., with type strain ZM35T (= JCM 13368T = DSM 17593T), respectively.
Collapse
|
250
|
Maltz RM, Keirsey J, Kim SC, Mackos AR, Gharaibeh RZ, Moore CC, Xu J, Bakthavatchalu V, Somogyi A, Bailey MT. Prolonged restraint stressor exposure in outbred CD-1 mice impacts microbiota, colonic inflammation, and short chain fatty acids. PLoS One 2018; 13:e0196961. [PMID: 29742146 PMCID: PMC5942810 DOI: 10.1371/journal.pone.0196961] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 04/24/2018] [Indexed: 12/13/2022] Open
Abstract
Stressor-exposure has been shown to exacerbate inflammation and change the composition of the gastrointestinal microbiota; however stressor-induced effects on microbiota-derived metabolites and their receptors are unknown. Thus, bacterial-produced short chain fatty acids (SCFAs), as well as microbial community composition, were assessed in the colons of mice exposed to stress during infection with Citrobacter rodentium. Mice were exposed to overnight restraint on 7 consecutive nights, or left undisturbed as a control. After the first exposure of restraint, mice were orally challenged with C. rodentium or with vehicle. Microbial community composition was assessed using 16S rRNA gene sequencing and SCFA levels measured using gas chromatography-mass spectrometry (GC-MS). Pathogen levels and colonic inflammation were also assessed 6 days post-infection. Results demonstrated that the microbial community structure and SCFA production were significantly affected by both stressor exposure and C. rodentium-infection. Exposure to prolonged restraint in the absence of infection significantly reduced SCFAs (acetic acid, butyric acid, and propionic acid). Multiple bacterial taxa were affected by stressor exposure, with the relative abundance of Lactobacillus being significantly reduced and directly correlated with propionic acid. Lactobacillus abundances were inversely correlated with colonic inflammation, supporting the contention that Lactobacillus helps to regulate mucosal inflammatory responses. Our data indicates that restraint stressor can have significant effects on pathogen-induced colonic inflammation and suggest that stressor-induced changes in the microbiota, microbial-produced SCFAs and their receptors may be involved.
Collapse
Affiliation(s)
- Ross M. Maltz
- Pediatric Gastroenterology, Nationwide Children's Hospital, Columbus, OH, United States of America
- Center for Microbial Pathogenesis, The Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States of America
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Jeremy Keirsey
- Campus Chemical Instrumentation Center Mass Spec and Proteomics, The Ohio State University, Columbus, OH, United States of America
| | - Sandra C. Kim
- Pediatric Gastroenterology, Nationwide Children's Hospital, Columbus, OH, United States of America
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Amy R. Mackos
- Center for Microbial Pathogenesis, The Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States of America
| | - Raad Z. Gharaibeh
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, United States of America
- Bioinformatics Services Division, Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Kannapolis, NC, United States of America
| | - Cathy C. Moore
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, United States of America
| | - Jinyu Xu
- Center for Microbial Pathogenesis, The Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States of America
| | - Vasudevan Bakthavatchalu
- The Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Arpad Somogyi
- Campus Chemical Instrumentation Center Mass Spec and Proteomics, The Ohio State University, Columbus, OH, United States of America
| | - Michael T. Bailey
- Center for Microbial Pathogenesis, The Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States of America
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States of America
| |
Collapse
|