551
|
Wu R, Mei X, Wang J, Sun W, Xue T, Lin C, Xu D. Zn(ii)-Curcumin supplementation alleviates gut dysbiosis and zinc dyshomeostasis during doxorubicin-induced cardiotoxicity in rats. Food Funct 2019; 10:5587-5604. [PMID: 31432062 DOI: 10.1039/c9fo01034c] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Doxorubicin is a powerful anticancer agent used to treat a variety of human neoplasms. However, the clinical use of doxorubicin is hampered by cardiotoxicity and effective cardioprotective adjuvants do not exist. Dietary zinc, an essential nutrient, is required to maintain steady-state tissue zinc levels and intestinal homeostasis and may yield therapeutic benefits in diseases associated with zinc dysregulation or gut dysbiosis. Here, we investigated the effects of dietary Zn(ii)-curcumin (ZnCM) solid dispersions on gut dysbiosis and zinc dyshomeostasis during doxorubicin-induced cardiotoxicity in rats. Rats were injected with multiple low doses of doxorubicin and orally administered ZnCM daily over four weeks. Daily administration of ZnCM not only alleviated Dox-induced gut dysbiosis-as indicated by the increased Firmicutes-to-Bacteroidetes ratio and the maintenance of the relative abundances of major beneficial bacteria including Clostridium_XIVa, Clostridium_IV, Roseburia, Butyricicoccus and Akkermansia-but also maintained intestinal barrier integrity and decreased the lipopolysaccharide (LPS) contents of feces and plasma. ZnCM also significantly attenuated doxorubicin-induced zinc dyshomeostasis, which was mirrored by preservation of zinc levels and expression of zinc-related transporters. Furthermore, ZnCM significantly improved heart function and reduced cardiomyocyte apoptosis and myocardial injury in doxorubicin-treated rats. Notably, the regulation of zinc homeostasis and cardioprotective and microbiota-modulating effects of ZnCM were transmissible through horizontal feces transfer from ZnCM-treated rats to normal rats. Thus, ZnCM supplementation has potential as an effective therapeutic strategy to alleviate gut dysbiosis and zinc dyshomeostasis during doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Rihui Wu
- Laboratory of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, 510275 Guangzhou, China.
| | - Xueting Mei
- Laboratory Animal Center of Sun Yat-sen University, Sun Yat-sen University, 510275 Guangzhou, China
| | - Jiasheng Wang
- Laboratory of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, 510275 Guangzhou, China.
| | - Wenjia Sun
- Laboratory of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, 510275 Guangzhou, China.
| | - Ting Xue
- Laboratory of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, 510275 Guangzhou, China.
| | - Caixia Lin
- Laboratory of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, 510275 Guangzhou, China.
| | - Donghui Xu
- Laboratory of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, 510275 Guangzhou, China.
| |
Collapse
|
552
|
Jin M, Kalainy S, Baskota N, Chiang D, Deehan EC, McDougall C, Tandon P, Martínez I, Cervera C, Walter J, Abraldes JG. Faecal microbiota from patients with cirrhosis has a low capacity to ferment non-digestible carbohydrates into short-chain fatty acids. Liver Int 2019; 39:1437-1447. [PMID: 30919578 DOI: 10.1111/liv.14106] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/18/2019] [Accepted: 03/20/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Cirrhosis is associated with dysbiosis, but its functional consequences are still largely unknown. Short-chain fatty acids (SCFAs) account for physiological interactions between the gut microbiota and host. Our aim was to assess the impact of cirrhotic dysbiosis on the production of SCFAs. METHODS Seventeen patients with cirrhosis and 17 controls were selected. Microbiota composition in faecal samples was assessed by next-generation 16S rRNA gene sequencing. SCFAs were measured with GC-MS in faecal samples and after in vitro batch fermentations using arabinoxylan, resistant starch, pectin, and lactulose as substrates. RESULTS Among the 17 cirrhotic patients (mean age 58, eight males), six, nine and two were, respectively, Child-Pugh class A, B and C. Eleven patients were on oral antibiotics, 11 on lactulose and 13 on proton pump inhibitors. Cirrhotic patients showed marked differences in the composition and diversity of gut microbiome when compared to controls, that were more pronounced with increased severity. Stool samples from cirrhotic patients showed lower SCFAs content and reduced capacity to produce SCFAs in batch fermentations, with butyrate production being the most abnormal. These functional aberrancies were more pronounced with greater liver disease severity. Abundance of Ruminococcus faecis (in family Ruminococcaceae), Faecalicatena fissicatena and Fusicatenibacter saccharivorans (in family Lachnospiraceae) was positively correlated with the SCFAs production. CONCLUSION Cirrhotic dysbiosis is associated with a decreased capacity to ferment non-digestible carbohydrates into SCFAs, especially into butyrate. These functional abnormalities are more pronounced as disease progresses. These results might inform the design of gut-targeted therapies for cirrhosis.
Collapse
Affiliation(s)
- Mingliang Jin
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada.,College of Animal Sciences, Zhejiang University, Hangzhou, PR China.,School of Life Sciences, Northwestern Polytechnical University, Xi'an, PR China
| | - Sylvia Kalainy
- Division of Gastroenterology (Liver Unit), University of Alberta, Edmonton, Alberta, Canada
| | - Nami Baskota
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Diana Chiang
- Division of Infectious Diseases, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Edward C Deehan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Chelsea McDougall
- Division of Gastroenterology (Liver Unit), University of Alberta, Edmonton, Alberta, Canada
| | - Puneeta Tandon
- Division of Gastroenterology (Liver Unit), University of Alberta, Edmonton, Alberta, Canada
| | - Inés Martínez
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Carlos Cervera
- Division of Infectious Diseases, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Jens Walter
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada.,Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Juan G Abraldes
- Division of Gastroenterology (Liver Unit), University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
553
|
Parker A, Fonseca S, Carding SR. Gut microbes and metabolites as modulators of blood-brain barrier integrity and brain health. Gut Microbes 2019; 11:135-157. [PMID: 31368397 PMCID: PMC7053956 DOI: 10.1080/19490976.2019.1638722] [Citation(s) in RCA: 336] [Impact Index Per Article: 67.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/22/2019] [Accepted: 06/26/2019] [Indexed: 02/03/2023] Open
Abstract
The human gastrointestinal (gut) microbiota comprises diverse and dynamic populations of bacteria, archaea, viruses, fungi, and protozoa, coexisting in a mutualistic relationship with the host. When intestinal homeostasis is perturbed, the function of the gastrointestinal tract and other organ systems, including the brain, can be compromised. The gut microbiota is proposed to contribute to blood-brain barrier disruption and the pathogenesis of neurodegenerative diseases. While progress is being made, a better understanding of interactions between gut microbes and host cells, and the impact these have on signaling from gut to brain is now required. In this review, we summarise current evidence of the impact gut microbes and their metabolites have on blood-brain barrier integrity and brain function, and the communication networks between the gastrointestinal tract and brain, which they may modulate. We also discuss the potential of microbiota modulation strategies as therapeutic tools for promoting and restoring brain health.
Collapse
Affiliation(s)
- Aimée Parker
- Gut Microbes and Health Research Programme, Quadram Institute Bioscience, Norwich, UK
| | - Sonia Fonseca
- Gut Microbes and Health Research Programme, Quadram Institute Bioscience, Norwich, UK
| | - Simon R. Carding
- Gut Microbes and Health Research Programme, Quadram Institute Bioscience, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| |
Collapse
|
554
|
Bodkhe R, Balakrishnan B, Taneja V. The role of microbiome in rheumatoid arthritis treatment. Ther Adv Musculoskelet Dis 2019; 11:1759720X19844632. [PMID: 31431810 PMCID: PMC6685117 DOI: 10.1177/1759720x19844632] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 03/25/2019] [Indexed: 12/15/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder with multifactorial etiology; both genetic and environmental factors are known to be involved in pathogenesis. Treatment with disease-modifying antirheumatic drugs (DMARDs) plays an essential role in controlling disease progression and symptoms. DMARDs have immunomodulatory properties and suppress immune response by interfering in various pro-inflammatory pathways. Recent evidence has shown that the gut microbiota directly and indirectly modulates the host immune system. RA has been associated with dysbiosis of the gut microbiota. Patients with RA treated with DMARDs show partial restoration of eubiotic gut microbiome. Hence, it is essential to understand the impact of DMARDs on the microbial composition and its consequent influences on the host immune system to identify novel therapies for RA. In this review, we discuss the importance of antirheumatic-drug-induced host microbiota modulations and possible probiotics that can generate eubiosis.
Collapse
Affiliation(s)
- Rahul Bodkhe
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | | | - Veena Taneja
- Department of Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
555
|
Blood Pressure Abnormalities Associated with Gut Microbiota-Derived Short Chain Fatty Acids in Children with Congenital Anomalies of the Kidney and Urinary Tract. J Clin Med 2019; 8:jcm8081090. [PMID: 31344888 PMCID: PMC6722976 DOI: 10.3390/jcm8081090] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/14/2019] [Accepted: 07/19/2019] [Indexed: 12/12/2022] Open
Abstract
Both kidney disease and hypertension can originate from early life. Congenital anomalies of the kidney and urinary tract (CAKUT) are the leading cause of chronic kidney disease (CKD) in children. Since gut microbiota and their metabolite short chain fatty acids (SCFAs) have been linked to CKD and hypertension, we examined whether gut microbial composition and SCFAs are correlated with blood pressure (BP) load and renal outcome in CKD children with CAKUT. We enrolled 78 children with CKD stage G1-G4. Up to 65% of children with CAKUT had BP abnormalities on 24 h ambulatory blood pressure monitoring (ABPM). CKD children with CAKUT had lower risk of developing BP abnormalities and CKD progression than those with non-CAKUT. Reduced plasma level of propionate was found in children with CAKUT, which was related to increased abundance of phylum Verrucomicrobia, genus Akkermansia, and species Bifidobacterium bifidum. CKD children with abnormal ABPM profile had higher plasma levels of propionate and butyrate. Our findings highlight that gut microbiota-derived SCFAs like propionate and butyrate are related to BP abnormalities in children with an early stage of CKD. Early assessments of these microbial markers may aid in developing potential targets for early life intervention for lifelong hypertension prevention in childhood CKD.
Collapse
|
556
|
Singh RP. Glycan utilisation system in Bacteroides and Bifidobacteria and their roles in gut stability and health. Appl Microbiol Biotechnol 2019; 103:7287-7315. [PMID: 31332487 DOI: 10.1007/s00253-019-10012-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 02/07/2023]
Abstract
Gut residential hundred trillion microbial cells are indispensable for maintaining gut homeostasis and impact on host physiology, development and immune systems. Many of them have displayed excellence in utilising dietary- and host-derived complex glycans and are producing useful postbiotics including short-chain fatty acids to primarily fuel different organs of the host. Therefore, employing individual microbiota is nowadays becoming a propitious target in biomedical for improving gut dysbiosis conditions of the host. Among other gut microbial communities, Bacteroides and Bifidobacteria are coevolved to utilise diverse ranges of diet- and host-derived glycans through harmonising distinct glycan utilisation systems. These gut symbionts frequently share digested oligosaccharides, carbohydrate-active enzymes and fermentable intermediate molecules for sustaining gut microbial symbiosis and improving fitness of own or other communities. Genomics approaches have provided unprecedented insights into these functions, but their precise mechanisms of action have poorly known. Sympathetic glycan-utilising strategy of each gut commensal will provide overview of mechanistic dynamic nature of the gut environment and will then assist in applying aptly personalised nutritional therapy. Thus, the review critically summarises cutting edge understanding of major plant- and host-derived glycan-utilising systems of Bacteroides and Bifidobacteria. Their evolutionary adaptation to gut environment and roles of postbiotics in human health are also highlighted.
Collapse
Affiliation(s)
- Ravindra Pal Singh
- Food and Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute (NABI), SAS, Nagar, Punjab, 140306, India.
| |
Collapse
|
557
|
Abstract
The gut microbiome is a complex microbial community that plays a key role in human health. Diet is an important factor dictating gut microbiome composition. This is mediated by multiple microbe-microbe interactions that result in the fermentation of nondigestible carbohydrates and the production of short-chain fatty acids. Certain species play key metabolic roles in the microbiome, and their disappearance could result in dysbiosis. In this work, a synthetic consortium of 14 gut microbes was studied during the utilization of prebiotic inulin in batch bioreactors. Fermentations were repeated leaving one species out every time, in order to evaluate the impact of their elimination on the system. Substrate consumption, microbial composition, and metabolite production were determined. Single deletions never resulted in a complete loss of bacterial growth or inulin consumption, suggesting functional redundancy. Deletions of Bacteroides dorei and Lachnoclostridium clostridioforme resulted in lower biomass and higher residual inulin. The absence of B. dorei impacted the abundance of the other 10 species negatively. Lachnoclostridium symbiosum, a butyrate producer, appeared to be the most sensitive species to deletions, being stimulated by the presence of Escherichia coli, Bifidobacterium adolescentis, B. dorei, and Lactobacillus plantarum Conversely, bioreactors without these species did not show butyrate production. L. clostridioforme was observed to be essential for propionate production, and B. dorei for lactate production. Our analysis identified specific members that were essential for the function of the consortium. In conclusion, species deletions from microbial consortia could be a useful approach to identify relevant interactions between microorganisms and defining metabolic roles in the gut microbiome.IMPORTANCE Gut microbes associate, compete for, and specialize in specific metabolic tasks. These interactions are dictated by the cross-feeding of degradation or fermentation products. However, the individual contribution of microbes to the function of the gut microbiome is difficult to evaluate. It is essential to understand the complexity of microbial interactions and how the presence or absence of specific microorganisms affects the stability and functioning of the gut microbiome. The experimental approach of this study could be used for identifying keystone species, in addition to redundant functions and conditions that contribute to community stability. Redundancy is an important feature of the microbiome, and its reduction could be useful for the design of microbial consortia with desired metabolic properties enhancing the tasks of the keystone species.
Collapse
|
558
|
Zaiss MM, Jones RM, Schett G, Pacifici R. The gut-bone axis: how bacterial metabolites bridge the distance. J Clin Invest 2019; 129:3018-3028. [PMID: 31305265 DOI: 10.1172/jci128521] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The gut microbiome is a key regulator of bone health that affects postnatal skeletal development and skeletal involution. Alterations in microbiota composition and host responses to the microbiota contribute to pathological bone loss, while changes in microbiota composition that prevent, or reverse, bone loss may be achieved by nutritional supplements with prebiotics and probiotics. One mechanism whereby microbes influence organs of the body is through the production of metabolites that diffuse from the gut into the systemic circulation. Recently, short-chain fatty acids (SCFAs), which are generated by fermentation of complex carbohydrates, have emerged as key regulatory metabolites produced by the gut microbiota. This Review will focus on the effects of SCFAs on the musculoskeletal system and discuss the mechanisms whereby SCFAs regulate bone cells.
Collapse
Affiliation(s)
- Mario M Zaiss
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Georg Schett
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, Georgia, USA.,Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
559
|
Discovery of potential genes contributing to the biosynthesis of short-chain fatty acids and lactate in gut microbiota from systematic investigation in E. coli. NPJ Biofilms Microbiomes 2019; 5:19. [PMID: 31312512 PMCID: PMC6626047 DOI: 10.1038/s41522-019-0092-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 06/19/2019] [Indexed: 12/17/2022] Open
Abstract
Microbiota play important roles in the internal environment and health of humans, livestock and wild animals. Short-chain fatty acids (SCFAs) and lactate are primary metabolites that can impact the composition and function of human microbiota. According to the well-characterized key synthesis genes, many SCFA- and lactate-producing bacteria have been identified in the gut microbiota. However, unknown genes may also contribute to the formation of SCFAs and lactate. The identification of such genes will provide new engineering targets and new strategies for maintaining a stable structure of beneficial microbiota. In this study, we used Escherichia coli as a model to analyze possible genes related to SCFAs and lactate production besides the well-characterized ones. The functions of nineteen candidate genes were studied by targeted gene deletion and overexpression. Results indicated thioesterase genes such as yciA, tesA, tesB, and menI can contribute to acetate and/or butyrate formation. As for lactate, mgsA and lldD can function in addition to ldh gene. At the same time, the distribution of these functional genes in gut microbiota was investigated. Most bacteria contain the well-studied genes whereas some bacteria contain some of the described unusual ones. The results provide insights and genetic targets for the discovery of new SCFA- and lactate-producing bacteria in gut microbiota.
Collapse
|
560
|
Gut Prevotella as a possible biomarker of diet and its eubiotic versus dysbiotic roles: a comprehensive literature review. Br J Nutr 2019; 122:131-140. [PMID: 30924428 DOI: 10.1017/s0007114519000680] [Citation(s) in RCA: 192] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The gut microbiota has a profound impact on human health. Emerging data show that dietary patterns are associated with different communities of bacterial species within the gut. Prevotella species have been correlated with plant-rich diets, abundant in carbohydrates and fibres. Dysbiosis within the gut ecosystem has been associated with the development of non-communicable diseases such as obesity, the metabolic syndrome, inflammatory bowel disease, irritable bowel syndrome, colorectal cancer, type 1 diabetes, allergies and other diseases. The purpose of this comprehensive literature review was to evaluate the available data on the impact of diet on the Prevotella genus, as a dietary fibre fermenter in the gut as well as its implications as a potential biomarker for homeostasis or disease state through its metabolite signature. Studies were identified by conducting PubMed, Web of Science Core Collection and Google Scholar electronic searches. We found eighty-five publications reporting the impact of dietary patterns on gut microbial communities, including Prevotella or Prevotella/Bacteroides ratio in particular. Moreover, the role of Prevotella species on health status was also evaluated. Prevotella possess a high genetic diversity, representing one of the important groups found in the oral cavity and large intestine of man. The gut commensal Prevotella bacteria contribute to polysaccharide breakdown, being dominant colonisers of agrarian societies. However, studies also suggested a potential role of Prevotella species as intestinal pathobionts. Further metagenomic studies are needed in order to reveal health- or disease-modulating properties of Prevotella species in the gut.
Collapse
|
561
|
Herrera S, Martínez-Sanz J, Serrano-Villar S. HIV, Cancer, and the Microbiota: Common Pathways Influencing Different Diseases. Front Immunol 2019; 10:1466. [PMID: 31316514 PMCID: PMC6610485 DOI: 10.3389/fimmu.2019.01466] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022] Open
Abstract
HIV infection exerts profound and perhaps irreversible damage to the gut mucosal-associated lymphoid tissues, resulting in long-lasting changes in the signals required for the coordination of commensal colonization and in perturbations at the compositional and functional level of the gut microbiota. These abnormalities in gut microbial communities appear to affect clinical outcomes, including T-cell recovery, vaccine responses, HIV transmission, cardiovascular disease, and cancer pathogenesis. For example, the microbial signature associated with HIV infection has been shown to induce tryptophan catabolism, affect the butyrate synthesis pathway, impair anti-tumoral immunity and affect oxidative stress, which have also been linked to the pathogenesis of cancer. Furthermore, some of the taxa that are depleted in subjects with HIV have proved to modulate the anti-tumor efficacy of various chemotherapies and immunotherapeutic agents. The aim of this work is to provide a broad overview of recent advances in our knowledge of how HIV might affect the microbiota, with a focus on the pathways shared with cancer pathogenesis.
Collapse
Affiliation(s)
- Sabina Herrera
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, Universidad de Alcalá (IRYCIS), Madrid, Spain
| | - Javier Martínez-Sanz
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, Universidad de Alcalá (IRYCIS), Madrid, Spain
| | - Sergio Serrano-Villar
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, Universidad de Alcalá (IRYCIS), Madrid, Spain
| |
Collapse
|
562
|
Chai LJ, Lu ZM, Zhang XJ, Ma J, Xu PX, Qian W, Xiao C, Wang ST, Shen CH, Shi JS, Zheng-Hong X. Zooming in on Butyrate-Producing Clostridial Consortia in the Fermented Grains of Baijiu via Gene Sequence-Guided Microbial Isolation. Front Microbiol 2019; 10:1397. [PMID: 31316481 PMCID: PMC6611424 DOI: 10.3389/fmicb.2019.01397] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/04/2019] [Indexed: 11/13/2022] Open
Abstract
Butyrate, one of the key aroma compounds in Luzhou-flavor baijiu, is synthesized through two alternative pathways: butyrate kinase (buk) and butyryl-CoA: acetate CoA-transferase (but). A lack of knowledge of butyrate-producing microorganisms hinders our ability to understand the flavor formation mechanism of baijiu. Here, temporal dynamics of microbial metabolic profiling in fermented grains (FG) was explored via PICRUSt based on 16S rRNA gene sequences. We found Bacilli and Bacteroidia were the major potential butyrate producers in buk pathway at the beginning of fermentation, while later Clostridia dominated the two pathways. Clone library analysis also revealed that Clostridia (~73% OTUs) was predominant in buk pathway throughout fermentation, followed by Bacilli and Bacteroidia, and but pathway was merely possessed by Clostridia. Afterward, Clostridia-specific 16S rRNA gene sequencing demonstrated Clostridium might be the major butyrate-producing genus in two pathways, which was subsequently evaluated using culture approach. Seventeen Clostridium species were isolated from FG based on 16S rRNA gene sequence-guided medium prediction method. Profiles of short-chain fatty acids and but and buk genes in these species demonstrated phylogenetic and functional diversities of butyrate-producing Clostridium in FG. These findings add to illustrate the diversity of potential butyrate producers during brewing and provide a workflow for targeting functional microbes in complex microbial community.
Collapse
Affiliation(s)
- Li-Juan Chai
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China.,Jiangsu Engineering Research Center for Bioactive Products Processing Technology, Jiangnan University, Wuxi, China
| | - Zhen-Ming Lu
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China.,Jiangsu Engineering Research Center for Bioactive Products Processing Technology, Jiangnan University, Wuxi, China.,School of Pharmaceutical Science, Jiangnan University, Wuxi, China
| | - Xiao-Juan Zhang
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China.,Jiangsu Engineering Research Center for Bioactive Products Processing Technology, Jiangnan University, Wuxi, China.,National Engineering Research Center of Solid-State Brewing, Luzhou, China
| | - Jian Ma
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Peng-Xiang Xu
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Wei Qian
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Chen Xiao
- Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Song-Tao Wang
- National Engineering Research Center of Solid-State Brewing, Luzhou, China
| | - Cai-Hong Shen
- National Engineering Research Center of Solid-State Brewing, Luzhou, China
| | - Jin-Song Shi
- School of Pharmaceutical Science, Jiangnan University, Wuxi, China
| | - Xu Zheng-Hong
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China.,Key Laboratory of Industrial Biotechnology of Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China.,National Engineering Research Center of Solid-State Brewing, Luzhou, China
| |
Collapse
|
563
|
Bilotta AJ, Cong Y. Gut microbiota metabolite regulation of host defenses at mucosal surfaces: implication in precision medicine. PRECISION CLINICAL MEDICINE 2019; 2:110-119. [PMID: 31281735 PMCID: PMC6598739 DOI: 10.1093/pcmedi/pbz008] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/27/2019] [Accepted: 05/02/2019] [Indexed: 12/19/2022] Open
Abstract
The gut microbiota has a well-established role in the regulation of host homeostasis. Multiple factors control the composition and function of the microbiota. The westernization of diet, a shift away from nutrient-dense foods toward diets high in saturated fats, has been implicated in the rise of chronic inflammatory diseases such as inflammatory bowel disease (IBD). Diet is critical in the development and maintenance of a healthy microbiome, where dietary fiber (found in the highest amounts in fruits, vegetables, and legumes) is metabolized by the microbiome. In turn, the bacterial metabolites of dietary fiber, short chain fatty acids (SCFAs), regulate gut homeostasis. SCFAs engage G-protein coupled receptors (GPRs) and act as histone deacetylase inhibitors (HDACi) to module epithelial and immune cell functions in the intestines, where they generally promote an anti-inflammatory state. This review highlights the functions of SCFAs and their roles in the pathogenesis of IBD to provide insights into their potential therapeutic application for the treatment of IBD for the purposes of precision medicine.
Collapse
Affiliation(s)
- Anthony J Bilotta
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.,Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
564
|
Abstract
Innate pattern recognition receptors have been implicated in the obesity-associated imbalance of gut microbiota. In this issue of Cell Host & Microbe, Truax et al. (2018) report that NLRP12 prevents high-fat-diet-induced obesity by maintaining beneficial short-chain fatty acid-producing microbiota.
Collapse
|
565
|
Sargautiene V, Nakurte I, Nikolajeva V. Broad Prebiotic Potential of Non-starch Polysaccharides from Oats ( Avena sativa L.): an in vitro Study. Pol J Microbiol 2019; 67:307-313. [PMID: 30451447 PMCID: PMC7256768 DOI: 10.21307/pjm-2018-036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2018] [Indexed: 12/20/2022] Open
Abstract
Prebiotics inducing the growth or activity of beneficial intestinal bacteria – probiotics producing short-chain fatty acids (SCFA) have lately received wide recognition for their beneficial influence on host intestinal microbiota and metabolic health. Some non-starch polysaccharides (NSP) are defined as prebiotics and oats being one of richest sources of NSP in grains are considered as potentially having prebiotic effect. However, information on fermentation of specific NSP of oats is limited. Moreover, bacterial cross-feeding interactions in which fermentation of prebiotics is involved is poorly characterized. Here, we report the exploration of new candidates for the syntrophic bacterial interactions and fermentability of oat non-starch polysaccharides (NSP). The results obtained by differentiating composition, viscosity and concentration of oats NSP in fermentation medium showed that Bacillus licheniformis pre-digests oat NSP, degrades high viscosity of oat β-glucan and makes hemicellulose easier to access for other bacteria. Because of fermentation, B. licheniformis produces lactic and succinic acids, which further can be used by other bacteria for cross-feeding and SCFA production.
Collapse
Affiliation(s)
| | - Ilva Nakurte
- Department of Physical Chemistry, University of Latvia, Riga, Latvia
| | - Vizma Nikolajeva
- Department of Microbiology and Biotechnology, University of Latvia, Riga, Latvia
| |
Collapse
|
566
|
Wang Q, Fu W, Guo Y, Tang Y, Du H, Wang M, Liu Z, Li Q, An L, Tian J, Li M, Wu Z. Drinking Warm Water Improves Growth Performance and Optimizes the Gut Microbiota in Early Postweaning Rabbits during Winter. Animals (Basel) 2019; 9:E346. [PMID: 31212853 PMCID: PMC6616395 DOI: 10.3390/ani9060346] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/04/2019] [Accepted: 06/07/2019] [Indexed: 12/31/2022] Open
Abstract
Accumulating evidence indicates that cold exposure changes the composition of the gut microbiota and reduces intestinal immunity in early postweaning livestock. However, little is known about the effects of drinking warm water (WW) on gut microbiota during winter. In this study, we investigated the effects of drinking WW in winter on the growth performance and gut microbiota structure of rabbits raised in poorly insulated housing from the early postweaning period (day 46) to the subadult period (day 82). The average daily gain and feed conversion ratio in rabbits drinking WW were significantly improved compared to those of the rabbits drinking cold water (CW) during 47-58 days. In addition, rabbits drinking WW had a significantly decreased the risk of diarrhea during 71-82 days. 16S rRNA sequence analysis revealed that the alpha diversity of the cecal microbiota was not significantly different between the WW and CW groups, but significantly increased with age. The relative abundance of cecal microorganisms, such as Coprococcus spp. was considerably increased at day 70 in the group drinking WW. Correlation analysis indicated that Coprococcus spp. was negatively associated with pro-inflammatory factors. In conclusion, our results suggest that drinking WW has a positive effect on growth performance and gut microbiota in rabbits during the early postweaning stage in winter.
Collapse
Affiliation(s)
- Qiangjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Wei Fu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Yao Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Yuhan Tang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
- ZhaoTong Technology Promotion Workstation of Animal Husbandry and Veterinary Medicine, ZhaoTong 657000, China.
| | - Haoxuan Du
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Meizhi Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Zhongying Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Qin Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Lei An
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Jianhui Tian
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Mingyong Li
- National Rabbit Industry Technology System Qingdao Comprehensive Experimental Station, Qingdao 266431, China.
| | - Zhonghong Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
567
|
Zhou M, Yan B, Lang Q, Zhang Y. Elevated volatile fatty acids production through reuse of acidogenic off-gases during electro-fermentation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 668:295-302. [PMID: 30852206 DOI: 10.1016/j.scitotenv.2019.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/27/2019] [Accepted: 03/01/2019] [Indexed: 06/09/2023]
Abstract
Electro-fermentation is gaining attention for its advantage in promoting product recovery and valorization of organic wastes. However, emission of by-product gases during acidogenic fermentation is one of the key reasons for reduced product recovery whereas high gas pressure in the acidogenic headspace could pose an inhibitory effect on the production of volatile fatty acids (VFAs). This study presents a novel electro-fermentation (EF) system for enhancing VFAs production by in situ reuse of anodic off-gases (mainly CO2 and H2) in the cathode. A total VFAs production of 0.57 g-VFAs/g-VS was achieved through reuse of acidogenic off-gases in EF system, corresponding to 48.70% increase in comparison with the treatment without off-gases reuse. Consequently, the conversion efficiency of carbon to VFAs was improved significantly by 13.92%. Acidogenic metabolic pathway in the anode shifted to mixed -acid fermentation with the succession of dominant microbes from genus of Escherichia in the seeding inocula to Bacteroides and Desulfovibrio in the anode and cathode chambers, respectively. This would provide a way to enhance VFAs recovery from organic wastes, which also contributes to reduced carbon footprint and increased environmental sustainability.
Collapse
Affiliation(s)
- Miaomiao Zhou
- Lab of Waste Valorization and Water Reuse, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China; State Key Laboratory of Petroleum Pollution Control, Beijing 102206, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Binghua Yan
- Lab of Waste Valorization and Water Reuse, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China.
| | - Qiaolin Lang
- Lab of Waste Valorization and Water Reuse, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China
| | - Yang Zhang
- Lab of Waste Valorization and Water Reuse, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China
| |
Collapse
|
568
|
Di S, Wang Y, Han L, Bao Q, Gao Z, Wang Q, Yang Y, Zhao L, Tong X. The Intervention Effect of Traditional Chinese Medicine on the Intestinal Flora and Its Metabolites in Glycolipid Metabolic Disorders. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:2958920. [PMID: 31275408 PMCID: PMC6582858 DOI: 10.1155/2019/2958920] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/31/2019] [Accepted: 04/24/2019] [Indexed: 12/22/2022]
Abstract
Metabolic syndrome (MS), which includes metabolic disorders such as protein disorder, glucose disorder, lipid disorder, and carbohydrate disorder, has been growing rapidly around the world. Glycolipid disorders are a main type of metabolic syndrome and are characterized by abdominal obesity and abnormal metabolic disorders of lipid, glucose, and carbohydrate utilization, which can cause cardiovascular and cerebrovascular diseases. Glycolipid disorders are closely related to intestinal flora and its metabolites. However, studies about the biological mechanisms of the intestinal flora and its metabolites with glycolipid disorders have not been clear. When glycolipid disorders are treated with drugs, a challenging problem is side effects. Traditional Chinese medicine (TCM) and dietary supplements have fewer side effects to treat it. Numerous basic and clinical studies have confirmed that TCM decoctions, Chinese medicine monomers, or compounds can treat glycolipid disorders and reduce the incidence of cardiovascular disease. In this study, we reviewed the relationship between the intestinal flora and its metabolites in glycolipid metabolic disorders and the effect of TCM in treating glycolipid metabolic disorders through the intestinal flora and its metabolites. This review provides new perspectives and strategies for future glycolipid disorders research and treatment.
Collapse
Affiliation(s)
- Sha Di
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100054, China
| | - Yitian Wang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100054, China
| | - Lin Han
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100054, China
| | - Qi Bao
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100054, China
| | - Zezheng Gao
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100054, China
| | - Qing Wang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100054, China
| | - Yingying Yang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100054, China
| | - Linhua Zhao
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100054, China
| | - Xiaolin Tong
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100054, China
- Shenzhen Hospital, Guangzhou University of Chinese Medicine, Guangzhou 518034, China
| |
Collapse
|
569
|
El Hage R, Hernandez-Sanabria E, Calatayud Arroyo M, Props R, Van de Wiele T. Propionate-Producing Consortium Restores Antibiotic-Induced Dysbiosis in a Dynamic in vitro Model of the Human Intestinal Microbial Ecosystem. Front Microbiol 2019; 10:1206. [PMID: 31214145 PMCID: PMC6554338 DOI: 10.3389/fmicb.2019.01206] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 05/13/2019] [Indexed: 12/25/2022] Open
Abstract
Metabolic syndrome is a growing public health concern. Efforts at searching for links with the gut microbiome have revealed that propionate is a major fermentation product in the gut with several health benefits toward energy homeostasis. For instance, propionate stimulates satiety-inducing hormones, leading to lower energy intake and reducing weight gain and associated risk factors. In (disease) scenarios where microbial dysbiosis is apparent, gut microbial production of propionate may be decreased. Here, we investigated the effect of a propionogenic bacterial consortium composed of Lactobacillusplantarum, Bacteroidesthetaiotaomicron, Ruminococcusobeum, Coprococcuscatus, Bacteroidesvulgatus, Akkermansiamuciniphila, and Veillonellaparvula for its potential to restore in vitro propionate concentrations upon antibiotic-induced microbial dysbiosis. Using the mucosal simulator of the human intestinal microbial ecosystem (M-SHIME), we challenged the simulated colon microbiome with clindamycin. Addition of the propionogenic consortium resulted in successful colonization and subsequent restoration of propionate levels, while a positive effect on the mitochondrial membrane potential (ΔΨm) was observed in comparison with the controls. Our results support the development and application of next generation probiotics, which are composed of multiple bacterial strains with diverse functionality and phylogenetic background.
Collapse
Affiliation(s)
- Racha El Hage
- Center for Microbial Ecology and Technology, Ghent University, Ghent, Belgium
| | | | | | - Ruben Props
- Center for Microbial Ecology and Technology, Ghent University, Ghent, Belgium
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology, Ghent University, Ghent, Belgium
| |
Collapse
|
570
|
Ke S, Fang S, He M, Huang X, Yang H, Yang B, Chen C, Huang L. Age-based dynamic changes of phylogenetic composition and interaction networks of health pig gut microbiome feeding in a uniformed condition. BMC Vet Res 2019; 15:172. [PMID: 31126262 PMCID: PMC6534858 DOI: 10.1186/s12917-019-1918-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 05/16/2019] [Indexed: 12/26/2022] Open
Abstract
Background The gut microbiota impacts on a range of host biological processes, and the imbalances in its composition are associated with pathology. Though the understanding of contribution of the many factors, e.g. gender, diet and age, in the development of gut microbiota has been well established, the dynamic changes of the phylogenetic composition and the interaction networks along with the age remain unclear in pigs. Results Here we applied 16S ribosomal RNA gene sequencing, enterotype-like clustering (Classification of the gut microbiome into distinct types) and phylogenetic co-occurrence network to explore the dynamic changes of pig gut microbiome following the ages with a successive investigation at four ages in a cohort of 953 pigs. We found that Firmicutes and Bacteroidetes are two predominant phyla throughout the experimental period. The richness of gut microbiota was significantly increased from 25 to 240 days of age. Principal coordinates analysis showed a clear difference in the gut microbial community compositions between pre-weaning piglets and the pigs at the other three age groups. The gut microbiota of pre-weaning piglets was clearly classified into two enterotypes, which were dominated by Fusobacterium and p-75-a5, respectively. However, Prevotella and Treponema were the main drivers of the enterotypes for pigs at the age of 80, 120 and 240 days. Besides the piglets, even some adult pigs switched putative enterotypes between ages. We confirmed that the topological features of phylogenetic co-occurrence networks, including scale, stability and complexity were increased along with the age. The biological significance for modules in the network of piglets were mainly associated with the utilization of simple carbohydrate and lactose, whereas the sub-networks identified at the ages of 80, 120 and 240 days may be involved in the digestion of complex dietary polysaccharide. The modules related to the metabolism of protein and amino acids could be identified in the networks at 120 and 240 days. This dynamic change of the functional capacities of gut microbiome was further supported by functional prediction analysis. Conclusions The present study provided meaningful biological insights into the age-based dynamic shifts of ecological community of porcine gut microbiota. Electronic supplementary material The online version of this article (10.1186/s12917-019-1918-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shanlin Ke
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, 330045, Nanchang, People's Republic of China
| | - Shaoming Fang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, 330045, Nanchang, People's Republic of China
| | - Maozhang He
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, 330045, Nanchang, People's Republic of China
| | - Xiaochang Huang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, 330045, Nanchang, People's Republic of China
| | - Hui Yang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, 330045, Nanchang, People's Republic of China
| | - Bin Yang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, 330045, Nanchang, People's Republic of China
| | - Congying Chen
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, 330045, Nanchang, People's Republic of China.
| | - Lusheng Huang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, 330045, Nanchang, People's Republic of China.
| |
Collapse
|
571
|
Lordan C, Thapa D, Ross RP, Cotter PD. Potential for enriching next-generation health-promoting gut bacteria through prebiotics and other dietary components. Gut Microbes 2019; 11:1-20. [PMID: 31116628 PMCID: PMC6973326 DOI: 10.1080/19490976.2019.1613124] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The human intestinal commensal microbiota and associated metabolic products have long been regarded as contributors to host health. As the identity and activities of the various members of this community have become clearer, newly identified health-associated bacteria, such as Faecalibacterium prausnitzii, Akkermansia muciniphila, Ruminococcus bromii and Roseburia species, have emerged. Notably, the abundance of many of these bacteria is inversely correlated to several disease states. While technological and regulatory hurdles may limit the use of strains from these taxa as probiotics, it should be possible to utilize prebiotics and other dietary components to selectively enhance their growth in situ. Dietary components of potential relevance include well-established prebiotics, such as galacto-oligosaccharides, fructo-oligosaccharides and inulin, while other putative prebiotics, such as other oligosaccharides, polyphenols, resistant starch, algae and seaweed as well as host gut metabolites such as lactate and acetate, may also be applied with the aim of selectively and/or differentially affecting the beneficial bacterial community within the gastrointestinal environment. The present review provides an overview of the dietary components that could be applied in this manner.
Collapse
Affiliation(s)
- Cathy Lordan
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland,School of Microbiology, University College Cork, Ireland
| | - Dinesh Thapa
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - R. Paul Ross
- School of Microbiology, University College Cork, Ireland,APC Microbiome Ireland, University College Cork, Ireland
| | - Paul D. Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland,APC Microbiome Ireland, University College Cork, Ireland,CONTACT Paul D. Cotter
| |
Collapse
|
572
|
Zhu Y, Chen K, Ding Y, Situ D, Li Y, Long Y, Wang L, Ye J. Metabolic and proteomic mechanism of benzo[a]pyrene degradation by Brevibacillus brevis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 172:1-10. [PMID: 30665150 DOI: 10.1016/j.ecoenv.2019.01.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/07/2019] [Accepted: 01/10/2019] [Indexed: 06/09/2023]
Abstract
Benzo[a]pyrene (BaP) is a model compound of polycyclic aromatic hydrocarbons. The relationship between its toxicity and some target biomolecules has been investigated. To reveal the interactions of BaP biodegradation and metabolic network, BaP intermediates, proteome, carbon metabolism and ion transport were analyzed. The results show that 76% BaP was degraded by Brevibacillus brevis within 7 d through the cleavage of aromatic rings with the production of 1-naphthol and 2-naphthol. During this process, the expression of xylose isomerase was induced for xylose metabolism, whereas, α-cyclodextrin could no longer be metabolized. Lactic acid, acetic acid and oxalic acid at 0.1-1.2 mg dm-3 were released stemming from their enhanced biosynthesis in the pathways of pyruvate metabolism and citrate cycle, while 5-7 mg dm-3 of PO43- were transported for energy metabolism. The relative abundance of 43 proteins was significantly increased for pyruvate metabolism, citrate cycle, amino acid metabolism, purine metabolism, ribosome metabolism and protein synthesis.
Collapse
Affiliation(s)
- Yueping Zhu
- Technology Research Center for Petrochemical Resources Clean Utilization of Guangdong Province, Faculty of Environmental Science and Engineering, Guangdong University of Petrochemical Technology, Maoming 525000, Guangdong, China
| | - Kaiyun Chen
- Child Developmental-Behavioral Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Yingqi Ding
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, Guangdong, China
| | - Donglin Situ
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, Guangdong, China
| | - Yi Li
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, Guangdong, China
| | - Yan Long
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, Guangdong, China
| | - Lili Wang
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, Guangdong, China
| | - Jinshao Ye
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, Guangdong, China.
| |
Collapse
|
573
|
Targeting gut microbiota as a possible therapy for mastitis. Eur J Clin Microbiol Infect Dis 2019; 38:1409-1423. [PMID: 31079312 DOI: 10.1007/s10096-019-03549-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 03/25/2019] [Indexed: 02/07/2023]
Abstract
Mastitis, a disease that affects both dairy herds and humans, is recognized as the most common source of losses in the dairy industry. Antibiotics have been used for years as the primary treatment for mastitis. However, abuse of antibiotics has led to the emergence of resistant strains and the presence of drug residues and has increased the difficulty of curing this disease. In addition, antibiotics kill most of the microbes that are present in the digestive tract, leading to imbalances in the gut microbiome and destruction of the ecosystem that is normally present in the gut. Gut microbiota play an important role in the host's health and could be considered the "second brain" of the body. In recent years, the gut microbiota and their metabolites, including lipopolysaccharide (LPS) and short-chain fatty acids (SCFAs), have been shown to participate in the development of mastitis. LPS is the main component of the cell walls of gram-negative bacteria. Overproduction of rumen-derived LPS injures the rumen epithelium, resulting in the entry of LPS into the blood and damaged liver function; once in the blood, it circulates into the mammary gland, increasing blood-barrier permeability and leading to mammary gland inflammation. SCFAs, which are produced by gut microbiota as fermentation products, have a protective effect on mammary gland inflammatory responses and help maintain the function of the blood-milk barrier. Recently, increasing attention has been focused on the use of probiotics as a promising alternative for the treatment of mastitis. This review summarizes the effects of the gut microbiome and its metabolites on mastitis as well as the current of probiotics in mastitis. This work may provide a valuable theoretical foundation for the development of fresh ideas for the prevention and treatment of mastitis.
Collapse
|
574
|
Almeida D, Machado D, Andrade JC, Mendo S, Gomes AM, Freitas AC. Evolving trends in next-generation probiotics: a 5W1H perspective. Crit Rev Food Sci Nutr 2019; 60:1783-1796. [PMID: 31062600 DOI: 10.1080/10408398.2019.1599812] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In recent years, scientific community has been gathering increasingly more insight on the dynamics that are at play in metabolic and inflammatory disorders. These rapidly growing conditions are reaching epidemic proportions, bringing clinicians and researcher's new challenges. The specific roles and modulating properties that beneficial/probiotic bacteria hold in the context of the gut ecosystem seem to be key to avert these inflammatory and diet-related disorders. Faecalibacterium prausnitzii, Akkermansia muciniphila and Eubacterium hallii have been identified as candidates for next generation probiotics (NGPs) with exciting potential for the prevention and treatment of such of dysbiosis-associated diseases. The challenges of these non-conventional native gut bacteria lie mainly on their extreme sensitivity to O2 traces. If these strains are to be used successfully in food, supplements or drugs they need to be stable and active in humans. In the present review, we present an overall perspective of the most updated scientific literature on the newly called NGPs through the 5W1H (What, Why, Who, Where, When, and How) method, an innovative and attractive problem-solving approach that provides the reader an effective understanding of the issue at hand.
Collapse
Affiliation(s)
- Diana Almeida
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal.,Department of Biology and CESAM, University of Aveiro, Aveiro, Portugal
| | - Daniela Machado
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - José Carlos Andrade
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Gandra PRD, Portugal
| | - Sónia Mendo
- Department of Biology and CESAM, University of Aveiro, Aveiro, Portugal
| | - Ana Maria Gomes
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Ana Cristina Freitas
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| |
Collapse
|
575
|
Díaz-Sánchez S, Perrotta AR, Rockafellow I, Alm EJ, Okimoto R, Hawken R, Hanning I. Using fecal microbiota as biomarkers for predictions of performance in the selective breeding process of pedigree broiler breeders. PLoS One 2019; 14:e0216080. [PMID: 31063485 PMCID: PMC6504170 DOI: 10.1371/journal.pone.0216080] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 04/12/2019] [Indexed: 01/12/2023] Open
Abstract
Much work has been dedicated to identifying members of the microbial gut community that have potential to augment the growth rate of agricultural animals including chickens. Here, we assessed any correlations between the fecal microbiome, a proxy for the gut microbiome, and feed efficiency or weight gain at the pedigree chicken level, the highest tier of the production process. Because selective breeding is conducted at the pedigree level, our aim was to determine if microbiome profiles could be used to predict feed conversion or weight gain in order to improve selective breeding. Using 16s rRNA amplicon sequencing, we profiled the microbiomes of high and low weight gain (WG) birds and good and poor feed efficient (FE) birds in two pedigree lineages of broiler chickens. We also aimed to understand the dynamics of the microbiome with respect to maturation. A time series experiment was conducted, where fecal samples of chickens were collected at 6 points of the rearing process and the microbiome of these samples profiled. We identified OTUs differences at different taxonomic levels in the fecal community between high and low performing birds within each genetic line, indicating a specificity of the microbial community profiles correlated to performance factors. Using machine-learning methods, we built a classification model that could predict feed conversion performance from the fecal microbial community. With respect to maturation, we found that the fecal microbiome is dynamic in early life but stabilizes after 3 weeks of age independent of lineage. Our results indicate that the fecal microbiome profile can be used to predict feed conversion, but not weight gain in these pedigree lines. From the time series experiments, it appears that these predictions can be evaluated as early as 20 days of age. Our data also indicates that there is a genetic factor for the microbiome profile.
Collapse
Affiliation(s)
- Sandra Díaz-Sánchez
- Department of Food Science, University of Tennessee, Knoxville, TN, United States of America
- SaBio IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain
| | - Allison R. Perrotta
- Department of Civil & Environmental Engineering, MIT, Cambridge, MA, United States of America
| | - Isaac Rockafellow
- Department of Biological Engineering, MIT, Cambridge, MA, United States of America
| | - Eric J. Alm
- Department of Civil & Environmental Engineering, MIT, Cambridge, MA, United States of America
- Department of Biological Engineering, MIT, Cambridge, MA, United States of America
- The Center for Microbiome Informatics and Therapeutics, MIT, Cambridge, MA, United States of America
| | - Ron Okimoto
- Cobb-Vantress, Inc. Research and Development, Genomics and Quantitative Genetics, Siloam Springs, AR, United States of America
| | - Rachel Hawken
- Cobb-Vantress, Inc. Research and Development, Genomics and Quantitative Genetics, Siloam Springs, AR, United States of America
| | - Irene Hanning
- The Graduate School of Genome Sciences and Technology, University of Tennessee, Knoxville, TN, United States of America
- * E-mail:
| |
Collapse
|
576
|
Marietta E, Horwath I, Balakrishnan B, Taneja V. Role of the intestinal microbiome in autoimmune diseases and its use in treatments. Cell Immunol 2019; 339:50-58. [DOI: 10.1016/j.cellimm.2018.10.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/11/2018] [Accepted: 10/18/2018] [Indexed: 02/06/2023]
|
577
|
Ahmadi S, Nagpal R, Wang S, Gagliano J, Kitzman DW, Soleimanian-Zad S, Sheikh-Zeinoddin M, Read R, Yadav H. Prebiotics from acorn and sago prevent high-fat-diet-induced insulin resistance via microbiome-gut-brain axis modulation. J Nutr Biochem 2019; 67:1-13. [PMID: 30831458 PMCID: PMC6520164 DOI: 10.1016/j.jnutbio.2019.01.011] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/30/2018] [Accepted: 01/18/2019] [Indexed: 01/06/2023]
Abstract
Role of gut microbiome in obesity and type 2 diabetes (T2D) became apparent from several independent studies indicating that gut microbiome modulators like prebiotics may improve microbiome perturbations (dysbiosis) to ameliorate metabolic derangements. We herein isolate water soluble, nondigestible polysaccharides from five plant-based foods (acorn, quinoa, sunflower, pumpkin seeds and sago) and assess their impact on human fecal microbiome and amelioration of high-fat-diet (HFD)-induced obesity/T2D in mice. During polysaccharide isolation, purification, biochemical and digestion resistance characterization, and fermentation pattern by human fecal microbiome, we select acorn- and sago-derived prebiotics (on the basis of relatively higher purity and yield and lower protein contamination) and examine their effects in comparison to inulin. Prebiotics treatments in human fecal microbiome culture system not only preserve microbial diversity but also appear to foster beneficial bacteria and short-chain fatty acids (SCFAs). Feeding of acorn- and sago-derived prebiotics ameliorates HFD-induced glucose intolerance and insulin resistance in mice, with effects comparatively superior to those seen in inulin-fed mice. Feeding of both of novel prebiotics as well as inulin increases SCFAs levels in the mouse gut. Interestingly, gut hyperpermeability and mucosal inflammatory markers were significantly reduced upon prebiotics feeding in HFD-fed mice. Hypothalamic energy signaling in terms of increased expression of pro-opiomelanocortin was also modulated by prebiotics administration. Results demonstrate that these (and/or such) novel prebiotics can ameliorate HFD-induced defects in glucose metabolism via positive modulation of gut-microbiome-brain axis and hence could be useful in preventing/treating diet-induced obesity/T2D.
Collapse
Affiliation(s)
- Shokouh Ahmadi
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Food Science and Technology, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Ravinder Nagpal
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Shaohua Wang
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jason Gagliano
- National Center for the Biotechnology Workforce, Forsyth Technical Community College, Winston-Salem, NC, USA
| | - Dalane W Kitzman
- Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Sabihe Soleimanian-Zad
- Department of Food Science and Technology, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Mahmoud Sheikh-Zeinoddin
- Department of Food Science and Technology, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Russel Read
- National Center for the Biotechnology Workforce, Forsyth Technical Community College, Winston-Salem, NC, USA
| | - Hariom Yadav
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
578
|
Butowski CF, Thomas DG, Young W, Cave NJ, McKenzie CM, Rosendale DI, Bermingham EN. Addition of plant dietary fibre to a raw red meat high protein, high fat diet, alters the faecal bacteriome and organic acid profiles of the domestic cat (Felis catus). PLoS One 2019; 14:e0216072. [PMID: 31042730 PMCID: PMC6493751 DOI: 10.1371/journal.pone.0216072] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/12/2019] [Indexed: 02/07/2023] Open
Abstract
Commercial diets high in animal protein and fat are increasingly being developed for pets, however little is understood about the impacts of feeding such diets to domestic cats. The carbohydrate content of these diets is typically low, and dietary fibre is often not included. Dietary fibre is believed to be important in the feline gastrointestinal tract, promoting stool formation and providing a substrate for the hindgut microbiome. Therefore, we aimed to determine the effects of adding plant-based dietary fibre to a high animal protein and fat diet. Twelve domestic short hair cats were fed three complete and balanced diets in a cross-over design for blocks of 21 days: raw meat (Raw), raw meat plus fibre (2%, ‘as is’ inclusion of inulin and cellulose; Raw+Fibre) and a commercially available Kibble diet. A commercially available canned diet was fed for 21 days as a washout phase. Apparent macronutrient digestibility, faecal output, score, pH, organic acid concentrations and bacteriome profiles were determined. Diet significantly affected all faecal parameters measured. The addition of dietary fibre to the raw meat diet was found to reduce apparent macronutrient digestibility, increase faecal output, pH and score. Thirty one bacterial taxa were significantly affected by diet. Prevotella was found to dominate in the Kibble diet, Clostridium and Fusobacterium in the Raw diet, and Prevotella and a group of unclassified Peptostreptococcaceae in the Raw+Fibre diet. Our results show that diets of different macronutrient proportions can strongly influence the faecal microbiome composition and metabolism, as shown by altered organic acid concentrations and faecal pH, in the domestic cat. The addition of 2% of each fibre to the Raw diet shifted faecal parameters closer to those produced by feeding a Kibble diet. These results provide a basis for further research assessing raw red meat diets to domestic cats.
Collapse
Affiliation(s)
- Christina F Butowski
- Food & Bio-based Products, AgResearch Grasslands, Palmerston North, New Zealand.,Centre for Feline Nutrition, Massey University, Palmerston North, New Zealand
| | - David G Thomas
- Centre for Feline Nutrition, Massey University, Palmerston North, New Zealand
| | - Wayne Young
- Food & Bio-based Products, AgResearch Grasslands, Palmerston North, New Zealand.,Riddet Institute, Massey University, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Nick J Cave
- Centre for Feline Nutrition, Massey University, Palmerston North, New Zealand
| | | | - Douglas I Rosendale
- The New Zealand Institute for Plant & Food Research Ltd., Palmerston North, New Zealand
| | - Emma N Bermingham
- Food & Bio-based Products, AgResearch Grasslands, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| |
Collapse
|
579
|
Feeding Essential Oils to Neonatal Holstein Dairy Calves Results in Increased Ruminal Prevotellaceae Abundance and Propionate Concentrations. Microorganisms 2019; 7:microorganisms7050120. [PMID: 31052458 PMCID: PMC6560426 DOI: 10.3390/microorganisms7050120] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 04/26/2019] [Accepted: 04/29/2019] [Indexed: 12/31/2022] Open
Abstract
Since antibiotic use in animal production has become a public health concern, great efforts are being dedicated to find effective and viable alternatives. While essential oils (EO) have become attractive candidates for use in the livestock industry, their mode of action and microbial targets in food animals remain largely uncharacterized. To gain further insight, we investigated the rumen environment of neonatal calves fed calf starter pellets and milk replacer supplemented with a commercial blend of EO. Propionate concentrations were not only found to be higher in EO-fed calves compared to controls (P < 0.05), but ruminal bacterial communities also differed greatly. For instance, the abundance of Firmicutes was significantly lower in samples from EO-fed calves than in controls, which appeared to be mostly due to lower Lachnospiraceae levels (P < 0.05). In contrast, Bacteriodetes were more abundant in EO-fed calves compared to controls, which was primarily the result of higher Prevotellaceae (P < 0.05). Notably, two bacterial operational taxonomic units (OTUs) were significantly more abundant in EO-fed calves; SD_Bt-00966 was found to be a close relative of Prevotella ruminicola (97%), while SD_Bt-00978 likely corresponded to an uncharacterized species of Gammaproteobacteria. In addition, Pearson correlation and canonical correspondence analyses revealed potential associations between other ruminal bacterial OTUs and either short chain fatty acids (SCFA) parameters or metrics for calf growth. Together, these results support that EO supplementation in growing dairy calves can modulate rumen function through SCFA production and growth of specific rumen bacterial groups.
Collapse
|
580
|
Shigeno Y, Kitahara M, Shime M, Benno Y. Phascolarctobacterium wakonense sp. nov., isolated from common marmoset (Callithrix jacchus) faeces. Int J Syst Evol Microbiol 2019; 69:1941-1946. [PMID: 31038451 DOI: 10.1099/ijsem.0.003407] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Two strictly anaerobic strains (MB11T and MB56) were isolated from common marmoset (Callithrixjacchus) faeces. Cells of the two strains were Gram-stain-negative, pleomorphic short (strain MB11T) or long (strain MB56) rods. Phylogenetic analysis based on 16S rRNA gene sequences revealed that both isolates were related to the genus Phascolarctobacterium. They had 16S rRNA gene sequences similarities lower than 93 % to previously described species, Phascolarctobacterium faecium ACM 3679T and Phascolarctobacterium succinatutens YIT 12067T, and 98.7 % between themselves. DNA-DNA hybridization values showed that strains MB11T and MB56 were the same species. The genomic DNA G+C content of strains MB11T and MB56 were 47.3-47.4 mol% and 47.7-48.0 mol%. The isolates had different enzymatic activities compared with P. succinatutens JCM 16074T and different major cellular fatty acids compared with P. faecium ACM 3679T. Substrate availability revealed that they utilized not only succinate, but also pyruvate. With pyruvate supplementation, they produced both propionate and acetate, while only propionate production occured with succinate. As suggested by the phylogenic and physiological properties of strains MB11T and MB56, we propose the name Phascolarctobacteriumwakonense sp. nov. with the type strain MB11T (=JCM 32899T=DSM 107697T).
Collapse
Affiliation(s)
- Yuko Shigeno
- Benno Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science Technology and Innovation Hab, Wako, Saitama 351-0198, Japan
| | - Maki Kitahara
- Benno Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science Technology and Innovation Hab, Wako, Saitama 351-0198, Japan
| | - Mari Shime
- Benno Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science Technology and Innovation Hab, Wako, Saitama 351-0198, Japan
| | - Yoshimi Benno
- Benno Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science Technology and Innovation Hab, Wako, Saitama 351-0198, Japan
| |
Collapse
|
581
|
Menni C, Hernandez MM, Vital M, Mohney RP, Spector TD, Valdes AM. Circulating levels of the anti-oxidant indoleproprionic acid are associated with higher gut microbiome diversity. Gut Microbes 2019; 10:688-695. [PMID: 31030641 PMCID: PMC6866703 DOI: 10.1080/19490976.2019.1586038] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [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
The gut microbiome has recently emerged as an important regulator of insulin resistance and abdominal obesity. The tryptophan metabolite generated by the gut microbiome, indoleproprionic acid (IPA) has been shown to predict the onset of type 2 diabetes. IPA is a metabolite produced by gut microbes from dietary tryptophan that exhibits a high degree of inter-individual variation. The microbiome composition parameters that are associated with circulating levels of this potent anti-oxidant have however not been investigated to date in human populations. In 1018 middle-aged women from the TwinsUK cohort, we assessed the relationship between serum IPA levels and gut microbiome composition targeting the 16S rRNA gene. Microbiome alpha-diversity was positively correlated with serum indoleproprionic acid levels (Shannon Diversity: Beta[95%CI] = 0.19[0.13;0.25], P = 6.41 × 10-10) after adjustment for covariates. Sixteen taxa and 12 operational taxonomic units (OTUs) associated with IPA serum levels. Among these are positive correlations with the butyrate-producing Faecalibacterium prausnitzii, the class Mollicutes and the order RF39 of the Tenericutes, and Coprococcus Negative correlations instead were observed with Eubacterium dolichum previously shown to correlate with visceral fat mass and several genera in the Lachnospiraceae family such as Blautia and Ruminococcus previously shown to correlate with obesity. Microbiome composition parameters explained ~20% of the variation in circulating levels of IPA, whereas nutritional and host genetic parameters explained only ~4%. Our data confirm an association between IPA circulating levels and metabolic syndrome parameters and indicate that gut microbiome composition influences IPA levels.
Collapse
Affiliation(s)
- Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, UK,CONTACT Ana M Valdes School of Medicine, University of Nottingham, Clinical Sciences Bldg, City Hospital, Hucknall Rd, Nottingham NG5 1PB, UK
| | | | - Marius Vital
- Microbial Interactions and Processes, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Robert P. Mohney
- Discovery and Translational Sciences, Metabolon Inc, Raleigh-Durham, NC, USA
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, UK
| | - Ana M. Valdes
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, UK,MSK Theme, NIHR Nottingham Biomedical Research Centre, Nottingham, UK,School of Medicine, Nottingham City Hospital, Nottingham, UK
| |
Collapse
|
582
|
Mills S, Stanton C, Lane JA, Smith GJ, Ross RP. Precision Nutrition and the Microbiome, Part I: Current State of the Science. Nutrients 2019; 11:nu11040923. [PMID: 31022973 PMCID: PMC6520976 DOI: 10.3390/nu11040923] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/10/2019] [Accepted: 04/17/2019] [Indexed: 12/11/2022] Open
Abstract
The gut microbiota is a highly complex community which evolves and adapts to its host over a lifetime. It has been described as a virtual organ owing to the myriad of functions it performs, including the production of bioactive metabolites, regulation of immunity, energy homeostasis and protection against pathogens. These activities are dependent on the quantity and quality of the microbiota alongside its metabolic potential, which are dictated by a number of factors, including diet and host genetics. In this regard, the gut microbiome is malleable and varies significantly from host to host. These two features render the gut microbiome a candidate ‘organ’ for the possibility of precision microbiomics—the use of the gut microbiome as a biomarker to predict responsiveness to specific dietary constituents to generate precision diets and interventions for optimal health. With this in mind, this two-part review investigates the current state of the science in terms of the influence of diet and specific dietary components on the gut microbiota and subsequent consequences for health status, along with opportunities to modulate the microbiota for improved health and the potential of the microbiome as a biomarker to predict responsiveness to dietary components. In particular, in Part I, we examine the development of the microbiota from birth and its role in health. We investigate the consequences of poor-quality diet in relation to infection and inflammation and discuss diet-derived microbial metabolites which negatively impact health. We look at the role of diet in shaping the microbiome and the influence of specific dietary components, namely protein, fat and carbohydrates, on gut microbiota composition.
Collapse
Affiliation(s)
- Susan Mills
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland.
| | - Catherine Stanton
- APC Microbiome Ireland, Teagasc Food Research Centre, Fermoy P61 C996, Co Cork, Ireland.
| | - Jonathan A Lane
- H&H Group, Technical Centre, Global Research and Technology Centre, Cork P61 C996, Ireland.
| | - Graeme J Smith
- H&H Group, Technical Centre, Global Research and Technology Centre, Cork P61 C996, Ireland.
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland.
| |
Collapse
|
583
|
Prebiotic Supplementation of In Vitro Fecal Fermentations Inhibits Proteolysis by Gut Bacteria, and Host Diet Shapes Gut Bacterial Metabolism and Response to Intervention. Appl Environ Microbiol 2019; 85:AEM.02749-18. [PMID: 30824442 DOI: 10.1128/aem.02749-18] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 02/13/2019] [Indexed: 12/22/2022] Open
Abstract
Metabolism of protein by gut bacteria is potentially detrimental due to the production of toxic metabolites, such as ammonia, amines, p-cresol, and indole. The consumption of prebiotic carbohydrates results in specific changes in the composition and/or activity of the microbiota that may confer benefits to host well-being and health. Here, we have studied the impact of prebiotics on proteolysis within the gut in vitro Anaerobic stirred batch cultures were inoculated with feces from omnivores (n = 3) and vegetarians (n = 3) and four protein sources (casein, meat, mycoprotein, and soy protein) with and without supplementation by an oligofructose-enriched inulin. Bacterial counts and concentrations of short-chain fatty acids (SCFA), ammonia, phenol, indole, and p-cresol were monitored during fermentation. Addition of the fructan prebiotic Synergy1 increased levels of bifidobacteria (P = 0.000019 and 0.000013 for omnivores and vegetarians, respectively). Branched-chain fatty acids (BCFA) were significantly lower in fermenters with vegetarians' feces (P = 0.004), reduced further by prebiotic treatment. Ammonia production was lower with Synergy1. Bacterial adaptation to different dietary protein sources was observed through different patterns of ammonia production between vegetarians and omnivores. In volunteer samples with high baseline levels of phenol, indole, p-cresol, and skatole, Synergy1 fermentation led to a reduction of these compounds.IMPORTANCE Dietary protein intake is high in Western populations, which could result in potentially harmful metabolites in the gut from proteolysis. In an in vitro fermentation model, the addition of prebiotics reduced the negative consequences of high protein levels. Supplementation with a prebiotic resulted in a reduction of proteolytic metabolites in the model. A difference was seen in protein fermentation between omnivore and vegetarian gut microbiotas: bacteria from vegetarian donors grew more on soy and Quorn than on meat and casein, with reduced ammonia production. Bacteria from vegetarian donors produced less branched-chain fatty acids (BCFA).
Collapse
|
584
|
Zhang X, Grosfeld A, Williams E, Vasiliauskas D, Barretto S, Smith L, Mariadassou M, Philippe C, Devime F, Melchior C, Gourcerol G, Dourmap N, Lapaque N, Larraufie P, Blottière HM, Herberden C, Gerard P, Rehfeld JF, Ferraris RP, Fritton JC, Ellero-Simatos S, Douard V. Fructose malabsorption induces cholecystokinin expression in the ileum and cecum by changing microbiota composition and metabolism. FASEB J 2019; 33:7126-7142. [PMID: 30939042 DOI: 10.1096/fj.201801526rr] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Current fructose consumption levels often overwhelm the intestinal capacity to absorb fructose. We investigated the impact of fructose malabsorption on intestinal endocrine function and addressed the role of the microbiota in this process. To answer this question, a mouse model of moderate fructose malabsorption [ketohexokinase mutant (KHK)-/-] and wild-type (WT) littermate mice were used and received a 20%-fructose (KHK-F and WT-F) or 20%-glucose diet. Cholecystokinin (Cck) mRNA and protein expression in the ileum and cecum, as well as preproglucagon (Gcg) and neurotensin (Nts) mRNA expression in the cecum, increased in KHK-F mice. In KHK-F mice, triple-label immunohistochemistry showed major up-regulation of CCK in enteroendocrine cells (EECs) that were glucagon-like peptide-1 (GLP-1)+/Peptide YY (PYY-) in the ileum and colon and GLP-1-/PYY- in the cecum. The cecal microbiota composition was drastically modified in the KHK-F in association with an increase in glucose, propionate, succinate, and lactate concentrations. Antibiotic treatment abolished fructose malabsorption-dependent induction of cecal Cck mRNA expression and, in mouse GLUTag and human NCI-H716 cells, Cck mRNA expression levels increased in response to propionate, both suggesting a microbiota-dependent process. Fructose reaching the lower intestine can modify the composition and metabolism of the microbiota, thereby stimulating the production of CCK from the EECs possibly in response to propionate.-Zhang, X., Grosfeld, A., Williams, E., Vasiliauskas, D., Barretto, S., Smith, L., Mariadassou, M., Philippe, C., Devime, F., Melchior, C., Gourcerol, G., Dourmap, N., Lapaque, N., Larraufie, P., Blottière, H. M., Herberden, C., Gerard, P., Rehfeld, J. F., Ferraris, R. P., Fritton, J. C., Ellero-Simatos, S., Douard, V. Fructose malabsorption induces cholecystokinin expression in the ileum and cecum by changing microbiota composition and metabolism.
Collapse
Affiliation(s)
- Xufei Zhang
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France.,Collège Doctoral, Sorbonne Université, Paris, France
| | - Alexandra Grosfeld
- Centre de Recherche des Cordeliers, INSERM Unité Mixte de Recherche (UMR) S1138, Sorbonne Université, Sorbonne Cités, Université Paris-Diderot (UPD), Centre National de la Recherche Scientifique (CNRS)-Instituts Hospitalo-Universitaires (IHU), Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Edek Williams
- Department of Orthopedics, Rutgers University, Newark, New Jersey, USA
| | - Daniel Vasiliauskas
- Paris-Saclay Institute of Neuroscience, Université Paris Sud, Centre National de la Recherche Scientifique (CNRS), Université Paris-Saclay, Gif-sur-Yvette, France
| | | | | | - Mahendra Mariadassou
- Mathématiques et Informatique Appliquées du Génome à l'Environnement (MaIAGE), Unité de Recherche (UR) 1404, INRA, Jouy-en-Josas, France
| | - Catherine Philippe
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Fabienne Devime
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Chloé Melchior
- INSERM Unit 1073, University of Rouen (UNIROUEN), Normandie University, Rouen, France
| | - Guillaume Gourcerol
- INSERM Unit 1073, University of Rouen (UNIROUEN), Normandie University, Rouen, France
| | - Nathalie Dourmap
- UNIROUEN, INSERM U1245 and Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Normandy University, Rouen, France
| | - Nicolas Lapaque
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Pierre Larraufie
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Hervé M Blottière
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Christine Herberden
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Philippe Gerard
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; and
| | - Ronaldo P Ferraris
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University, Newark, New Jersey, USA
| | | | | | - Veronique Douard
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
585
|
Wang H, Ren P, Mang L, Shen N, Chen J, Zhang Y. In vitro fermentation of novel microwave-synthesized non-digestible oligosaccharides and their impact on the composition and metabolites of human gut microbiota. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.02.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
586
|
Dou J, Zeng J, Wu K, Tan W, Gao L, Lu J. Microbiosis in pathogenesis and intervention of atopic dermatitis. Int Immunopharmacol 2019; 69:263-269. [DOI: 10.1016/j.intimp.2019.01.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 01/21/2019] [Indexed: 12/19/2022]
|
587
|
Lee CS, Tan PL, Eor JY, Choi DH, Park M, Seo SK, Yoon S, Yang S, Kim SH. Prophylactic use of probiotic chocolate modulates intestinal physiological functions in constipated rats. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2019; 99:3045-3056. [PMID: 30488458 DOI: 10.1002/jsfa.9518] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/19/2018] [Accepted: 11/26/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND This study investigated the in vivo prophylactic effect of probiotic chocolate on constipation. Rats were administered chocolate containing 2.5 × 1010 CFU g-1 of probiotics daily for 4 weeks and treated with loperamide (5 mg kg-1 ) daily at the fourth week of treatment. RESULTS Probiotic chocolate treatment significantly (P < 0.05) increased the intestinal motility, colon length, fecal moisture content and number of excreted fecal pellets in constipated rats. Moreover, quantitative real-time polymerase chain reaction data and histological images also revealed that both probiotic chocolate LYC and BB12 treatments were capable of upregulating the mRNA expression levels of colonic ZO-1, occludin and AQP8, leading to the maintenance of the defensive barrier function in the constipated rats compared with the negative controls. Interestingly, these treatments also modulated gut bacterial populations by increasing the abundance levels of Lactobacillus and Bifidobacterium, as well as reducing the abundance level of Enterobacteriaceae. CONCLUSION The present study demonstrated that probiotic chocolate LYC and BB12 could potentially be used as alternative agents for prophylactic constipation. © 2018 Society of Chemical Industry.
Collapse
Affiliation(s)
- Chul Sang Lee
- College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Pei Lei Tan
- College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Ju Young Eor
- College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | | | - Miri Park
- Lotte R&D Center, Seoul, South Korea
| | | | | | | | - Sae Hun Kim
- College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| |
Collapse
|
588
|
Mikó E, Kovács T, Sebő É, Tóth J, Csonka T, Ujlaki G, Sipos A, Szabó J, Méhes G, Bai P. Microbiome-Microbial Metabolome-Cancer Cell Interactions in Breast Cancer-Familiar, but Unexplored. Cells 2019; 8:E293. [PMID: 30934972 PMCID: PMC6523810 DOI: 10.3390/cells8040293] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/18/2022] Open
Abstract
Breast cancer is a leading cause of death among women worldwide. Dysbiosis, an aberrant composition of the microbiome, characterizes breast cancer. In this review we discuss the changes to the metabolism of breast cancer cells, as well as the composition of the breast and gut microbiome in breast cancer. The role of the breast microbiome in breast cancer is unresolved, nevertheless it seems that the gut microbiome does have a role in the pathology of the disease. The gut microbiome secretes bioactive metabolites (reactivated estrogens, short chain fatty acids, amino acid metabolites, or secondary bile acids) that modulate breast cancer. We highlight the bacterial species or taxonomical units that generate these metabolites, we show their mode of action, and discuss how the metabolites affect mitochondrial metabolism and other molecular events in breast cancer. These metabolites resemble human hormones, as they are produced in a "gland" (in this case, the microbiome) and they are subsequently transferred to distant sites of action through the circulation. These metabolites appear to be important constituents of the tumor microenvironment. Finally, we discuss how bacterial dysbiosis interferes with breast cancer treatment through interfering with chemotherapeutic drug metabolism and availability.
Collapse
Affiliation(s)
- Edit Mikó
- Department of Medical Chemistry, University of Debrecen, 4032 Debrecen, Hungary.
- Department of Microbiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - Tünde Kovács
- Department of Medical Chemistry, University of Debrecen, 4032 Debrecen, Hungary.
| | - Éva Sebő
- Kenézy Breast Center, Kenézy Gyula County Hospital, 4032 Debrecen, Hungary.
| | - Judit Tóth
- Kenézy Breast Center, Kenézy Gyula County Hospital, 4032 Debrecen, Hungary.
| | - Tamás Csonka
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - Gyula Ujlaki
- Department of Medical Chemistry, University of Debrecen, 4032 Debrecen, Hungary.
| | - Adrienn Sipos
- Department of Medical Chemistry, University of Debrecen, 4032 Debrecen, Hungary.
| | - Judit Szabó
- Department of Microbiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - Péter Bai
- Department of Medical Chemistry, University of Debrecen, 4032 Debrecen, Hungary.
- MTA-DE Lendület Laboratory of Cellular Metabolism, 4032 Debrecen, Hungary.
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| |
Collapse
|
589
|
Liao X, Song L, Zeng B, Liu B, Qiu Y, Qu H, Zheng Y, Long M, Zhou H, Wang Y, Du Y, Xu J, Shen R, Tong Q, Cai L, Li X, Guo S, Yang G, Zhu Z, Pu X, Wei H, Zheng H. Alteration of gut microbiota induced by DPP-4i treatment improves glucose homeostasis. EBioMedicine 2019; 44:665-674. [PMID: 30922964 PMCID: PMC6603491 DOI: 10.1016/j.ebiom.2019.03.057] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/09/2019] [Accepted: 03/19/2019] [Indexed: 12/12/2022] Open
Abstract
Background Increasing evidence indicates that the gut microbiota contributes to the occurrence and development of metabolic diseases. However, little is known about the effects of commonly used antidiabetic agents on the gut microbiota. In this study, we investigated the roles of dipeptidyl peptidase-4 inhibitors (DPP-4i) and α-glucosidase inhibitor in modulating the gut microbiota. Methods 16S-rDNA sequencing was performed to analyse the effects of DPP-4i and acarbose on the gut microbiota in mice fed a high-fat diet (HFD). Fecal microbiota transplantation (FMT) from type 2 diabetes patients to germ-free mice was performed to investigate the contribution of the altered microbiome to antidiabetic effects of the drugs. Fecal metabolomics was also analysed by untargeted and targeted GC–MS systems. Findings Although DPP-4i and α-glucosidase inhibitor both altered the gut microbial composition, only the microbiome modulation of DPP-4i contributed to its hypoglycemic effect. Specifically, the changes of 68.6% genera induced by HFD were rescued by DPP-4i. FMT showed that the DPP-4i-altered microbiome improved glucose tolerance in colonized mice, while acarbose did not. Moreover, DPP-4i increased the abundance of Bacteroidetes, and also promoted a functional shift in the gut microbiome, especially increasing the production of succinate. Interpretation Our findings demonstrate an important effect of DPP-4i on the gut microbiota, revealing a new hypoglycemic mechanism and an additional benefit of it. Furthermore, modulating the microbial composition, and the functional shift arising from changes in the microbiome, might be a potential strategy for improving glucose homeostasis. Fund This work was supported by grants from the National Natural Science Foundation of China (No. 81700757, No. 81471039, No. 81700714 and No. 81770434), the National Key R&D Program of China (No. 2017YFC1309602, No. 2016YFC1101100, No. 2017YFD0500503 and No. 2017YFD0501001), and the Natural Science Foundation of Chongqing (No. cstc2014jcyjjq10006, No. cstc2016jcyjA0093 and No. cstc2016jcyjA0518).
Collapse
Affiliation(s)
- Xiaoyu Liao
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Lingyu Song
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing, China; Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Benhua Zeng
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Bingyao Liu
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yuyang Qiu
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Hua Qu
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yi Zheng
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Min Long
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Houdi Zhou
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yuan Wang
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yingxuan Du
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jing Xu
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Rufei Shen
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Qiang Tong
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Leiqin Cai
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xing Li
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shaodong Guo
- Department of Nutrition and Food Science, Texas A&M University, TX 77843, USA
| | - Gangyi Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xiaoyun Pu
- Department of Clinical Laboratory, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Hong Wei
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China.
| | - Hongting Zheng
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
590
|
Reddel S, Del Chierico F, Quagliariello A, Giancristoforo S, Vernocchi P, Russo A, Fiocchi A, Rossi P, Putignani L, El Hachem M. Gut microbiota profile in children affected by atopic dermatitis and evaluation of intestinal persistence of a probiotic mixture. Sci Rep 2019; 9:4996. [PMID: 30899033 PMCID: PMC6428866 DOI: 10.1038/s41598-019-41149-6] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 02/11/2019] [Indexed: 12/17/2022] Open
Abstract
Atopic dermatitis (AD) has been hypothesised to be associated with gut microbiota (GM) composition. We performed a comparative study of the GM profile of 19 AD children and 18 healthy individuals aimed at identifying bacterial biomarkers associated with the disease. The effect of probiotic intake (Bifidobacterium breve plus Lactobacillus salivarius) on the modulation of GM and the probiotic persistence in the GM were also evaluated. Faecal samples were analysed by real-time PCR and 16S rRNA targeted metagenomics. Although the probiotics, chosen for this study, did not shape the entire GM profile, we observed the ability of these species to pass through the gastrointestinal tract and to persist (only B. breve) in the GM. Moreover, the GM of patients compared to CTRLs showed a dysbiotic status characterised by an increase of Faecalibacterium, Oscillospira, Bacteroides, Parabacteroides and Sutterella and a reduction of short-chain fatty acid (SCFA)-producing bacteria (i.e., Bifidobacterium, Blautia, Coprococcus, Eubacterium and Propionibacterium). Taken togheter these results show an alteration in AD microbiota composition with the depletion or absence of some species, opening the way to future probiotic intervention studies.
Collapse
Affiliation(s)
- Sofia Reddel
- Human Microbiome Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | | | | | | | - Pamela Vernocchi
- Human Microbiome Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Alessandra Russo
- Human Microbiome Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Alessandro Fiocchi
- Unit of Allergology, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Paolo Rossi
- University Department of Pediatrics, Unit of Immune and Infectious Diseases, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Lorenza Putignani
- Human Microbiome Unit and Parasitology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy.
| | - May El Hachem
- Dermatology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| |
Collapse
|
591
|
Peterson CT, Sharma V, Iablokov SN, Albayrak L, Khanipov K, Uchitel S, Chopra D, Mills PJ, Fofanov Y, Rodionov DA, Peterson SN. 16S rRNA gene profiling and genome reconstruction reveal community metabolic interactions and prebiotic potential of medicinal herbs used in neurodegenerative disease and as nootropics. PLoS One 2019; 14:e0213869. [PMID: 30889210 PMCID: PMC6424447 DOI: 10.1371/journal.pone.0213869] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/01/2019] [Indexed: 12/31/2022] Open
Abstract
The prebiotic potential of nervine herbal medicines has been scarcely studied. We therefore used anaerobic human fecal cultivation to investigate whether medicinal herbs commonly used as treatment in neurological health and disease in Ayurveda and other traditional systems of medicine modulate gut microbiota. Profiling of fecal cultures supplemented with either Kapikacchu, Gotu Kola, Bacopa/Brahmi, Shankhapushpi, Boswellia/Frankincense, Jatamansi, Bhringaraj, Guduchi, Ashwagandha or Shatavari by 16S rRNA sequencing revealed profound changes in diverse taxa. Principal coordinate analysis highlights that each herb drives the formation of unique microbial communities predicted to display unique metabolic potential. The relative abundance of approximately one-third of the 243 enumerated species was altered by all herbs. Additional species were impacted in an herb-specific manner. In this study, we combine genome reconstruction of sugar utilization and short chain fatty acid (SCFA) pathways encoded in the genomes of 216 profiled taxa with monosaccharide composition analysis of each medicinal herb by quantitative mass spectrometry to enhance the interpretation of resulting microbial communities and discern potential drivers of microbiota restructuring. Collectively, our results indicate that gut microbiota engage in both protein and glycan catabolism, providing amino acid and sugar substrates that are consumed by fermentative species. We identified taxa that are efficient amino acid fermenters and those capable of both amino acid and sugar fermentation. Herb-induced microbial communities are predicted to alter the relative abundance of taxa encoding SCFA (butyrate and propionate) pathways. Co-occurrence network analyses identified a large number of taxa pairs in medicinal herb cultures. Some of these pairs displayed related culture growth relationships in replicate cultures highlighting potential functional interactions among medicinal herb-induced taxa.
Collapse
Affiliation(s)
- Christine Tara Peterson
- UC San Diego, School of Medicine, Center of Excellence for Research and Training in Integrative Health, Department of Family Medicine and Public Health, La Jolla, California, United States of America
- * E-mail:
| | - Vandana Sharma
- Sanford Burnham Prebys Medical Discovery Institute, Bioinformatics and Structural Biology Program, La Jolla, California, United States of America
| | - Stanislav N. Iablokov
- Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
- P.G. Demidov Yaroslavl State University, Yaroslavl, Russia
| | - Levent Albayrak
- Department of Pharmacology and Toxicology, Sealy Center for Structural Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kamil Khanipov
- Department of Pharmacology and Toxicology, Sealy Center for Structural Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Sasha Uchitel
- Washington University, Department of Biology, St. Louis, Missouri, United States of America
| | - Deepak Chopra
- UC San Diego, School of Medicine, Center of Excellence for Research and Training in Integrative Health, Department of Family Medicine and Public Health, La Jolla, California, United States of America
- Chopra Foundation, Department of Ayurveda and Yoga Research, Carlsbad, California, United States of America
| | - Paul J. Mills
- UC San Diego, School of Medicine, Center of Excellence for Research and Training in Integrative Health, Department of Family Medicine and Public Health, La Jolla, California, United States of America
| | - Yuriy Fofanov
- Department of Pharmacology and Toxicology, Sealy Center for Structural Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Dmitry A. Rodionov
- Sanford Burnham Prebys Medical Discovery Institute, Bioinformatics and Structural Biology Program, La Jolla, California, United States of America
- Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
| | - Scott N. Peterson
- Sanford Burnham Prebys Medical Discovery Institute, Bioinformatics and Structural Biology Program, La Jolla, California, United States of America
- Sanford Burnham Prebys Medical Discovery Institute, Tumor Microenvironment and Cancer Immunology Program, La Jolla, California, United States of America
| |
Collapse
|
592
|
Ferreira-Lazarte A, Moreno FJ, Cueva C, Gil-Sánchez I, Villamiel M. Behaviour of citrus pectin during its gastrointestinal digestion and fermentation in a dynamic simulator (simgi®). Carbohydr Polym 2019; 207:382-390. [DOI: 10.1016/j.carbpol.2018.11.088] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 11/26/2018] [Accepted: 11/27/2018] [Indexed: 12/31/2022]
|
593
|
Untargeted metabolomic evaluation of mango bagasse and mango bagasse based confection under in vitro simulated colonic fermentation. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.01.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
|
594
|
Su A, Ma G, Xie M, Ji Y, Li X, Zhao L, Hu Q. Characteristic of polysaccharides from
Flammulina velutipes in vitro
digestion under salivary, simulated gastric and small intestinal conditions and fermentation by human gut microbiota. Int J Food Sci Technol 2019. [DOI: 10.1111/ijfs.14142] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Anxiang Su
- College of Food Science and Technology Nanjing Agricultural University Nanjing 210095 Jiangsu China
| | - Gaoxing Ma
- College of Food Science and Engineering Nanjing University of Finance and Economics Nanjing 210023 Jiangsu China
| | - Minhao Xie
- College of Food Science and Engineering Nanjing University of Finance and Economics Nanjing 210023 Jiangsu China
| | - Yang Ji
- College of Food Science and Engineering Nanjing University of Finance and Economics Nanjing 210023 Jiangsu China
| | - Xiangfei Li
- College of Food Science and Engineering Nanjing University of Finance and Economics Nanjing 210023 Jiangsu China
| | - Liyan Zhao
- College of Food Science and Technology Nanjing Agricultural University Nanjing 210095 Jiangsu China
| | - Qiuhui Hu
- College of Food Science and Technology Nanjing Agricultural University Nanjing 210095 Jiangsu China
- College of Food Science and Engineering Nanjing University of Finance and Economics Nanjing 210023 Jiangsu China
| |
Collapse
|
595
|
Larsen N, Bussolo de Souza C, Krych L, Barbosa Cahú T, Wiese M, Kot W, Hansen KM, Blennow A, Venema K, Jespersen L. Potential of Pectins to Beneficially Modulate the Gut Microbiota Depends on Their Structural Properties. Front Microbiol 2019; 10:223. [PMID: 30828323 PMCID: PMC6384267 DOI: 10.3389/fmicb.2019.00223] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 01/28/2019] [Indexed: 01/16/2023] Open
Abstract
Pectins are plant cell-wall polysaccharides which can be utilized by commensal bacteria in the gut, exhibiting beneficial properties for the host. Knowledge of the impact of pectins on intestinal bacterial communities is insufficient and limited to a few types of pectins. This study characterized the relationship between the structural properties of pectins and their potential to modulate composition and activity of the gut microbiota in a beneficial way. For this purpose we performed in vitro fermentations of nine structurally diverse pectins from citrus fruits and sugar beet, and a pectic derivative, rhamnogalacturonan I (RGI), using a TIM-2 colon model. The composition of microbiota during TIM-2 fermentations was assessed by 16S rRNA gene amplicon sequencing. Both general and pectin-specific changes were observed in relative abundances of numerous bacterial taxa in a time-dependent way. Bacterial populations associated with human health, such as Faecalibacterium prausnitzii, Coprococcus, Ruminococcus, Dorea, Blautia, Oscillospira, Sutterella, Bifidobacterium, Christensenellaceae, Prevotella copri, and Bacteroides spp. were either increased or decreased depending on the substrate, suggesting that these bacteria can be controlled using structurally different pectins. The main structural features linked to the pectin-mediated shifts in microbiota included degree of esterification, composition of neutral sugars, distribution of homogalacturonan and rhamnogalacturonan fractions, degree of branching, and the presence of amide groups. Cumulative production of the total short chain fatty acids and propionate was largest in fermentations of the high methoxyl pectins. Thus, this study indicates that microbial communities in the gut can be specifically modulated by pectins and identifies the features in pectin molecules linked to microbial alterations. This knowledge can be used to define preferred dietary pectins, targeting beneficial bacteria, and favoring more balanced microbiota communities in the gut.
Collapse
Affiliation(s)
- Nadja Larsen
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - Carlota Bussolo de Souza
- Center for Healthy Eating and Food Innovation, Maastricht University - Campus Venlo, Maastricht, Netherlands
| | - Lukasz Krych
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | | | - Maria Wiese
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - Witold Kot
- Department of Environmental Science, Aarhus University, Roskilde, Denmark
| | | | - Andreas Blennow
- Department of Plant and Environmental Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Koen Venema
- Center for Healthy Eating and Food Innovation, Maastricht University - Campus Venlo, Maastricht, Netherlands.,Beneficial Microbes Consultancy, Wageningen, Netherlands
| | - Lene Jespersen
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
596
|
Tian X, Hellman J, Horswill AR, Crosby HA, Francis KP, Prakash A. Elevated Gut Microbiome-Derived Propionate Levels Are Associated With Reduced Sterile Lung Inflammation and Bacterial Immunity in Mice. Front Microbiol 2019; 10:159. [PMID: 30891007 PMCID: PMC6413706 DOI: 10.3389/fmicb.2019.00159] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 01/22/2019] [Indexed: 12/14/2022] Open
Abstract
Short-chain fatty acids (SCFA) are important dietary and microbiome metabolites that can have roles in gut immunity as well as further afield. We previously observed that gut microbiome alteration via antibiotics led to attenuated lung inflammatory responses. The rationale for this study was to identify gut microbiome factors that regulate lung immune homeostasis. We first investigated key factors within mouse colonic lumen filtrates (CLF) which could elicit direct inflammatory effects in vitro. We identified lipopolysaccharide (LPS) and SCFAs as key CLF ingredients whose levels and inflammatory capacity changed after antibiotic exposure in mice. Specifically, the SCFA propionate appeared to be a key regulator of LPS responses in vitro. Elevated propionate: acetate ratios, as seen in CLF after antibiotic exposure, strongly blunted inflammatory responses in vitro. In vivo, exposure of lungs to high dose propionate, to mimic how prior antibiotic exposure changed SCFA levels, resulted in diminished immune containment of Staphylococcus aureus pneumonia. Finally, we discovered an enrichment of propionate-producing gut bacteria in mice with reduced lung inflammation following lung ischemia reperfusion injury in vivo. Overall, our data show that propionate levels can distinctly modulate lung immune responses in vitro and in vivo and that gut microbiome increased production of propionate is associated with reduced lung inflammation.
Collapse
Affiliation(s)
- Xiaoli Tian
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
| | - Judith Hellman
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
| | - Heidi A. Crosby
- Department of Immunology and Microbiology, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
| | | | - Arun Prakash
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States.,San Francisco General Hospital, University of California, San Francisco, San Francisco, CA, United States
Approved by: Frontiers in Microbiology Editorial Office, Frontiers Media SA, Switzerland
| |
Collapse
|
597
|
Perdijk O, van Baarlen P, Fernandez-Gutierrez MM, van den Brink E, Schuren FHJ, Brugman S, Savelkoul HFJ, Kleerebezem M, van Neerven RJJ. Sialyllactose and Galactooligosaccharides Promote Epithelial Barrier Functioning and Distinctly Modulate Microbiota Composition and Short Chain Fatty Acid Production In Vitro. Front Immunol 2019; 10:94. [PMID: 30809221 PMCID: PMC6380229 DOI: 10.3389/fimmu.2019.00094] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/14/2019] [Indexed: 12/20/2022] Open
Abstract
Human milk oligosaccharides (HMO) and prebiotic oligosaccharides are proposed to confer several health benefits to the infant. They shape the microbiota, have anti-inflammatory properties, and support epithelial barrier functioning. However, in order to select the best oligosaccharides for inclusion in infant formulas, there is a need to increase our understanding of the specific effects of HMO and prebiotics on the host immune system. Therefore, we investigated the effects of the HMO sialyllactose (SL), and galactooligosaccharides (GOS) on epithelial barrier functioning, microbiota composition, and SCFA production. The effect of GOS and SL on epithelial barrier functioning and microbiota composition was investigated using in vitro models. Epithelial barrier function was investigated by transcriptome analysis of fully polarized Caco-2 cells exposed for 6 h to SL or GOS. In addition, epithelial cell growth, alkaline phosphatase production, and re-epithelization was studied. Further, we investigated the effect of SL and GOS on microbiota composition and SCFA production using in vitro fecal batch cultures. Transcriptome analysis showed that SL and GOS both induced pathways that regulate cell cycle control. This gene-expression profile translated to a phenotype of halted proliferation and included the induction of alkaline phosphatase activity, a marker of epithelial cell differentiation. SL and GOS also promoted re-epithelialization in an in vitro epithelial wound repair assay. SL and GOS did show distinct modulation of microbiota composition, promoting the outgrowth of Bacteroides and bifidobacteria, respectively, which resulted in distinct changes in SCFA production profiles. Our results show that SL and GOS can both modulate epithelial barrier function by inducing differentiation and epithelial wound repair, but differentially promote the growth of specific genera in the microbiota, which is associated with differential changes in SCFA profiles.
Collapse
Affiliation(s)
- Olaf Perdijk
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands
| | - Peter van Baarlen
- Host-Microbe Interactomics Group, Wageningen University & Research, Wageningen, Netherlands
| | | | - Erik van den Brink
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands
| | - Frank H. J. Schuren
- Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research, Zeist, Netherlands
| | - Sylvia Brugman
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands
| | - Huub F. J. Savelkoul
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands
| | - Michiel Kleerebezem
- Host-Microbe Interactomics Group, Wageningen University & Research, Wageningen, Netherlands
| | - R. J. Joost van Neerven
- Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands
- FrieslandCampina, Amersfoort, Netherlands
| |
Collapse
|
598
|
Wu CH, Ko JL, Liao JM, Huang SS, Lin MY, Lee LH, Chang LY, Ou CC. D-methionine alleviates cisplatin-induced mucositis by restoring the gut microbiota structure and improving intestinal inflammation. Ther Adv Med Oncol 2019; 11:1758835918821021. [PMID: 30792823 PMCID: PMC6376546 DOI: 10.1177/1758835918821021] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 11/13/2018] [Indexed: 02/06/2023] Open
Abstract
Background There are close links between chemotherapy-induced intestinal mucositis and microbiota dysbiosis. Previous studies indicated that D-methionine was an excellent candidate for a chemopreventive agent. Here, we investigated the effects of D-methionine on cisplatin-induced mucositis. Materials and methods Male Wistar rats (176-200 g, 6 weeks old) were given cisplatin (5 mg/kg) and treated with D-methionine (300 mg/kg). Histopathological, digestive enzymes activity, oxidative/antioxidant status, proinflammatory/anti-inflammatory cytokines in intestinal tissues were measured. Next-generation sequencing technologies were also performed to investigate the gut microbial ecology. Results D-methionine administration increased villus length and crypt depth and improved digestive enzyme (leucine aminopeptidase, sucrose and alkaline phosphatase) activities in the brush-border membrane of cisplatin-treated rats (p < 0.05). Furthermore, D-methionine significantly attenuated oxidative stress and inflammatory reaction and increased interleukin-10 levels in cisplatin-induced intestinal mucositis (p < 0.05). Cisplatin administration resulted in high relative abundances of Deferribacteres and Proteobacteria and a low diversity of the microbiota when compared with control groups, D-methionine only and cisplatin plus D-methionine. Cisplatin markedly increased comparative abundances of Bacteroides caccae, Escherichia coli, Mucispirillum schaedleri, Bacteroides uniformis and Desulfovibrio C21-c20, while Lactobacillus was almost completely depleted, compared with the control group. There were higher abundances of Lactobacillus, Lachnospiraceae, and Clostridium butyrium in cisplatin plus D-methionine rats than in cisplatin rats. D-methionine treatment alone significantly increased the number of Lactobacillus reuteri. Conclusion D-methionine protects against cisplatin-induced intestinal damage through antioxidative and anti-inflammatory effects. By enhancing growth of beneficial bacteria (Lachnospiraceae and Lactobacillus), D-methionine attenuates gut microbiome imbalance caused by cisplatin and maintains gut homeostasis.
Collapse
Affiliation(s)
- Cheng-Hsi Wu
- Department of Family Medicine, Jen-Ai Hospital, Dali, Taichung, Taiwan
| | - Jiunn-Liang Ko
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Jiuan-Miaw Liao
- Department of Physiology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Shiang-Suo Huang
- Department of Pharmacology and Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Meei-Yn Lin
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Ling-Hui Lee
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Li-Yu Chang
- Department of Nursing, Jen-Ai Hospital, Dali, Taichung, Taiwan
| | - Chu-Chyn Ou
- School of Nutrition, Chung Shan Medical University, 110, Sec. 1, Chien-Kuo N. Road, Taichung 40203, Taiwan
| |
Collapse
|
599
|
Moraïs S, Mizrahi I. The Road Not Taken: The Rumen Microbiome, Functional Groups, and Community States. Trends Microbiol 2019; 27:538-549. [PMID: 30679075 DOI: 10.1016/j.tim.2018.12.011] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/28/2018] [Accepted: 12/19/2018] [Indexed: 02/07/2023]
Abstract
The rumen ecosystem represents a classic example of host-microbiome symbiosis. In this obligatory relationship, the host feeds on plant fibers that can only be degraded through a set of complex metabolic cascades, exclusively encoded in rumen-associated microbes. These various metabolic pathways are distributed across a multitude of microbial populations. Application of basic ecological principles to this ecosystem can contribute to profound understanding of the rules that shape it. Here, we discuss recent studies by examining the mapping between host attributes, rumen ecosystem composition, and functionality to propose simple, yet powerful concepts to guide the interpretation of microbiome data and enable a better understanding of how the system responds to perturbations.
Collapse
Affiliation(s)
- Sarah Moraïs
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, 8410501 Beer-Sheva, Israel
| | - Itzhak Mizrahi
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, 8410501 Beer-Sheva, Israel.
| |
Collapse
|
600
|
Wu Y, Hu H, Dai X, Che H, Zhang H. Effects of dietary intake of potatoes on body weight gain, satiety-related hormones, and gut microbiota in healthy rats. RSC Adv 2019; 9:33290-33301. [PMID: 35529109 PMCID: PMC9073283 DOI: 10.1039/c9ra04867g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/03/2019] [Indexed: 12/27/2022] Open
Abstract
Potatoes, as a prominent staple food, have exerted diverse intestinal health benefits, but few studies have addressed the gut microecology modulatory effects of consuming potatoes in realistic quantities. The objective of this study was to evaluate the effects of ingesting potatoes in different doses on body weight gain (BWG), food intake, short chain fatty acids (SCFAs), fecal microbiota, gut hormones, and colon morphology of healthy rats. Male Sprague-Dawley rats of 6–8 weeks old were randomized to five groups and fed AIN-93 G or diets containing graded concentrations of potato powder (low, medium, high, and higher) for 7 weeks. Accordingly, the final body weight was significantly lower for rats fed the high and/or higher potato diets than their control counterparts (P < 0.05). Potato intervention caused a significant dose-dependent increment in full cecum, and SCFAs production. The relative abundance of “S24-7” (order Bacteroidales), Bifidobacterium, “NK3B31” (family Prevotellaceae), Parasutterella, and Ruminococcus_1 increased in high and higher potato diets. Furthermore, a Spearman's correlation analysis revealed that Parasutterella was negatively correlated with BWG, triglyceride (TG), and low-density lipoproteins (LDL). The maximum number of goblet cells, longest crypt depth, and highest level of PYY were found in the distal colon of rats fed higher potato diets. The results suggested that potato powder could provide the potential for hopeful impact on weight control. Supplementation of potato powders with 54.88 g kg−1 would significantly reduce the body weight gain by enriching Bifidobacterium and Parasutterella.![]()
Collapse
Affiliation(s)
- Yu Wu
- Institute of Food Science and Technology
- Chinese Academy of Agricultural Sciences (CAAS)/Key Laboratory of Agro-Products Processing
- Ministry of Agriculture
- Beijing 100193
- China
| | - Honghai Hu
- Institute of Food Science and Technology
- Chinese Academy of Agricultural Sciences (CAAS)/Key Laboratory of Agro-Products Processing
- Ministry of Agriculture
- Beijing 100193
- China
| | - Xiaofeng Dai
- Institute of Food Science and Technology
- Chinese Academy of Agricultural Sciences (CAAS)/Key Laboratory of Agro-Products Processing
- Ministry of Agriculture
- Beijing 100193
- China
| | - Huilian Che
- College of Food Science and Nutritional Engineering
- China Agricultural University
- Beijing 100083
- China
| | - Hong Zhang
- Institute of Food Science and Technology
- Chinese Academy of Agricultural Sciences (CAAS)/Key Laboratory of Agro-Products Processing
- Ministry of Agriculture
- Beijing 100193
- China
| |
Collapse
|