1
|
He X, Zhang Z, Jiang H, Luo H, Gan Q, Wei K, Liu Y, Qin Y, Xiao M. Causal association of gut microbes and blood metabolites with acne identified through systematic mendelian randomization. Sci Rep 2024; 14:26816. [PMID: 39501024 PMCID: PMC11538280 DOI: 10.1038/s41598-024-78603-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024] Open
Abstract
Acne is a prevalent inflammatory disease in dermatology, and its pathogenesis may be associated with inflammation, immunity, and other mechanisms. It commonly manifests in young individuals and frequently imposes a heavy economic, physical, and psychological burden on patients. Gut microbes and blood metabolites, as significant immune and inflammatory regulators in the body, have been hypothesized to form the "neurocutaneous axis." Nonetheless, the precise causal relationships among the gut microbes, circulating blood metabolites, and acne development have yet to be elucidated. This study employed bidirectional two-sample Mendelian randomization (MR) to probe the causal impacts of 412 distinct gut microbes and 249 blood metabolites on acne. Single nucleotide polymorphisms (SNPs), which are closely associated with gut microbes and blood metabolites, were utilized as instrumental variables. This approach was taken to discern whether these elements serve as pathogenic or protective factors in relation to acne. Furthermore, a mediation analysis encompassing gut microbes, blood metabolites, and acne was conducted to explore potential correlations between gut microbes and blood metabolites, as well as their cumulative effects on acne. This was done to substantiate the notion of causality. Bidirectional two-sample MR analysis revealed 8 gut bacteria, 6 bacterial metabolic abundance pathways determined by birdshot, and 8 blood metabolites significantly associated with acne. The mediation MR analysis revealed 2 potential causal relationships, namely, Bifidobacterium-DHA-Acne and Bifidobacterium-Degree of Unsaturation-Acne. This study identified gut microbes and blood metabolites that are causally associated with acne. A potential causal relationship between gut microbes and blood metabolites was obtained via mediation analysis. These insights pave the way for the identification of new targets and the formulation of innovative approaches for the prevention and treatment of acne.
Collapse
Affiliation(s)
- Xin He
- Chengdu Integrated TCM and Western Medicine Hospital, Chengdu, Sichuan Province, P R China
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
| | - Zhongyi Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
| | - Hengyu Jiang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
| | - Hui Luo
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
| | - Qianrong Gan
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
| | - Kebo Wei
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
| | - Ying Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
| | - Yuesi Qin
- Chengdu Integrated TCM and Western Medicine Hospital, Chengdu, Sichuan Province, P R China.
| | - Min Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China.
| |
Collapse
|
2
|
Sempach L, Doll JPK, Limbach V, Marzetta F, Schaub AC, Schneider E, Kettelhack C, Mählmann L, Schweinfurth-Keck N, Ibberson M, Lang UE, Schmidt A. Examining immune-inflammatory mechanisms of probiotic supplementation in depression: secondary findings from a randomized clinical trial. Transl Psychiatry 2024; 14:305. [PMID: 39048549 PMCID: PMC11269721 DOI: 10.1038/s41398-024-03030-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024] Open
Abstract
We recently indicated that four-week probiotic supplementation significantly reduced depression along with microbial and neural changes in people with depression. Here we further elucidated the biological modes of action underlying the beneficial clinical effects of probiotics by focusing on immune-inflammatory processes. The analysis included a total of N = 43 participants with depression, from which N = 19 received the probiotic supplement and N = 24 received a placebo over four weeks, in addition to treatment as usual. Blood and saliva were collected at baseline, at post-intervention (week 4) and follow-up (week 8) to assess immune-inflammatory markers (IL-1β, IL-6, CRP, MIF), gut-related hormones (ghrelin, leptin), and a stress marker (cortisol). Furthermore, transcriptomic analyses were conducted to identify differentially expressed genes. Finally, we analyzed the associations between probiotic-induced clinical and immune-inflammatory changes. We observed a significant group x time interaction for the gut hormone ghrelin, indicative of an increase in the probiotics group. Additionally, the increase in ghrelin was correlated with the decrease in depressive symptoms in the probiotics group. Transcriptomic analyses identified 51 up- and 57 down-regulated genes, which were involved in functional pathways related to enhanced immune activity. We identified a probiotic-dependent upregulation of the genes ELANE, DEFA4 and OLFM4 associated to immune activation and ghrelin concentration. These results underscore the potential of probiotic supplementation to produce biological meaningful changes in immune activation in patients with depression. Further large-scale mechanistic trials are warranted to validate and extend our understanding of immune-inflammatory measures as potential biomarkers for stratification and treatment response in depression. Trial Registration: www.clinicaltrials.gov , identifier: NCT02957591.
Collapse
Affiliation(s)
- Lukas Sempach
- Translational Neuroscience, Department of Clinical Research (DKF), University of Basel, Basel, Switzerland.
- University Psychiatric Clinics Basel (UPK), University of Basel, Basel, Switzerland.
| | - Jessica P K Doll
- Translational Neuroscience, Department of Clinical Research (DKF), University of Basel, Basel, Switzerland
- University Psychiatric Clinics Basel (UPK), University of Basel, Basel, Switzerland
| | - Verena Limbach
- Translational Neuroscience, Department of Clinical Research (DKF), University of Basel, Basel, Switzerland
- University Psychiatric Clinics Basel (UPK), University of Basel, Basel, Switzerland
| | - Flavia Marzetta
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Anna-Chiara Schaub
- University Psychiatric Clinics Basel (UPK), University of Basel, Basel, Switzerland
- Translational Psychiatry, Department of Clinical Research (DKF), University of Basel, Basel, Switzerland
| | - Else Schneider
- University Psychiatric Clinics Basel (UPK), University of Basel, Basel, Switzerland
- Experimental Cognitive and Clinical Affective Neuroscience (ECAN) Laboratory, Department of Clinical Research (DKF), University of Basel, Basel, Switzerland
| | - Cedric Kettelhack
- University Psychiatric Clinics Basel (UPK), University of Basel, Basel, Switzerland
| | - Laura Mählmann
- University Psychiatric Clinics Basel (UPK), University of Basel, Basel, Switzerland
| | | | - Mark Ibberson
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Undine E Lang
- University Psychiatric Clinics Basel (UPK), University of Basel, Basel, Switzerland
| | - André Schmidt
- Translational Neuroscience, Department of Clinical Research (DKF), University of Basel, Basel, Switzerland
- University Psychiatric Clinics Basel (UPK), University of Basel, Basel, Switzerland
| |
Collapse
|
3
|
Zhao Y, Bao D, Sun Y, Meng Y, Li Z, Liu R, Lang J, Liu L, Gao L. Comparative analysis of the gut bacteria of the relict gull ( Larus Relictus) and black-necked grebe ( Podiceps Nigricollis) in Erdos Relic Gull National Nature Reserve in Inner Mongolia, China. PeerJ 2023; 11:e15462. [PMID: 37456862 PMCID: PMC10340111 DOI: 10.7717/peerj.15462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/04/2023] [Indexed: 07/18/2023] Open
Abstract
The gut microbiota promotes host health by maintaining homeostasis and enhancing digestive efficiency. The gut microflora in wild birds affects host physiological characteristics, nutritional status, and stress response. The relict gull (Larus Relictus, a Chinese national first-class protected species) and the black-necked grebe (Podiceps Nigricollis, a secondary protected species) bred in the Ordos Relic Gull National Nature Reserve share similar feeding habits and living environments but are distantly related genetically. To explore the composition and differences in the gut microbiota of these two key protected avian species in Erdos Relic Gull National Nature Reserve and provide a basis for their protection, 16S rRNA gene high-throughput sequencing was performed and the gut microbial diversity and composition of the relict gull (L. Relictus) and black-necked grebe (P. Nigricollis) was characterized. In total, 445 OTUs (operational taxonomic units) were identified and classified into 15 phyla, 22 classes, 64 orders, 126 families, and 249 genera. Alpha diversity analysis indicates that the gut microbial richness of the relict gull is significantly lower than that of the black-necked grebe. Gut microbe composition differs significantly between the two species. The most abundant bacterial phyla in these samples were Proteobacteria, Firmicutes, Fusobacteria, and Bacteroidetes. The prominent phylum in the relict gull was Proteobacteria, whereas the prominent phylum in the black-necked grebe was Firmicutes. The average relative abundance of the 17 genera identified was greater than 1%. The dominant genus in the relict gull was Escherichia-Shigella, whereas Halomonas was dominant in the black-necked grebe. Microbial functional analyses indicate that environmental factors exert a greater impact on relict gulls than on black-necked grebes. Compared with the relict gull, the black-necked grebe was able to use food more efficiently to accumulate its nutrient requirements, and the gut of the relict gull harbored more pathogenic bacteria, which may be one reason for the decline in the relict gull population, rendering it an endangered species. This analysis of the gut microbial composition of these two wild avian species in the same breeding grounds is of great significance, offers important guidance for the protection of these two birds, especially relict gulls, and provides a basis for understanding the propagation of related diseases.
Collapse
Affiliation(s)
- Yaru Zhao
- Faculty of Biological Science and Technology, Baotou Teachers' College, Baotou, Inner Mongolia, China
| | - Dulan Bao
- Faculty of Biological Science and Technology, Baotou Teachers' College, Baotou, Inner Mongolia, China
| | - Ying Sun
- Faculty of Biological Science and Technology, Baotou Teachers' College, Baotou, Inner Mongolia, China
| | - Yajie Meng
- Faculty of Biological Science and Technology, Baotou Teachers' College, Baotou, Inner Mongolia, China
| | - Ziteng Li
- Faculty of Biological Science and Technology, Baotou Teachers' College, Baotou, Inner Mongolia, China
| | - Rui Liu
- Faculty of Biological Science and Technology, Baotou Teachers' College, Baotou, Inner Mongolia, China
| | - Jiwei Lang
- Faculty of Biological Science and Technology, Baotou Teachers' College, Baotou, Inner Mongolia, China
| | - Li Liu
- Faculty of Biological Science and Technology, Baotou Teachers' College, Baotou, Inner Mongolia, China
| | - Li Gao
- Faculty of Biological Science and Technology, Baotou Teachers' College, Baotou, Inner Mongolia, China
| |
Collapse
|
4
|
Wei L, Zeng B, Zhang S, Guo W, Li F, Zhao J, Li Y. Hybridization altered the gut microbiota of pigs. Front Microbiol 2023; 14:1177947. [PMID: 37465027 PMCID: PMC10350513 DOI: 10.3389/fmicb.2023.1177947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/14/2023] [Indexed: 07/20/2023] Open
Abstract
Mammalian gut microbiota plays an important role in the host's nutrient metabolism, growth, and immune regulation. Hybridization can enable a progeny to acquire superior traits of the parents, resulting in the hybridization advantage. However, studies on the effects of hybridization on the pigs' gut microbiota are lacking. Therefore, this study used multi-omics technologies to compare and analyze the gut microbiota of the primary wild boar and its offspring. The 16S rRNA gene sequencing results revealed that the gut microbiota of F4 exhibited a host-like dominance phenomenon with a significant increase in the abundance of Lactobacillus and Bifidobacterium. The beta diversity of Duroc was significantly different from those of F0, F2, and F4; after the host hybridization, the similarity of the beta diversity in the progeny decreased with the decrease in the similarity of the F0 lineage. The metagenomic sequencing results showed that the significantly enriched metabolic pathways in F4, such as environmental, circulatory system, fatty acid degradation adaptation, and fatty acid biosynthesis, were similar to those in F0. Moreover, it also exhibited similar significantly enriched metabolic pathways as those in Duroc, such as carbohydrate metabolism, starch and sucrose metabolism, starch-degrading CAZymes, lactose-degrading CAZymes, and various amino acid metabolism pathways. However, the alpha-amylase-related KOs, lipid metabolism, and galactose metabolism in F4 were significantly higher than those in Duroc and F0. Non-targeted metabolome technology analysis found that several metabolites, such as docosahexaenoic acid, arachidonic acid, and citric acid were significantly enriched in the F4 pigs as compared to those in F0. Based on Spearman correlation analysis, Lactobacillus and Bifidobacterium were significantly positively correlated with these metabolites. Finally, the combined metagenomic and metabolomic analysis suggested that the metabolic pathways, such as valine, leucine, and isoleucine biosynthesis and alanine aspartate and glutamate metabolism were significantly enriched in F4 pigs. In conclusion, the gut microbiota of F4 showed a similar host "dominance" phenomenon, which provided reference data for the genetics and evolution of microbiota and the theory of microbial-assisted breeding.
Collapse
Affiliation(s)
- Limin Wei
- Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Medical and Pharmaceutical College, Chongqing, China
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, College of Life Science and Engineering, Foshan University, Foshan, China
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Bo Zeng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Siyuan Zhang
- School of Laboratory Medicine/Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-Origin Food, Chengdu Medical College, Chengdu, China
| | - Wei Guo
- School of Laboratory Medicine/Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-Origin Food, Chengdu Medical College, Chengdu, China
| | - Feng Li
- Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, Nanchong, China
| | - Jiangchao Zhao
- Division of Agriculture, Department of Animal Science, University of Arkansas, Fayetteville, AR, United States
| | - Ying Li
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, College of Life Science and Engineering, Foshan University, Foshan, China
| |
Collapse
|
5
|
Moore RL, Feehily C, Killeen SL, Yelverton CA, Geraghty AA, Walsh CJ, O'Neill IJ, Nielsan IB, Lawton EM, Sanchez-Gallardo R, Nori SRC, Shanahan F, Murphy EF, Van Sinderen D, Cotter PD, McAuliffe FM. Ability of Bifidobacterium breve 702258 to transfer from mother to infant: the MicrobeMom randomized controlled trial. Am J Obstet Gynecol MFM 2023; 5:100994. [PMID: 37142190 DOI: 10.1016/j.ajogmf.2023.100994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND The composition of the infant microbiome can have a variety of short- and long-term implications for health. It is unclear if maternal probiotic supplementation in pregnancy can affect the infant gut microbiome. OBJECTIVE This study aimed to investigate if maternal supplementation of a formulation of Bifidobacterium breve 702258 from early pregnancy until 3 months postpartum could transfer to the infant gut. STUDY DESIGN This was a double-blinded, placebo-controlled, randomized controlled trial of B breve 702258 (minimum 1 × 109 colony-forming units) or placebo taken orally from 16 weeks' gestation until 3 months postpartum in healthy pregnant women. The primary outcome was presence of the supplemented strain in infant stool up to 3 months of life, detected by at least 2 of 3 methods: strain-specific polymerase chain reaction, shotgun metagenomic sequencing, or genome sequencing of cultured B breve. A total of 120 individual infants' stool samples were required for 80% power to detect a difference in strain transfer between groups. Rates of detection were compared using the Fisher exact test. RESULTS A total of 160 pregnant women with average age of 33.6 (3.9) years and mean body mass index of 24.3 (22.5-26.5) kg/m2, of whom 43% were nulliparous (n=58), were recruited from September 2016 to July 2019. Neonatal stool samples were obtained from 135 infants (65 in intervention and 70 in control group). The presence of the supplemented strain was detected through at least 2 methods (polymerase chain reaction and culture) in 2 infants in the intervention group (n=2/65; 3.1%) and none in the control group (n=0; 0%; P=.230). CONCLUSION Direct mother-to-infant strain transfer of B breve 702258 occurred, albeit infrequently. This study highlights the potential for maternal supplementation to introduce microbial strains into the infant microbiome.
Collapse
Affiliation(s)
- Rebecca L Moore
- UCD Perinatal Research Centre, UCD School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Ireland (Drs Moore, Killeen, Yelverton, Geraghty, and McAuliffe)
| | - Conor Feehily
- APC Microbiome Ireland, National University of Ireland, Cork, Ireland (Drs Feehily, Walsh, and O'Neill, Mses Nielsan, Lawton, and Sanchez-Gallardo, Mr Nori, and Drs Shanahan and Cotter); Teagasc Food Research Centre, Moorepark, Fermoy, Ireland (Drs Feehily and Walsh, Ms Lawton, Mr Nori, and Drs Sinderen and Cotter); Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom (Dr Feehily)
| | - Sarah Louise Killeen
- UCD Perinatal Research Centre, UCD School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Ireland (Drs Moore, Killeen, Yelverton, Geraghty, and McAuliffe)
| | - Cara A Yelverton
- UCD Perinatal Research Centre, UCD School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Ireland (Drs Moore, Killeen, Yelverton, Geraghty, and McAuliffe)
| | - Aisling A Geraghty
- UCD Perinatal Research Centre, UCD School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Ireland (Drs Moore, Killeen, Yelverton, Geraghty, and McAuliffe); Institute of Food and Health, UCD School of Agriculture and Food Science, University College Dublin, Dublin, Ireland (Dr Geraghty)
| | - Calum J Walsh
- APC Microbiome Ireland, National University of Ireland, Cork, Ireland (Drs Feehily, Walsh, and O'Neill, Mses Nielsan, Lawton, and Sanchez-Gallardo, Mr Nori, and Drs Shanahan and Cotter); Teagasc Food Research Centre, Moorepark, Fermoy, Ireland (Drs Feehily and Walsh, Ms Lawton, Mr Nori, and Drs Sinderen and Cotter)
| | - Ian J O'Neill
- APC Microbiome Ireland, National University of Ireland, Cork, Ireland (Drs Feehily, Walsh, and O'Neill, Mses Nielsan, Lawton, and Sanchez-Gallardo, Mr Nori, and Drs Shanahan and Cotter); School of Microbiology, University College Cork, Cork, Ireland (Dr O'Neill, Mses Nielsan and Sanchez-Gallardo, and Dr Van Sinderen)
| | - Ida Busch Nielsan
- APC Microbiome Ireland, National University of Ireland, Cork, Ireland (Drs Feehily, Walsh, and O'Neill, Mses Nielsan, Lawton, and Sanchez-Gallardo, Mr Nori, and Drs Shanahan and Cotter); School of Microbiology, University College Cork, Cork, Ireland (Dr O'Neill, Mses Nielsan and Sanchez-Gallardo, and Dr Van Sinderen)
| | - Elaine M Lawton
- APC Microbiome Ireland, National University of Ireland, Cork, Ireland (Drs Feehily, Walsh, and O'Neill, Mses Nielsan, Lawton, and Sanchez-Gallardo, Mr Nori, and Drs Shanahan and Cotter); Teagasc Food Research Centre, Moorepark, Fermoy, Ireland (Drs Feehily and Walsh, Ms Lawton, Mr Nori, and Drs Sinderen and Cotter)
| | - Rocio Sanchez-Gallardo
- APC Microbiome Ireland, National University of Ireland, Cork, Ireland (Drs Feehily, Walsh, and O'Neill, Mses Nielsan, Lawton, and Sanchez-Gallardo, Mr Nori, and Drs Shanahan and Cotter); School of Microbiology, University College Cork, Cork, Ireland (Dr O'Neill, Mses Nielsan and Sanchez-Gallardo, and Dr Van Sinderen)
| | - Sai Ravi Chandra Nori
- APC Microbiome Ireland, National University of Ireland, Cork, Ireland (Drs Feehily, Walsh, and O'Neill, Mses Nielsan, Lawton, and Sanchez-Gallardo, Mr Nori, and Drs Shanahan and Cotter); Teagasc Food Research Centre, Moorepark, Fermoy, Ireland (Drs Feehily and Walsh, Ms Lawton, Mr Nori, and Drs Sinderen and Cotter); Science Foundation Ireland Centre for Research Training in Genomics Data Science, School of Mathematics, Statistics and Applied Mathematics, National University of Ireland, Galway, Ireland (Mr Nori)
| | - Fergus Shanahan
- APC Microbiome Ireland, National University of Ireland, Cork, Ireland (Drs Feehily, Walsh, and O'Neill, Mses Nielsan, Lawton, and Sanchez-Gallardo, Mr Nori, and Drs Shanahan and Cotter); Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland (Dr Shanahan)
| | - Eileen F Murphy
- PrecisionBiotics Group Ltd, Novozymes, Cork, Ireland (Dr Murphy)
| | - Douwe Van Sinderen
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland (Drs Feehily and Walsh, Ms Lawton, Mr Nori, and Drs Sinderen and Cotter); School of Microbiology, University College Cork, Cork, Ireland (Dr O'Neill, Mses Nielsan and Sanchez-Gallardo, and Dr Van Sinderen)
| | - Paul D Cotter
- APC Microbiome Ireland, National University of Ireland, Cork, Ireland (Drs Feehily, Walsh, and O'Neill, Mses Nielsan, Lawton, and Sanchez-Gallardo, Mr Nori, and Drs Shanahan and Cotter); Teagasc Food Research Centre, Moorepark, Fermoy, Ireland (Drs Feehily and Walsh, Ms Lawton, Mr Nori, and Drs Sinderen and Cotter)
| | - Fionnuala M McAuliffe
- UCD Perinatal Research Centre, UCD School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Ireland (Drs Moore, Killeen, Yelverton, Geraghty, and McAuliffe).
| |
Collapse
|
6
|
Zhang Q, Chen B, Zhang J, Dong J, Ma J, Zhang Y, Jin K, Lu J. Effect of prebiotics, probiotics, synbiotics on depression: results from a meta-analysis. BMC Psychiatry 2023; 23:477. [PMID: 37386630 PMCID: PMC10308754 DOI: 10.1186/s12888-023-04963-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/15/2023] [Indexed: 07/01/2023] Open
Abstract
Accumulating studies have shown the effects of gut microbiota management tools in improving depression. We conducted a meta-analysis to evaluate the effects of prebiotics, probiotics, and synbiotics on patients with depression. We searched six databases up to July 2022. In total, 13 randomized controlled trials (RCTs) with 786 participants were included. The overall results demonstrated that patients who received prebiotics, probiotics or synbiotics had significantly improved symptoms of depression compared with those in the placebo group. However, subgroup analysis only confirmed the significant antidepressant effects of agents that contained probiotics. In addition, patients with mild or moderate depression could both benefit from the treatment. Studies with a lower proportion of females reported stronger effects for alleviating depressive symptoms. In conclusion, agents that manipulate gut microbiota might improve mild-to-moderate depression. It is necessary to further investigate the benefits of prebiotic, probiotic and synbiotic treatments relative to antidepressants and follow up with individuals over a longer time before these therapies are implemented in clinical practice.
Collapse
Affiliation(s)
- Qin Zhang
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bing Chen
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinghui Zhang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingyi Dong
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianglin Ma
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuyan Zhang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Kangyu Jin
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jing Lu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, China.
| |
Collapse
|
7
|
Omega-3-Rich Fish-Oil-Influenced Mouse Gut Microbiome Shaped by Intermittent Consumption of Beef. Curr Microbiol 2023; 80:119. [PMID: 36855004 DOI: 10.1007/s00284-023-03223-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 02/11/2023] [Indexed: 03/02/2023]
Abstract
Beef consumption can provide various amino acids, lipids, vitamins, and minerals; however, excessive intake causes metabolic disorders and increases the probability of obesity, atherosclerosis, and colorectal cancer. The intake of omega-3 fatty acids can ameliorate metabolic disorders by lowering blood glucose and triglyceride levels. In the present study, we investigated the effect of omega-3-rich fish oil on body performance and the gut microbiome in a beef-rich diet. Four-week-old C57BL/6 mice were distributed into four groups (chow diet [Chow], chow with beef diet [Beef], chow with omega-3 diet [Cw3], and chow with beef and omega-3 diet [Bw3]). We observed that body weight was unaltered between groups, and serum triglyceride levels were reduced in the omega-3 supplemented groups. The beta diversity indices, unweighted UniFrac distance (P = 0.001), and Jaccard distance (P = 0.001) showed statistically significant differences, and the principal coordinates analysis plot showed a clear separation between groups. In addition, the taxonomic comparison revealed that beef consumption increased numerous potentially pathogenic bacteria, including Escherichia-Shigella, Mucispirillum, Helicobacter, and Desulfovibrio, which were decreased following omega-3 supplementation. Metabolic comparison based on 16S rRNA revealed that energy and glucose metabolism were higher in omega-3 supplemented groups. Our findings suggest that the omega-3 supplementation under intermittent beef consumption contributes to changes in the gut microbiome and microbial metabolic pathways.
Collapse
|
8
|
Xie F, Xu HF, Zhang J, Liu XN, Kou BX, Cai MY, Wu J, Dong JL, Meng QH, Wang Y, Chen D, Zhang Y. Dysregulated hepatic lipid metabolism and gut microbiota associated with early-stage NAFLD in ASPP2-deficiency mice. Front Immunol 2022; 13:974872. [PMID: 36466835 PMCID: PMC9716097 DOI: 10.3389/fimmu.2022.974872] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/02/2022] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND Growing evidence indicates that lipid metabolism disorders and gut microbiota dysbiosis were related to the progression of non-alcoholic fatty liver disease (NAFLD). Apoptosis-stimulating p53 protein 2 (ASPP2) has been reported to protect against hepatocyte injury by regulating the lipid metabolism, but the mechanisms remain largely unknown. In this study, we investigate the effect of ASPP2 deficiency on NAFLD, lipid metabolism and gut microbiota using ASPP2 globally heterozygous knockout (ASPP2+/-) mice. METHODS ASPP2+/- Balb/c mice were fed with methionine and choline deficient diet for 3, 10 and 40 day to induce an early and later-stage of NAFLD, respectively. Fresh fecal samples were collected and followed by 16S rRNA sequencing. HPLC-MRM relative quantification analysis was used to identify changes in hepatic lipid profiles. The expression level of innate immunity-, lipid metabolism- and intestinal permeability-related genes were determined. A spearman's rank correlation analysis was performed to identify possible correlation between hepatic medium and long-chain fatty acid and gut microbiota in ASPP2-deficiency mice. RESULTS Compared with the WT control, ASPP2-deficiency mice developed moderate steatosis at day 10 and severe steatosis at day 40. The levels of hepatic long chain omega-3 fatty acid, eicosapentaenoic (EPA, 20:5 n-3) and docosahexaenoic (DHA, 22:6 n-3), were decreased at day 10 and increased at day 40 in ASPP+/- mice. Fecal microbiota analysis showed significantly increased alpha and beta diversity, as well as the composition of gut microbiota at the phylum, class, order, family, genus, species levels in ASPP2+/- mice. Moreover, ASPP-deficiency mice exhibited impaired intestinal barrier function, reduced expression of genes associated with chemical barrier (REG3B, REG3G, Lysozyme and IAP), and increased expression of innate immune components (TLR4 and TLR2). Furthermore, correlation analysis between gut microbiota and fatty acids revealed that EPA was significantly negatively correlated with Bifidobacterium family. CONCLUSION Our findings suggested that ASPP2-deficiency promotes the progression of NAFLD, alterations in fatty acid metabolism and gut microbiota dysbiosis. The long chain fatty acid EPA was significantly negatively correlated with Bifidobacterial abundance, which is a specific feature of NAFLD in ASPP2-deficiency mice. Totally, the results provide evidence for a mechanism of ASPP2 on dysregulation of fatty acid metabolism and gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Fang Xie
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing Institute of Hepatology, Beijing, China
| | - Hang-fei Xu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Department of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Jing Zhang
- Department of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xiao-ni Liu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing Institute of Hepatology, Beijing, China
| | - Bu-xin Kou
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing Institute of Hepatology, Beijing, China
| | - Meng-yin Cai
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing Institute of Hepatology, Beijing, China
| | - Jing Wu
- Department of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Jin-ling Dong
- Department of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Qing-hua Meng
- Department of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Dexi Chen
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing Institute of Hepatology, Beijing, China
| | - Yang Zhang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Engineering Research Center for Precision Medicine and Transformation of Hepatitis and Liver Cancer, Beijing Institute of Hepatology, Beijing, China
| |
Collapse
|
9
|
Mueed A, Shibli S, Korma SA, Madjirebaye P, Esatbeyoglu T, Deng Z. Flaxseed Bioactive Compounds: Chemical Composition, Functional Properties, Food Applications and Health Benefits-Related Gut Microbes. Foods 2022; 11:3307. [PMCID: PMC9602266 DOI: 10.3390/foods11203307] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Flaxseed (Linum usitatissimum L.) has gained worldwide recognition as a health food because of its abundance in diverse nutrients and bioactive compounds such as oil, fatty acids, proteins, peptides, fiber, lignans, carbohydrates, mucilage, and micronutrients. These constituents attribute a multitude of beneficial properties to flaxseed that makes its use possible in various applications, such as nutraceuticals, food products, cosmetics, and biomaterials. The importance of these flaxseed components has also increased in modern times because of the newer trend among consumers of greater reliance on a plant-based diet for fulfilling their nutritional requirements, which is perceived to be hypoallergenic, more environmentally friendly, sustainable, and humane. The role of flaxseed substances in the maintenance of a healthy composition of the gut microbiome, prevention, and management of multiple diseases has recently been elucidated in various studies, which have highlighted its importance further as a powerful nutritional remedy. Many articles previously reported the nutritive and health benefits of flaxseed, but no review paper has been published reporting the use of individual flaxseed components in a manner to improve the techno-functional properties of foods. This review summarizes almost all possible applications of flaxseed ingredients in food products from an extensive online literature survey; moreover, it also outlines the way forward to make this utilization even better.
Collapse
Affiliation(s)
- Abdul Mueed
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Sahar Shibli
- National Agriculture Research Center, Food Science Research Institute, Islamabad 44000, Pakistan
| | - Sameh A. Korma
- Department of Food Science, Faculty of Agriculture, Zagazig University, Zagazig 44519, Egypt
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Philippe Madjirebaye
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Tuba Esatbeyoglu
- Department of Food Development and Food Quality, Institute of Food Science and Human Nutrition, Gottfried Wilhelm Leibniz University Hannover, Am Kleinen Felde 30, 30167 Hannover, Germany
- Correspondence: (T.E.); (Z.D.); Tel.: +49-5117625589 (T.E.); +86-791-88304402 (Z.D.)
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
- Correspondence: (T.E.); (Z.D.); Tel.: +49-5117625589 (T.E.); +86-791-88304402 (Z.D.)
| |
Collapse
|
10
|
Zhang F, Zuo T, Wan Y, Xu Z, Cheung C, Li AY, Zhu W, Tang W, Chan PK, Chan FK, Ng SC. Multi-omic analyses identify mucosa bacteria and fecal metabolites associated with weight loss after fecal microbiota transplantation. Innovation (N Y) 2022; 3:100304. [PMID: 36091491 PMCID: PMC9460156 DOI: 10.1016/j.xinn.2022.100304] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/13/2022] [Indexed: 11/19/2022] Open
Abstract
Fecal microbiota transplantation (FMT) has shown promising results in animal models of obesity, while results in human studies are inconsistent. We aimed to determine factors associated with weight loss after FMT in nine obese subjects using serial multi-omics analysis of the fecal and mucosal microbiome. The mucosal microbiome, fecal microbiome, and fecal metabolome showed individual clustering in each subject after FMT. The colonic microbiome in patients showed more marked variance after FMT compared with the duodenal microbiome, characterized by an increased relative abundance of Bacteroides. Subjects who lost weight after FMT sustained enrichment of Bifidobacterium bifidum and Alistipes onderdonkii in the duodenal, colonic mucosal, and fecal microbiome and increased levels of phosphopantothenate biosynthesis and fecal metabolite eicosapentaenoic acid (EPA), compared with those without weight loss. Fecal levels of amino acid metabolism-associated were positively correlated with the fecal abundance of B. bifidum, and fatty acid metabolism-associated metabolites showed positive correlations with A. onderdonkii. We report for the first time the individualized response of fecal and mucosa microbiome to FMT in obese subjects and highlight that FMT is less capable of shaping the small intestine microbiota. These findings contribute to personalized microbe-based therapies for obesity.
Collapse
Affiliation(s)
- Fen Zhang
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
| | - Tao Zuo
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
| | - Yating Wan
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
| | - Zhilu Xu
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
| | - Chunpan Cheung
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
| | - Amy Y. Li
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Wenyi Zhu
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
| | - Whitney Tang
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
| | - Paul K.S. Chan
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong 999077, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Francis K.L. Chan
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Siew C. Ng
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Microbiota I-Center (MagIC), Hong Kong 999077, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
- Corresponding author
| |
Collapse
|
11
|
Research progress on conjugated linoleic acid bio-conversion in Bifidobacterium. Int J Food Microbiol 2022; 369:109593. [DOI: 10.1016/j.ijfoodmicro.2022.109593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/13/2022] [Accepted: 02/20/2022] [Indexed: 11/18/2022]
|
12
|
YONESHIGE R, KUSUDA E, ANDO T. Effect of probiotics on blood fatty acid metabolism during the late middle stage of fattening period in Japanese Black cattle. J Vet Med Sci 2022; 84:319-324. [PMID: 35110457 PMCID: PMC8983281 DOI: 10.1292/jvms.21-0354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study was conducted to investigate the effects of probiotics administration on fatty acid metabolism in Japanese Black cattle as per changes in blood fatty acid concentrations and
blood biochemical tests. Eighteen clinically healthy Japanese Black female fattening cattle bred on the same fattening farm were randomly classified into the probiotics administration group
(n=9) or the control group (n=9). In the probiotics administration group, 50 g of probiotics were started per animal per day at the age of 18 months, and the administration period was 2
months from the start date of the study. Blood was collected twice before starting the probiotics administration and at 2 months after starting the probiotics administration. In the
probiotics administration group, palmitic, linoleic, arachidonic and α-linolenic acid tended to be higher at the end of the administration compared with those before probiotics
administration. Additionally, as a result of multiple comparison test, monounsaturated fatty acids at Post was significantly higher, and the ω6 / ω3 ratio was significantly lower than in the
control group. Vitamin A, E and albumin were significantly higher at the end of the administration than in the control group. In this study that administering probiotics to Japanese Black
cattle in the late middle stage of fattening period did not have a significant effect on fatty acid metabolism during feed digestion and absorption, but suggested that may alter some blood
fatty acids concentrations.
Collapse
Affiliation(s)
| | | | - Takaaki ANDO
- The United Graduate School of Veterinary Science, Yamaguchi University
| |
Collapse
|
13
|
Guz M, Jeleniewicz W, Malm A, Korona-Glowniak I. A Crosstalk between Diet, Microbiome and microRNA in Epigenetic Regulation of Colorectal Cancer. Nutrients 2021; 13:2428. [PMID: 34371938 PMCID: PMC8308570 DOI: 10.3390/nu13072428] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 02/07/2023] Open
Abstract
A still growing interest between human nutrition in relation to health and disease states can be observed. Dietary components shape the composition of microbiota colonizing our gastrointestinal tract which play a vital role in maintaining human health. There is a strong evidence that diet, gut microbiota and their metabolites significantly influence our epigenome, particularly through the modulation of microRNAs. These group of small non-coding RNAs maintain cellular homeostasis, however any changes leading to impaired expression of miRNAs contribute to the development of different pathologies, including neoplastic diseases. Imbalance of intestinal microbiota due to diet is primary associated with the development of colorectal cancer as well as other types of cancers. In the present work we summarize current knowledge with particular emphasis on diet-microbiota-miRNAs axis and its relation to the development of colorectal cancer.
Collapse
Affiliation(s)
- Małgorzata Guz
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Witold Jeleniewicz
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Anna Malm
- Department of Pharmaceutical Microbiology, Medical University of Lublin, 20-093 Lublin, Poland; (A.M.); (I.K.-G.)
| | - Izabela Korona-Glowniak
- Department of Pharmaceutical Microbiology, Medical University of Lublin, 20-093 Lublin, Poland; (A.M.); (I.K.-G.)
| |
Collapse
|
14
|
Vlasov AA, Salikova SP, Golovkin NV, Grinevich VB. Intestinal Microbial-tissue Complex and Chronic Heart Failure (part 1): Pathogenesis. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2021. [DOI: 10.20996/1819-6446-2021-06-12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Antigenic and metabolic integration of the intestinal microbiota into the homeostasis of the human body is a factor that claims to play a key role in the pathogenesis of cardiovascular diseases. It acquires special significance against the background of the decrease in blood circulation and congestion in the digestive system during chronic heart failure. Aim of the review is analysis and synthesis of studies results on the role of intestinal microbiocenosis in the pathogenesis of heart remodeling and chronic heart failure. The search for articles was conducted in databases eLIBRARY.RU and Medline for the key terms "gut microbiota (microbiome, microbiocenosis)", "dysbiosis (dysbacteriosis)", "excessive bacterial growth syndrome", "lipopolysaccharide (endotoxin)", "trimethylamine-N-oxide" in combination with the terms "heart failure", "myocardial remodeling", "myocardium" in Russian and English, respectively. We selected articles containing the results of clinical and experimental studies published from 1995 to 2020. Review articles were considered only on the subject of the cited original publications. Most researchers have established the relationship between chronic heart failure and dysfunction and changes in the qualitative and quantitative composition of intestinal microbiocenosis. As negative changes, it is customary to note the proliferation of gram-negative opportunistic bacteria with concomitant endotoxinemia and a decrease in the pool of commensal microbiota. The available data suggest that the participation of the intestinal microbial-tissue complex in the pathogenesis of chronic heart failure and heart remodeling is realized through the activation of a local and then systemic inflammatory response, accompanied by cardiodepressive action of pro-inflammatory cytokines and universal proliferation factors, an imbalance of matrix metalloproteinases and their inhibitors, the initiation of apoptosis, fibrosis, and loss of contractile myocardium. Besides, a decrease in the production of short-chain and polyunsaturated fatty acids and vitamins by the commensal microbiota may be associated with changes in the electrical properties of cardiomyocyte membranes, a decrease in the systolic function of the left ventricle of the heart, and an increase in the risk of sudden cardiac death. It's also shown that the direct cardiotoxic effect of microbial molecules (lipopolysaccharides, peptidoglycans, trimethylamine-N-oxide, etc.), which interact with the receptors of cardiomyocytes and microenvironment cells, can cause the development of myocardial remodeling and its dysfunction. Recent studies have established mechanisms of myocardial remodeling mediated by microbial molecules, which may be associated with new strategies for the treatment and prevention of heart failure.
Collapse
|
15
|
Associations among Dietary Omega-3 Polyunsaturated Fatty Acids, the Gut Microbiota, and Intestinal Immunity. Mediators Inflamm 2021; 2021:8879227. [PMID: 33488295 PMCID: PMC7801035 DOI: 10.1155/2021/8879227] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/02/2020] [Accepted: 12/18/2020] [Indexed: 02/08/2023] Open
Abstract
Omega-3 polyunsaturated fatty acids (omega-3 PUFAs), which are essential fatty acids that humans should obtain from diet, have potential benefits for human health. In addition to altering the structure and function of cell membranes, omega-3 PUFAs (docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), alpha-linolenic acid (ALA), and docosapentaenoic acid (DPA)) exert different effects on intestinal immune tolerance and gut microbiota maintenance. Firstly, we review the effect of omega-3 PUFAs on gut microbiota. And the effects of omega-3 PUFAs on intestinal immunity and inflammation were described. Furthermore, the important roles of omega-3 PUFAs in maintaining the balance between gut immunity and the gut microbiota were discussed. Additional factors, such as obesity and diseases (NAFLD, gastrointestinal malignancies or cancer, bacterial and viral infections), which are associated with variability in omega-3 PUFA metabolism, can influence omega-3 PUFAs–microbiome–immune system interactions in the intestinal tract and also play roles in regulating gut immunity. This review identifies several pathways by which the microbiota modulates the gut immune system through omega-3 PUFAs. Omega-3 supplementation can be targeted to specific pathways to prevent and alleviate intestinal diseases, which may help researchers identify innovative diagnostic methods.
Collapse
|
16
|
Wu Q, Li Q, Zhang X, Ntim M, Wu X, Li M, Wang L, Zhao J, Li S. Treatment with Bifidobacteria can suppress Aβ accumulation and neuroinflammation in APP/PS1 mice. PeerJ 2020; 8:e10262. [PMID: 33194428 PMCID: PMC7602682 DOI: 10.7717/peerj.10262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 10/07/2020] [Indexed: 12/21/2022] Open
Abstract
Background Alzheimer’s disease (AD), being a complex disorder, is affected either by genetic or environmental factors or both. It is observed that there is an excessive accumulation of amyloid β (Aβ) in the extracellular space of the brain. AD is the first neurodegenerative disease in the elderly, and so far there is no effective treatment. In recent years, many studies have reported that Alzheimer’s disease has a relationship with gut microflora, indicating that regulating gut microbiota could offer therapeutic intervention for AD. This study explored the effect Bifidobacteria has in averting AD. Methods WT and APP/PS1 mice were used for the experiments. The mice were randomly assigned to four groups: WT group, WT + Bi group, AD group (APP/PS1 mouse) and AD + Bi group (Bifidobacteria-treated APP/PS1 mouse). Treatment with Bifidobacteria lasted for 6 months and mice were prepared for immunohistochemistry, immunofluorescence, Thioflavin S staining, Western blotting, PCR and Elisa quantitative assay. Results The results show that after 6 months of treatment with Bifidobacteria signiis to be lesficantly reduces Aβ deposition in cortex and hippocampus of AD mice. The level of insoluble Aβ in the hippocampus and cortex of AD+Bi mice was decreased compared with AD mice. Meanwhile, a significant decrease in the level of soluble Aβ in the cortex of AD+Bi mice but not in the hippocampus was observed. The activation of microglia and the release of inflammatory factors were also determined in this study. From the results, Bifidobacteria inhibited microglial activation and reduced IL-1β, TNF-α, IL-4, IL-6 and INF-γ release. Altogether, these results implied that Bifidobacteria can alleviate the pathological changes of AD through various effects.
Collapse
Affiliation(s)
- Qiong Wu
- Liaoning Provincial Key Laboratory of Cerebral Diseases in Department of Physiology, Dalian Medical University, Dalian, China
| | - Qifa Li
- Functional Laboratory, Dalian Medical University, Dalian, China
| | - Xuan Zhang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Michael Ntim
- Liaoning Provincial Key Laboratory of Cerebral Diseases in Department of Physiology, Dalian Medical University, Dalian, China
| | - Xuefei Wu
- Liaoning Provincial Key Laboratory of Cerebral Diseases in Department of Physiology, Dalian Medical University, Dalian, China
| | - Ming Li
- Department of Microecology, Dalian Medical University, Dalian, China
| | - Li Wang
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Jie Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Shao Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases in Department of Physiology, Dalian Medical University, Dalian, China
| |
Collapse
|
17
|
Xin J, Zeng D, Wang H, Sun N, Khalique A, Zhao Y, Wu L, Pan K, Jing B, Ni X. Lactobacillus johnsonii BS15 improves intestinal environment against fluoride-induced memory impairment in mice-a study based on the gut-brain axis hypothesis. PeerJ 2020; 8:e10125. [PMID: 33083147 PMCID: PMC7547597 DOI: 10.7717/peerj.10125] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 09/17/2020] [Indexed: 01/09/2023] Open
Abstract
Background Excessive fluoride can lead to chronic neurodegeneration characterized by neuron and myelin loss and memory dysfunction. The gut–brain axis hypothesis suggests that gut microbiota plays a crucial role in regulating brain function. Thus, using probiotics to adjust the gut microenvironment may be a potential therapy for mental diseases. Methods Mice in the prob group were administrated with Lactobacillus johnsonii BS15 for 28 days prior to and throughout a 70-day exposure to sodium fluoride. The drinking water of all groups (F and prob groups) except the control group were replaced by high-fluoride water (100 mg NaF/L) on day 28. Animals in each group were divided into two subsets: one underwent behavioral test, and the other was sacrificed for sampling. The mRNA expression level and protein content related to inflammatory reaction in the ileum and hippocampus were respectively detected by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA). The mRNA expression levels of proteins related to myelin structure, apoptosis, and memory in the hippocampus and tight junction proteins in the ileum were determined by RT-qPCR and/or immunohistochemistry. Gut permeability markers (D-lactate and diamine oxidase (DAO)) in the serum were also examined by ELISA. Results The results showed that fluoride exposure induced a lower spontaneous exploration (P < 0.05) in T-maze test, which indicated an impairment of memory. Spontaneous exploration of BS15-treated mice was significantly higher (P < 0.05) than that in F group. Fluoride reduced (P < 0.05) levels of myelin structural protein (proteolipid protein) and neurogenesis-associated proteins (brain-derived neurotrophic factor and cAMP/Ca2+ responsive element-binding protein), induced disordered inflammatory cytokines (TNF-α, IFN-γ, and IL-6; P < 0.05), increased pro-apoptotic genes (caspase-3; P < 0.05), and decreased anti-apoptotic genes (Bcl-2; P < 0.05) in the hippocampus, of which the influences were reversed by BS15. BS15 treatment exerted significant preventive effects on reversing the gut inflammation induced by excessive fluoride intake by reducing (P < 0.05) the levels of pro-inflammatory cytokines (tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ)) and remarkably increasing (P < 0.05) the level of anti-inflammatory cytokines (IL-10). Moreover, the serum DAO activity and D-lactate concentration significantly increased by fluoride were also reduced (P < 0.05) by BS15. This result indicated the profitable effect of BS15 on gut permeability. Conclusion L. johnsonii BS15 intake could benefit the neuroinflammation and demyelination in the hippocampus by improving the gut environment and ameliorating fluorine-induced memory dysfunction.
Collapse
Affiliation(s)
- Jinge Xin
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Dong Zeng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hesong Wang
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ning Sun
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Abdul Khalique
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Ying Zhao
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Liqian Wu
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Kangcheng Pan
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Bo Jing
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xueqin Ni
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
18
|
Hughes RL, Arnold CD, Young RR, Ashorn P, Maleta K, Fan YM, Ashorn U, Chaima D, Malamba-Banda C, Kable ME, Dewey KG. Infant gut microbiota characteristics generally do not modify effects of lipid-based nutrient supplementation on growth or inflammation: secondary analysis of a randomized controlled trial in Malawi. Sci Rep 2020; 10:14861. [PMID: 32908192 PMCID: PMC7481312 DOI: 10.1038/s41598-020-71922-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 08/21/2020] [Indexed: 12/22/2022] Open
Abstract
An unhealthy gut microbial community may act as a barrier to improvement in growth and health outcomes in response to nutritional interventions. The objective of this analysis was to determine whether the infant microbiota modified the effects of a randomized controlled trial of lipid-based nutrient supplements (LNS) in Malawi on growth and inflammation at 12 and 18 months, respectively. We characterized baseline microbiota composition of fecal samples at 6 months of age (n = 506, prior to infant supplementation, which extended to 18 months) using 16S rRNA gene sequencing of the V4 region. Features of the gut microbiota previously identified as being involved in fatty acid or micronutrient metabolism or in outcomes relating to growth and inflammation, especially in children, were investigated. Prior to correction for multiple hypothesis testing, the effects of LNS on growth appeared to be modified by Clostridium (p-for-interaction = 0.02), Ruminococcus (p-for-interaction = 0.007), and Firmicutes (p-for-interaction = 0.04) and effects on inflammation appeared to be modified by Faecalibacterium (p-for-interaction = 0.03) and Streptococcus (p-for-interaction = 0.004). However, after correction for multiple hypothesis testing these findings were not statistically significant, suggesting that the gut microbiota did not alter the effect of LNS on infant growth and inflammation in this cohort.
Collapse
Affiliation(s)
- Riley L Hughes
- Department of Nutrition, University of California, Davis, CA, USA
| | - Charles D Arnold
- Department of Nutrition, University of California, Davis, CA, USA
| | - Rebecca R Young
- Department of Nutrition, University of California, Davis, CA, USA
| | - Per Ashorn
- Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Pediatrics, Tampere University Hospital, Tampere, Finland
| | - Ken Maleta
- College of Medicine, University of Malawi, Blantyre 3, Malawi
| | - Yue-Mei Fan
- Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ulla Ashorn
- Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - David Chaima
- School of Public Health and Family Medicine, University of Malawi College of Medicine, Blantyre, Malawi
| | - Chikondi Malamba-Banda
- School of Public Health and Family Medicine, University of Malawi College of Medicine, Blantyre, Malawi
| | - Mary E Kable
- Immunity and Disease Prevention, Western Human Nutrition Research Center, Agricultural Research Service, USDA, Davis, CA, USA
| | - Kathryn G Dewey
- Department of Nutrition, University of California, Davis, CA, USA.
| |
Collapse
|
19
|
Salazar N, Neyrinck AM, Bindels LB, Druart C, Ruas-Madiedo P, Cani PD, de Los Reyes-Gavilán CG, Delzenne NM. Functional Effects of EPS-Producing Bifidobacterium Administration on Energy Metabolic Alterations of Diet-Induced Obese Mice. Front Microbiol 2019; 10:1809. [PMID: 31440225 PMCID: PMC6693475 DOI: 10.3389/fmicb.2019.01809] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/23/2019] [Indexed: 01/14/2023] Open
Abstract
Obesity has been recognized by the World Health Organization as a global epidemic. The gut microbiota is considered as a factor involved in the regulation of numerous metabolic pathways by impacting several functions of the host. It has been suggested that probiotics can modulate host gene expression and metabolism, and thereby positively influence host adipose tissue development and obesity related-metabolic disorders. The aim of the present work was to evaluate the effect of an exopolysaccharide (EPS)-producing Bifidobacterium strain on host glucose and lipid metabolism and the gut microbial composition in a short-term diet-induced obesity (DIO) in mice. C57BL/6J male mice were randomly divided into three groups: a control group that received control standard diet, a group fed a high-fat diet (HF), and a group fed HF supplemented with Bifidobacterium animalis IPLA R1. Fasting serum insulin as well as triglycerides accumulation in the liver were significantly reduced in the group receiving B. animalis IPLA R1. The treatment with the EPS-producing B. animalis IPLA R1 tended to down-regulate the expression of host genes involved in the hepatic synthesis of fatty acids which was concomitant with an upregulation in the expression of genes related with fatty acid oxidation. B. animalis IPLA R1 not only promoted the increase of Bifidobacterium but also the levels of Bacteroides-Prevotella. Our data indicate that the EPS-producing Bifidobacterium IPLA R1 strain may have beneficial effects in metabolic disorders associated with obesity, by modulating the gut microbiota composition and promoting changes in lipids metabolism and glucose homeostasis.
Collapse
Affiliation(s)
- Nuria Salazar
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), Brussels, Belgium.,Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Asturias, Spain.,Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Céline Druart
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Patricia Ruas-Madiedo
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Asturias, Spain
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), Brussels, Belgium.,Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Clara G de Los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Asturias, Spain.,Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
20
|
Effect of Pufa Substrates on Fatty Acid Profile of Bifidobacterium breve Ncimb 702258 and CLA/CLNA Production in Commercial Semi-Skimmed Milk. Sci Rep 2018; 8:15591. [PMID: 30349012 PMCID: PMC6197199 DOI: 10.1038/s41598-018-33970-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/28/2018] [Indexed: 12/16/2022] Open
Abstract
Current research on lipids is highlighting their relevant role in metabolic/signaling pathways. Conjugated fatty acids (CFA), namely isomers of linoleic and linolenic acid (i.e. CLA and CLNA, respectively) can positively modulate inflammation processes and energy metabolism, promoting anti-carcinogenic and antioxidant effects, improved lipid profiles and insulin resistance, among others. Bioactive doses have been indicated to be above 1 g/d, yet these cannot be achieved through a moderate intake (i.e. 1-2 servings) of natural sources, and certain CLA-containing products have limited commercial availability. Such handicaps have fueled research interest in finding alternative fortification strategies. In recent years, screening of dairy products for CFA-producing bacteria has attracted much attention and has led to the identification of some promising strains, including Bifidobacterium breve NCIMB 702258. This strain has shown interesting producing capabilities in model systems as well as positive modulation of lipid metabolism activities in animal studies. Accordingly, the aim of this research work was to assay B. breve NCIMB 702258 in semi-skimmed milk to produce a probiotic fermented dairy product enriched in bioactive CLA and CLNA. The effect of substrates (LA, α-LNA and γ-LNA) on growth performance and membrane fatty acids profile was also studied, as these potential modifications have been associated to stress response. When tested in cys-MRS culture medium, LA, α-LNA and γ-LNA impaired the fatty acid synthesis by B. breve since membrane concentrations for stearic and oleic acids decreased. Variations in the C18:1 c11 and lactobacillic acid concentrations, may suggest that these substrates are also affecting the membrane fluidity. Bifidobacterium breve CFA production capacity was first assessed in cys-MRS with LA, α-LNA, γ-LNA or all substrates together at 0.5 mg/mL each. This strain did not produce CFA from γ-LNA, but converted 31.12% of LA and 68.20% of α-LNA into CLA and CLNA, respectively, after incubation for 24 h at 37 °C. In a second phase, B. breve was inoculated in a commercial semi-skimmed milk with LA, α-LNA or both at 0.5 mg/mL each. Bifidobacterium breve revealed a limited capacity to synthesize CLA isomers, but was able to produce 0.062-0.115 mg/mL CLNA after 24 h at 37 °C. However, organoleptic problems were reported which need to be addressed in future studies. These results show that although CFA were produced at too low concentrations to be able to achieve solely the bioactive dose in one daily portion size, fermented dairy products are a suitable vector to deliver B. breve NCIMB 702258.
Collapse
|
21
|
Gerhauser C. Impact of dietary gut microbial metabolites on the epigenome. Philos Trans R Soc Lond B Biol Sci 2018; 373:20170359. [PMID: 29685968 PMCID: PMC5915727 DOI: 10.1098/rstb.2017.0359] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2018] [Indexed: 12/18/2022] Open
Abstract
Within the past decade, epigenetic mechanisms and their modulation by natural products have gained increasing interest. Dietary bioactive compounds from various sources, including green tea, soya, fruit and berries, cruciferous vegetables, whole grain foods, fish and others, have been shown to target enzymes involved in epigenetic gene regulation, including DNA methyltransferases, histone acetyltransferases, deacetylases and demethylases in vitro and in cell culture. Also, many dietary agents were shown to alter miRNA expression. In vivo studies in animal models and humans are still limited. Recent research has indicated that the gut microbiota and gut microbial metabolites might be important mediators of diet-epigenome interactions. Inter-individual differences in the gut microbiome might affect release, metabolism and bioavailability of dietary agents and explain variability in response to intervention in human studies. Only a few microbial metabolites, including folate, phenolic acids, S-(-)equol, urolithins, isothiocyanates, and short- and long-chain fatty acids have been tested with respect to their potential to influence epigenetic mechanisms. Considering that a complex mixture of intermediary and microbial metabolites is present in human circulation, a more systematic interdisciplinary investigation of nutri-epigenetic activities and their impact on human health is called for.This article is part of a discussion meeting issue 'Frontiers in epigenetic chemical biology'.
Collapse
Affiliation(s)
- Clarissa Gerhauser
- Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
22
|
Sundman MH, Chen NK, Subbian V, Chou YH. The bidirectional gut-brain-microbiota axis as a potential nexus between traumatic brain injury, inflammation, and disease. Brain Behav Immun 2017; 66:31-44. [PMID: 28526435 DOI: 10.1016/j.bbi.2017.05.009] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/25/2017] [Accepted: 05/10/2017] [Indexed: 02/06/2023] Open
Abstract
As head injuries and their sequelae have become an increasingly salient matter of public health, experts in the field have made great progress elucidating the biological processes occurring within the brain at the moment of injury and throughout the recovery thereafter. Given the extraordinary rate at which our collective knowledge of neurotrauma has grown, new insights may be revealed by examining the existing literature across disciplines with a new perspective. This article will aim to expand the scope of this rapidly evolving field of research beyond the confines of the central nervous system (CNS). Specifically, we will examine the extent to which the bidirectional influence of the gut-brain axis modulates the complex biological processes occurring at the time of traumatic brain injury (TBI) and over the days, months, and years that follow. In addition to local enteric signals originating in the gut, it is well accepted that gastrointestinal (GI) physiology is highly regulated by innervation from the CNS. Conversely, emerging data suggests that the function and health of the CNS is modulated by the interaction between 1) neurotransmitters, immune signaling, hormones, and neuropeptides produced in the gut, 2) the composition of the gut microbiota, and 3) integrity of the intestinal wall serving as a barrier to the external environment. Specific to TBI, existing pre-clinical data indicates that head injuries can cause structural and functional damage to the GI tract, but research directly investigating the neuronal consequences of this intestinal damage is lacking. Despite this void, the proposed mechanisms emanating from a damaged gut are closely implicated in the inflammatory processes known to promote neuropathology in the brain following TBI, which suggests the gut-brain axis may be a therapeutic target to reduce the risk of Chronic Traumatic Encephalopathy and other neurodegenerative diseases following TBI. To better appreciate how various peripheral influences are implicated in the health of the CNS following TBI, this paper will also review the secondary biological injury mechanisms and the dynamic pathophysiological response to neurotrauma. Together, this review article will attempt to connect the dots to reveal novel insights into the bidirectional influence of the gut-brain axis and propose a conceptual model relevant to the recovery from TBI and subsequent risk for future neurological conditions.
Collapse
Affiliation(s)
- Mark H Sundman
- Department of Psychology, University of Arizona, Tucson, AZ, USA.
| | - Nan-Kuei Chen
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, USA
| | - Vignesh Subbian
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, USA; Department of Systems and Industrial Engineering, University of Arizona, Tucson, AZ, USA
| | - Ying-Hui Chou
- Department of Psychology, University of Arizona, Tucson, AZ, USA; Cognitive Science Program, University of Arizona, Tucson, AZ, USA; Arizona Center on Aging, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
23
|
Patterson E, Wall R, Lisai S, Ross RP, Dinan TG, Cryan JF, Fitzgerald GF, Banni S, Quigley EM, Shanahan F, Stanton C. Bifidobacterium breve with α-linolenic acid alters the composition, distribution and transcription factor activity associated with metabolism and absorption of fat. Sci Rep 2017; 7:43300. [PMID: 28265110 PMCID: PMC5339701 DOI: 10.1038/srep43300] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 01/12/2017] [Indexed: 01/16/2023] Open
Abstract
This study focused on the mechanisms that fatty acid conjugating strains - Bifidobacterium breve NCIMB 702258 and Bifidobacterium breve DPC 6330 - influence lipid metabolism when ingested with α-linolenic acid (ALA) enriched diet. Four groups of BALB/c mice received ALA enriched diet (3% (w/w)) either alone or in combination with B. breve NCIMB 702258 or B. breve DPC 6330 (109 CFU/day) or unsupplemented control diet for six weeks. The overall n-3 PUFA score was increased in all groups receiving the ALA enriched diet. Hepatic peroxisomal beta oxidation increased following supplementation of the ALA enriched diet with B. breve (P < 0.05) and so the ability of the strains to produce c9t11 conjugated linoleic acid (CLA) was identified in adipose tissue. Furthermore, a strain specific effect of B. breve NCIMB 702258 was found on the endocannabinoid system (ECS). Liver triglycerides (TAG) were reduced following ALA supplementation, compared with unsupplemented controls (P < 0.01) while intervention with B. breve further reduced liver TAG (P < 0.01), compared with the ALA enriched control. These data indicate that the interactions of the gut microbiota with fatty acid metabolism directly affect host health by modulating n-3 PUFA score and the ECS.
Collapse
Affiliation(s)
- Elaine Patterson
- APC Microbiome Institute, Biosciences Building, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Food Biosciences Department, Moorepark, Fermoy, Cork, Ireland
| | - Rebecca Wall
- APC Microbiome Institute, Biosciences Building, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Food Biosciences Department, Moorepark, Fermoy, Cork, Ireland
| | - Sara Lisai
- Department of Biomedical Sciences, University of Cagliari, Monserrato, CA 09042, Italy
| | - R Paul Ross
- APC Microbiome Institute, Biosciences Building, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Institute, Biosciences Building, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, Biosciences Building, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Gerald F Fitzgerald
- APC Microbiome Institute, Biosciences Building, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | - Sebastiano Banni
- Department of Biomedical Sciences, University of Cagliari, Monserrato, CA 09042, Italy
| | - Eamonn M Quigley
- APC Microbiome Institute, Biosciences Building, University College Cork, Cork, Ireland
| | - Fergus Shanahan
- APC Microbiome Institute, Biosciences Building, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Institute, Biosciences Building, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Food Biosciences Department, Moorepark, Fermoy, Cork, Ireland
| |
Collapse
|
24
|
Joffre C, Dinel AL, Aubert A, Fressange-Mazda C, Le Ruyet P, Layé S. Impact of Lactobacillus fermentum and dairy lipids in the maternal diet on the fatty acid composition of pups' brain and peripheral tissues. Prostaglandins Leukot Essent Fatty Acids 2016; 115:24-34. [PMID: 27914510 DOI: 10.1016/j.plefa.2016.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/05/2016] [Accepted: 10/05/2016] [Indexed: 10/20/2022]
Abstract
The aim of the study was to determine the effect of maternal diets administered since day 1 of gestation and containing dairy lipids or vegetable oils differing in the supply of n-3 polyunsaturated fatty acids (n-3 PUFAs) (equilibrated or deficient) and of Lactobacillus fermentum (L. fermentum) on the docosahexaenoic acid (DHA) accretion in the pups at postnatal day 14 in the prefrontal cortex (PFC) and hippocampus (HC) for brain structures and in the liver and adipose tissue for peripheral tissues. Maternal milk fatty acid composition was also assessed by analyzing the fatty acid composition of the gastric content of the pups. DHA was higher in mice supplemented with L. fermentum than in mice in the deficient group in HC and PFC and also in liver and adipose tissue. This increase could be linked to the slight but significant increase in C18:3n-3 in the maternal milk. This proportion was comparable in the dairy lipid group for which the brain DHA level was the highest. L. fermentum may have a key role in the protection of the brain during the perinatal period via the neuronal accretion of n-3 PUFAs, especially during n-3 PUFA deficiency.
Collapse
Affiliation(s)
- C Joffre
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France.
| | - A L Dinel
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Nutribrain Research and Technology transfer of Nutrition et Neurobiologie Intégrée, 33076 Bordeaux, France
| | - A Aubert
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | | | - P Le Ruyet
- Lactalis Recherche et Développement, Retiers, France
| | - S Layé
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| |
Collapse
|
25
|
|
26
|
Jaudszus A, Mainz JG, Pittag S, Dornaus S, Dopfer C, Roth A, Jahreis G. Effects of a dietary intervention with conjugated linoleic acid on immunological and metabolic parameters in children and adolescents with allergic asthma--a placebo-controlled pilot trial. Lipids Health Dis 2016; 15:21. [PMID: 26843092 PMCID: PMC4739392 DOI: 10.1186/s12944-016-0187-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 01/20/2016] [Indexed: 01/09/2023] Open
Abstract
Background Circumstantial evidence suggests that conjugated linoleic acid (CLA) beneficially modulates immune function in allergic subjects. C9,t11-CLA, naturally occurring in ruminant fats, is suggested to be the effective isomer. In contrast, for the t10,c12-CLA isomer, which is naturally found only in traces but usually constitutes a relevant part in commercial CLA mixtures, adverse effects have been reported. Aim of this study was to assess putative immunomodulatory effects of highly enriched c9,t11-CLA in allergic subjects. To our best knowledge, our study is the first in that a CLA preparation was used for such purpose which was free of t10,c12-CLA. Design Twenty-nine asthmatic children and adolescents (age 6–18 y) with diagnosed allergic sensitization against grass pollen, house dust mite, or cat hair/epithelia consumed daily a portion of yoghurt containing either 3 g CLA (75 % c9,t11-CLA, 87 % purity) or placebo (safflower oil) over a period of 12 weeks. At study start and end, lung function parameters, specific IgE, in vitro allergen-induced cytokine production in peripheral blood mononuclear cells (PBMC), plasma ECP, urinary 8-oxodG as marker of oxidation, fatty acid profiles of erythrocytes, and routine haematological parameters were determined. Prior to blood samplings, 3-days dietary records were requested. Throughout the study, the participants documented daily their peak expiratory flow and kept protocol about their allergy symptoms and usage of demand medication. Results In contrast to the CLA group, PBMC-produced IFN-γ and IL-4 increased significantly and by trend, respectively, in the placebo group. Moreover, plasma ECP tended to increase in the placebo group. In the pollen subgroup, FEV1 improved upon both CLA and placebo oil supplementation. In both intervention groups, the n-6/n-3 PUFA ratio in red blood cells decreased, mainly due to an increase in n-3 PUFA. Moreover, 8-oxodG excretion increased in both groups. No changes occurred regarding specific IgE concentrations, allergy symptoms, and volume parameters. Conclusion Our results indicate that CLA modestly dampens the inflammatory response on the cellular level. A clinically relevant amelioration of the symptoms could not be proved in atopic manifest patients. Trial registration NCT01026506.
Collapse
Affiliation(s)
- Anke Jaudszus
- Present Address: Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Haid-und-Neu-Straße 9, D-76131, Karlsruhe, Germany.
| | - Jochen G Mainz
- Department of Pediatrics, Pediatric Pulmonology, CF-Centre, Jena University Hospital, Kochstraße 2, D-07745, Jena, Germany
| | - Sylvia Pittag
- Department of Nutritional Physiology, Institute of Nutrition, Friedrich Schiller University Jena, Dornburger Straße 24, D-07743, Jena, Germany
| | - Sabine Dornaus
- Department of Pediatrics, Pediatric Pulmonology, CF-Centre, Jena University Hospital, Kochstraße 2, D-07745, Jena, Germany
| | - Christian Dopfer
- Department of Pediatrics, Pediatric Pulmonology, CF-Centre, Jena University Hospital, Kochstraße 2, D-07745, Jena, Germany
| | - Alexander Roth
- Present Address: Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Haid-und-Neu-Straße 9, D-76131, Karlsruhe, Germany
| | - Gerhard Jahreis
- Department of Nutritional Physiology, Institute of Nutrition, Friedrich Schiller University Jena, Dornburger Straße 24, D-07743, Jena, Germany
| |
Collapse
|
27
|
Wang J, Chen H, Yang B, Gu Z, Zhang H, Chen W, Chen YQ. Lactobacillus plantarum ZS2058 produces CLA to ameliorate DSS-induced acute colitis in mice. RSC Adv 2016. [DOI: 10.1039/c5ra24491a] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Lactobacillus plantarumZS2058 is an efficient producer of conjugated linoleic acid (CLA)in vitro.
Collapse
Affiliation(s)
- Juntong Wang
- State Key Laboratory of Food Science and Technology
- School of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
| | - Haiqin Chen
- State Key Laboratory of Food Science and Technology
- School of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
| | - Bo Yang
- State Key Laboratory of Food Science and Technology
- School of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
| | - Zhennan Gu
- State Key Laboratory of Food Science and Technology
- School of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology
- School of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology
- School of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
| | - Yong Q. Chen
- State Key Laboratory of Food Science and Technology
- School of Food Science and Technology
- Jiangnan University
- Wuxi
- P. R. China
| |
Collapse
|
28
|
Does Diet Matter? The Use of Polyunsaturated Fatty Acids (PUFAs) and Other Dietary Supplements in Inflammation-Associated Depression. Curr Top Behav Neurosci 2016; 31:321-338. [PMID: 27431396 DOI: 10.1007/7854_2016_31] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
An increasingly pertinent issue in psychiatry in recent years is that of the limitations of conventional antidepressants, which are not effective in a large number of patients with major depressive disorder (MDD). Coupled with emerging hypotheses about the role of inflammation in depression, it would appear that it is time to look for alternative treatments for these symptoms.This review will examine an emerging area in psychiatry, that of dietary supplements and the diet in general to treat depressive symptoms, and inflammation in depression. In particular, polyunsaturated fatty acids (PUFAs), probiotics and folic acid are three supplements that demonstrate the ability to target inflammation and other underlying systems in depression. While there is a definite need for more research in all these supplements to determine true efficacy, dosage and target populations, they can be used as mono- or adjunctive therapies to good effect, and show superior safety profiles when compared with more traditional alternatives.
Collapse
|
29
|
Usami M, Miyoshi M, Yamashita H. Gut microbiota and host metabolism in liver cirrhosis. World J Gastroenterol 2015; 21:11597-11608. [PMID: 26556989 PMCID: PMC4631963 DOI: 10.3748/wjg.v21.i41.11597] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/06/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
The gut microbiota has the capacity to produce a diverse range of compounds that play a major role in regulating the activity of distal organs and the liver is strategically positioned downstream of the gut. Gut microbiota linked compounds such as short chain fatty acids, bile acids, choline metabolites, indole derivatives, vitamins, polyamines, lipids, neurotransmitters and neuroactive compounds, and hypothalamic-pituitary-adrenal axis hormones have many biological functions. This review focuses on the gut microbiota and host metabolism in liver cirrhosis. Dysbiosis in liver cirrhosis causes serious complications, such as bacteremia and hepatic encephalopathy, accompanied by small intestinal bacterial overgrowth and increased intestinal permeability. Gut dysbiosis in cirrhosis and intervention with probiotics and synbiotics in a clinical setting is reviewed and evaluated. Recent studies have revealed the relationship between gut microbiota and host metabolism in chronic metabolic liver disease, especially, non-alcoholic fatty liver disease, alcoholic liver disease, and with the gut microbiota metabolic interactions in dysbiosis related metabolic diseases such as diabetes and obesity. Recently, our understanding of the relationship between the gut and liver and how this regulates systemic metabolic changes in liver cirrhosis has increased. The serum lipid levels of phospholipids, free fatty acids, polyunsaturated fatty acids, especially, eicosapentaenoic acid, arachidonic acid, and docosahexaenoic acid have significant correlations with specific fecal flora in liver cirrhosis. Many clinical and experimental reports support the relationship between fatty acid metabolism and gut-microbiota. Various blood metabolome such as cytokines, amino acids, and vitamins are correlated with gut microbiota in probiotics-treated liver cirrhosis patients. The future evaluation of the gut-microbiota-liver metabolic network and the intervention of these relationships using probiotics, synbiotics, and prebiotics, with sufficient nutrition could aid the development of treatments and prevention for liver cirrhosis patients.
Collapse
|
30
|
Savignac HM, Tramullas M, Kiely B, Dinan TG, Cryan JF. Bifidobacteria modulate cognitive processes in an anxious mouse strain. Behav Brain Res 2015; 287:59-72. [PMID: 25794930 DOI: 10.1016/j.bbr.2015.02.044] [Citation(s) in RCA: 258] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 02/15/2015] [Accepted: 02/20/2015] [Indexed: 12/17/2022]
Abstract
Increasing evidence suggests that a brain-gut-microbiome axis exists, which has the potential to play a major role in modulating behaviour. However, the role of this axis in cognition remains relatively unexplored. Probiotics, which are commensal bacteria offering potential health benefit, have been shown to decrease anxiety, depression and visceral pain-related behaviours. In this study, we investigate the potential of two Bifidobacteria strains to modulate cognitive processes and visceral pain sensitivity. Adult male BALB/c mice were fed daily for 11 weeks with B. longum 1714, B. breve 1205 or vehicle treatment. Starting at week 4, animals were behaviourally assessed in a battery of tests relevant to different aspects of cognition, as well as locomotor activity and visceral pain. In the object recognition test, B. longum 1714-fed mice discriminated between the two objects faster than all other groups and B. breve 1205-fed mice discriminated faster than vehicle animals. In the Barnes maze, B. longum 1714-treated mice made fewer errors than other groups, suggesting a better learning. In the fear conditioning, B. longum 1714-treated group also showed better learning and memory, yet presenting the same extinction learning profile as controls. None of the treatments affected visceral sensitivity. Altogether, these data suggest that B. longum 1714 had a positive impact on cognition and also that the effects of individual Bifidobacteria strains do not generalise across the species. Clinical validation of the effects of probiotics on cognition is now warranted.
Collapse
Affiliation(s)
- H M Savignac
- Alimentary Pharmabiotic Centre, University College Cork, Ireland; School of Pharmacy, University College Cork, Ireland.
| | - M Tramullas
- Alimentary Pharmabiotic Centre, University College Cork, Ireland
| | - B Kiely
- Alimentary Health Ltd., Cork, Ireland
| | - T G Dinan
- Alimentary Pharmabiotic Centre, University College Cork, Ireland; Department of Psychiatry, University College Cork, Ireland.
| | - J F Cryan
- Alimentary Pharmabiotic Centre, University College Cork, Ireland; Department of Anatomy and Neurosciences, University College Cork, Ireland.
| |
Collapse
|
31
|
Gondalia S, Scholey A. Gastrointestinal microbiota, diet and brain functioning. MICROBIOLOGY AUSTRALIA 2015. [DOI: 10.1071/ma15008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
32
|
Dinan TG, Borre YE, Cryan JF. Genomics of schizophrenia: time to consider the gut microbiome? Mol Psychiatry 2014; 19:1252-7. [PMID: 25288135 DOI: 10.1038/mp.2014.93] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/06/2014] [Accepted: 07/08/2014] [Indexed: 12/12/2022]
Abstract
Research into the genomics of schizophrenia promises much, but so far is resplendent with failures to replicate, and has yielded little of therapeutic potential. Within our bodies resides a dynamic population of gut microbes forming a symbiotic superorganism comprising a myriad of bacteria of approximately 10(14) cells, containing 100 times the number of genes of the human genome and weighing approximately the same as the human brain. Recent preclinical investigations indicate that these microbes majorly impact on cognitive function and fundamental behavior patterns, such as social interaction and stress management. We are pivotally dependent on the neuroactive substances produced by such bacteria. The biological diversity of this ecosystem is established in the initial months of life and is highly impacted upon by environmental factors. To date, this vast quantity of DNA has been largely ignored in schizophrenia research. Perhaps it is time to reconsider this omission.
Collapse
Affiliation(s)
- T G Dinan
- 1] Alimentary Pharmabiotic Centre, University College, Cork, Ireland [2] Department of Psychiatry, Cork University Hospital, University College Cork, Cork, Ireland
| | - Y E Borre
- Alimentary Pharmabiotic Centre, University College, Cork, Ireland
| | - J F Cryan
- 1] Alimentary Pharmabiotic Centre, University College, Cork, Ireland [2] Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
33
|
Bowe W, Patel NB, Logan AC. Acne vulgaris, probiotics and the gut-brain-skin axis: from anecdote to translational medicine. Benef Microbes 2014; 5:185-99. [PMID: 23886975 DOI: 10.3920/bm2012.0060] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Acne vulgaris has long been postulated to feature a gastrointestinal mechanism, dating back 80 years to dermatologists John H. Stokes and Donald M. Pillsbury. They hypothesised that emotional states (e.g. depression and anxiety) could alter normal intestinal microbiota, increase intestinal permeability, and contribute to systemic inflammation. They were also among the first to propose the use of probiotic Lactobacillus acidophilus cultures. In recent years, aspects of this gut-brain-skin theory have been further validated via modern scientific investigations. It is evident that gut microbes and oral probiotics could be linked to the skin, and particularly acne severity, by their ability to influence systemic inflammation, oxidative stress, glycaemic control, tissue lipid content, and even mood. This intricate relationship between gut microbiota and the skin may also be influenced by diet, a current area of intense scrutiny by those who study acne. Here we provide a historical background to the gut-brain-skin theory in acne, followed by a summary of contemporary investigations and clinical implications.
Collapse
Affiliation(s)
- W Bowe
- Department of Dermatology, State University of New York Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| | - N B Patel
- New Jersey Medical School, University of Medicine and Dentistry of New Jersey, 150 Bergen Street, Newark, NJ 07103, USA
| | - A C Logan
- Genuine Health, 775 East Blithedale Avenue, Suite 364, Mill Valley, CA 94941, USA
| |
Collapse
|
34
|
Borre YE, Moloney RD, Clarke G, Dinan TG, Cryan JF. The impact of microbiota on brain and behavior: mechanisms & therapeutic potential. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 817:373-403. [PMID: 24997043 DOI: 10.1007/978-1-4939-0897-4_17] [Citation(s) in RCA: 209] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
There is increasing evidence that host-microbe interactions play a key role in maintaining homeostasis. Alterations in gut microbial composition is associated with marked changes in behaviors relevant to mood, pain and cognition, establishing the critical importance of the bi-directional pathway of communication between the microbiota and the brain in health and disease. Dysfunction of the microbiome-brain-gut axis has been implicated in stress-related disorders such as depression, anxiety and irritable bowel syndrome and neurodevelopmental disorders such as autism. Bacterial colonization of the gut is central to postnatal development and maturation of key systems that have the capacity to influence central nervous system (CNS) programming and signaling, including the immune and endocrine systems. Moreover, there is now expanding evidence for the view that enteric microbiota plays a role in early programming and later response to acute and chronic stress. This view is supported by studies in germ-free mice and in animals exposed to pathogenic bacterial infections, probiotic agents or antibiotics. Although communication between gut microbiota and the CNS are not fully elucidated, neural, hormonal, immune and metabolic pathways have been suggested. Thus, the concept of a microbiome-brain-gut axis is emerging, suggesting microbiota-modulating strategies may be a tractable therapeutic approach for developing novel treatments for CNS disorders.
Collapse
Affiliation(s)
- Yuliya E Borre
- Laboratory of NeuroGastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | | | | | | | | |
Collapse
|
35
|
Savignac HM, Kiely B, Dinan TG, Cryan JF. Bifidobacteria exert strain-specific effects on stress-related behavior and physiology in BALB/c mice. Neurogastroenterol Motil 2014; 26:1615-27. [PMID: 25251188 DOI: 10.1111/nmo.12427] [Citation(s) in RCA: 286] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 08/17/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND Accumulating evidence suggests that commensal bacteria consumption has the potential to have a positive impact on stress-related psychiatric disorders. However, the specific bacteria influencing behaviors related to anxiety and depression remain unclear. To this end, we compared the effects of two different Bifidobacteria on anxiety and depression-like behavior; an antidepressant was also used as a comparator. METHODS Innately anxious BALB/c mice received daily Bifidobacterium longum (B.) 1714, B. breve 1205, the antidepressant escitalopram or vehicle treatment for 6 weeks. Behavior was assessed in stress-induced hyperthermia test, marble burying, elevated plus maze, open field, tail suspension test, and forced swim test. Physiological responses to acute stress were also assessed. KEY RESULTS Both Bifidobacteria and escitalopram reduced anxiety in the marble burying test; however, only B. longum 1714 decreased stress-induced hyperthermia. B. breve 1205 induced lower anxiety in the elevated plus maze whereas B. longum 1714 induced antidepressant-like behavior in the tail suspension test. However, there was no difference in corticosterone levels between groups. CONCLUSIONS & INFERENCES These data show that these two Bifidobacteria strains reduced anxiety in an anxious mouse strain. These results also suggest that each bacterial strain has intrinsic effects and may be beneficially specific for a given disorder. These findings strengthen the role of gut microbiota supplementation as psychobiotic-based strategies for stress-related brain-gut axis disorders, opening new avenues in the field of neurogastroenterology.
Collapse
Affiliation(s)
- H M Savignac
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | | | | | | |
Collapse
|
36
|
Abstract
A healthy gut microbiota plays many crucial functions in the host, being involved in the correct development and functioning of the immune system, assisting in the digestion of certain foods and in the production of health-beneficial bioactive metabolites or 'pharmabiotics'. These include bioactive lipids (including SCFA and conjugated linoleic acid) antimicrobials and exopolysaccharides in addition to nutrients, including vitamins B and K. Alterations in the composition of the gut microbiota and reductions in microbial diversity are highlighted in many disease states, possibly rendering the host susceptible to infection and consequently negatively affecting innate immune function. Evidence is also emerging of microbially produced molecules with neuroactive functions that can have influences across the brain-gut axis. For example, γ-aminobutyric acid, serotonin, catecholamines and acetylcholine may modulate neural signalling within the enteric nervous system, when released in the intestinal lumen and consequently signal brain function and behaviour. Dietary supplementation with probiotics and prebiotics are the most widely used dietary adjuncts to modulate the gut microbiota. Furthermore, evidence is emerging of the interactions between administered microbes and dietary substrates, leading to the production of pharmabiotics, which may directly or indirectly positively influence human health.
Collapse
|
37
|
Clarke G, Stilling RM, Kennedy PJ, Stanton C, Cryan JF, Dinan TG. Minireview: Gut microbiota: the neglected endocrine organ. Mol Endocrinol 2014; 28:1221-38. [PMID: 24892638 DOI: 10.1210/me.2014-1108] [Citation(s) in RCA: 706] [Impact Index Per Article: 70.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The concept that the gut microbiota serves as a virtual endocrine organ arises from a number of important observations. Evidence for a direct role arises from its metabolic capacity to produce and regulate multiple compounds that reach the circulation and act to influence the function of distal organs and systems. For example, metabolism of carbohydrates results in the production of short-chain fatty acids, such as butyrate and propionate, which provide an important source of nutrients as well as regulatory control of the host digestive system. This influence over host metabolism is also seen in the ability of the prebiotic inulin to influence production of relevant hormones such as glucagon-like peptide-1, peptide YY, ghrelin, and leptin. Moreover, the probiotic Lactobacillus rhamnosus PL60, which produces conjugated linoleic acid, has been shown to reduce body-weight gain and white adipose tissue without effects on food intake. Manipulating the microbial composition of the gastrointestinal tract modulates plasma concentrations of tryptophan, an essential amino acid and precursor to serotonin, a key neurotransmitter within both the enteric and central nervous systems. Indirectly and through as yet unknown mechanisms, the gut microbiota exerts control over the hypothalamic-pituitary-adrenal axis. This is clear from studies on animals raised in a germ-free environment, who show exaggerated responses to psychological stress, which normalizes after monocolonization by certain bacterial species including Bifidobacterium infantis. It is tempting to speculate that therapeutic targeting of the gut microbiota may be useful in treating stress-related disorders and metabolic diseases.
Collapse
Affiliation(s)
- Gerard Clarke
- Alimentary Pharmabiotic Centre (G.C., R.M.S., P.J.K., C.S., J.F.C., T.G.D.) and Departments of Psychiatry (G.C., C.S., T.G.D.) and Anatomy and Neuroscience (J.F.C.), University College Cork, Cork, Ireland; and Teagasc (C.S.), Moorepark, Fermoy, Cork, Ireland
| | | | | | | | | | | |
Collapse
|
38
|
'The way to a man's heart is through his gut microbiota'--dietary pro- and prebiotics for the management of cardiovascular risk. Proc Nutr Soc 2014; 73:172-85. [PMID: 24495527 DOI: 10.1017/s0029665113003911] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The human gut microbiota has been identified as a possible novel CVD risk factor. This review aims to summarise recent insights connecting human gut microbiome activities with CVD and how such activities may be modulated by diet. Aberrant gut microbiota profiles have been associated with obesity, type 1 and type 2 diabetes and non-alcoholic fatty liver disease. Transfer of microbiota from obese animals induces metabolic disease and obesity in germ-free animals. Conversely, transfer of pathogen-free microbiota from lean healthy human donors to patients with metabolic disease can increase insulin sensitivity. Not only are aberrant microbiota profiles associated with metabolic disease, but the flux of metabolites derived from gut microbial metabolism of choline, phosphatidylcholine and l-carnitine has been shown to contribute directly to CVD pathology, providing one explanation for increased disease risk of eating too much red meat. Diet, especially high intake of fermentable fibres and plant polyphenols, appears to regulate microbial activities within the gut, supporting regulatory guidelines encouraging increased consumption of whole-plant foods (fruit, vegetables and whole-grain cereals), and providing the scientific rationale for the design of efficacious prebiotics. Similarly, recent human studies with carefully selected probiotic strains show that ingestion of viable microorganisms with the ability to hydrolyse bile salts can lower blood cholesterol, a recognised risk factor in CVD. Taken together such observations raise the intriguing possibility that gut microbiome modulation by whole-plant foods, probiotics and prebiotics may be at the base of healthy eating pyramids advised by regulatory agencies across the globe. In conclusion, dietary strategies which modulate the gut microbiota or their metabolic activities are emerging as efficacious tools for reducing CVD risk and indicate that indeed, the way to a healthy heart may be through a healthy gut microbiota.
Collapse
|
39
|
Selhub EM, Logan AC, Bested AC. Fermented foods, microbiota, and mental health: ancient practice meets nutritional psychiatry. J Physiol Anthropol 2014; 33:2. [PMID: 24422720 PMCID: PMC3904694 DOI: 10.1186/1880-6805-33-2] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 12/18/2013] [Indexed: 02/06/2023] Open
Abstract
The purposeful application of fermentation in food and beverage preparation, as a means to provide palatability, nutritional value, preservative, and medicinal properties, is an ancient practice. Fermented foods and beverages continue to make a significant contribution to the overall patterns of traditional dietary practices. As our knowledge of the human microbiome increases, including its connection to mental health (for example, anxiety and depression), it is becoming increasingly clear that there are untold connections between our resident microbes and many aspects of physiology. Of relevance to this research are new findings concerning the ways in which fermentation alters dietary items pre-consumption, and in turn, the ways in which fermentation-enriched chemicals (for example, lactoferrin, bioactive peptides) and newly formed phytochemicals (for example, unique flavonoids) may act upon our own intestinal microbiota profile. Here, we argue that the consumption of fermented foods may be particularly relevant to the emerging research linking traditional dietary practices and positive mental health. The extent to which traditional dietary items may mitigate inflammation and oxidative stress may be controlled, at least to some degree, by microbiota. It is our contention that properly controlled fermentation may often amplify the specific nutrient and phytochemical content of foods, the ultimate value of which may associated with mental health; furthermore, we also argue that the microbes (for example, Lactobacillus and Bifidobacteria species) associated with fermented foods may also influence brain health via direct and indirect pathways.
Collapse
Affiliation(s)
- Eva M Selhub
- Harvard Medical School and Massachusetts General Hospital, 40 Crescent St., Suite 201, Waltham, MA 02453, USA
| | - Alan C Logan
- CAMNR, 23679 Calabasas Road Suite 542, Calabasas, CA 91302, USA
| | - Alison C Bested
- Complex Chronic Diseases Program, BC Women’s Hospital and Health Centre, B223A-4500 Oak Street, Vancouver, BC V6H 3N1, Canada
| |
Collapse
|
40
|
Abstract
Bacterial colonisation of the gut plays a major role in postnatal development and maturation of key systems that have the capacity to influence central nervous system (CNS) programming and signaling, including the immune and endocrine systems. Individually, these systems have been implicated in the neuropathology of many CNS disorders and collectively they form an important bidirectional pathway of communication between the microbiota and the brain in health and disease. Regulation of the microbiome-brain-gut axis is essential for maintaining homeostasis, including that of the CNS. Moreover, there is now expanding evidence for the view that commensal organisms within the gut play a role in early programming and later responsivity of the stress system. Research has focused on how the microbiota communicates with the CNS and thereby influences brain function. The routes of this communication are not fully elucidated but include neural, humoral, immune and metabolic pathways. This view is underpinned by studies in germ-free animals and in animals exposed to pathogenic bacterial infections, probiotic agents or antibiotics which indicate a role for the gut microbiota in the regulation of mood, cognition, pain and obesity. Thus, the concept of a microbiome-brain-gut axis is emerging which suggests that modulation of the gut microflora may be a tractable strategy for developing novel therapeutics for complex stress-related CNS disorders where there is a huge unmet medical need.
Collapse
|
41
|
Tabbaa M, Golubic M, Roizen MF, Bernstein AM. Docosahexaenoic acid, inflammation, and bacterial dysbiosis in relation to periodontal disease, inflammatory bowel disease, and the metabolic syndrome. Nutrients 2013; 5:3299-310. [PMID: 23966110 PMCID: PMC3775255 DOI: 10.3390/nu5083299] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Revised: 07/30/2013] [Accepted: 08/08/2013] [Indexed: 02/06/2023] Open
Abstract
Docosahexaenoic acid (DHA), a long-chain omega-3 polyunsaturated fatty acid, has been used to treat a range of different conditions, including periodontal disease (PD) and inflammatory bowel disease (IBD). That DHA helps with these oral and gastrointestinal diseases in which inflammation and bacterial dysbiosis play key roles, raises the question of whether DHA may assist in the prevention or treatment of other inflammatory conditions, such as the metabolic syndrome, which have also been linked with inflammation and alterations in normal host microbial populations. Here we review established and investigated associations between DHA, PD, and IBD. We conclude that by beneficially altering cytokine production and macrophage recruitment, the composition of intestinal microbiota and intestinal integrity, lipopolysaccharide- and adipose-induced inflammation, and insulin signaling, DHA may be a key tool in the prevention of metabolic syndrome.
Collapse
Affiliation(s)
| | | | | | - Adam M. Bernstein
- Cleveland Clinic, Wellness Institute, 1950 Richmond Road/TR2-203, Lyndhurst, OH 44124, USA; E-Mails: (M.T.); (M.G.); (M.F.R.)
| |
Collapse
|
42
|
Li J, Zhang L, Han X, Yi H, Guo C, Zhang Y, Du M, Luo X, Zhang Y, Shan Y. Effect of incubation conditions and possible intestinal nutrients on cis-9, trans-11 conjugated linoleic acid production by Lactobacillus acidophilus F0221. Int Dairy J 2013. [DOI: 10.1016/j.idairyj.2012.10.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
43
|
Implication of fermentable carbohydrates targeting the gut microbiota on conjugated linoleic acid production in high-fat-fed mice. Br J Nutr 2013; 110:998-1011. [PMID: 23507010 DOI: 10.1017/s0007114513000123] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In vitro experiments have shown that isolated human gut bacteria are able to metabolise PUFA into conjugated PUFA like conjugated linoleic acids (CLA). The hypothesis of the present paper was that high-fat (HF) diet feeding and supplementation with fermentable carbohydrates that have prebiotic properties modulate the in vivo production of CLA by the mouse gut microbiota. Mice were treated for 4 weeks as follows: control (CT) groups were fed a standard diet; HF groups were fed a HF diet rich in linoleic acid (18 : 2n-6); the third groups were fed with the HF diet supplemented with either inulin-type fructans (HF-ITF) or arabinoxylans (HF-Ax). HF diet feeding increased rumenic acid (cis-9,trans-11-18 : 2 CLA) content both in the caecal and liver tissues compared with the CT groups. ITF supplementation had no major effect compared with the HF diet whereas Ax supplementation increased further rumenic acid (cis-9,trans-11-18 : 2 CLA) in the caecal tissue. These differences between both prebiotics may be linked to the high fat-binding capacity of Ax that provides more substrates for bacterial metabolism and to differential modulation of the gut microbiota (specific increase in Roseburia spp. in HF-Ax v. HF). In conclusion, these experiments supply the proof of concept that the mouse gut microbiota produces CLA in vivo, with consequences on the level of CLA in the caecal and liver tissues. We postulate that the CLA-producing bacteria could be a mediator to consider in the metabolic effects of both HF diet feeding and prebiotic supplementation.
Collapse
|
44
|
Saulnier DM, Ringel Y, Heyman MB, Foster JA, Bercik P, Shulman RJ, Versalovic J, Verdu EF, Dinan TG, Hecht G, Guarner F. The intestinal microbiome, probiotics and prebiotics in neurogastroenterology. Gut Microbes 2013; 4. [PMID: 23202796 PMCID: PMC3555881 DOI: 10.4161/gmic.22973] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The brain-gut axis allows bidirectional communication between the central nervous system (CNS) and the enteric nervous system (ENS), linking emotional and cognitive centers of the brain with peripheral intestinal functions. Recent experimental work suggests that the gut microbiota have an impact on the brain-gut axis. A group of experts convened by the International Scientific Association for Probiotics and Prebiotics (ISAPP) discussed the role of gut bacteria on brain functions and the implications for probiotic and prebiotic science. The experts reviewed and discussed current available data on the role of gut microbiota on epithelial cell function, gastrointestinal motility, visceral sensitivity, perception and behavior. Data, mostly gathered from animal studies, suggest interactions of gut microbiota not only with the enteric nervous system but also with the central nervous system via neural, neuroendocrine, neuroimmune and humoral links. Microbial colonization impacts mammalian brain development in early life and subsequent adult behavior. These findings provide novel insights for improved understanding of the potential role of gut microbial communities on psychological disorders, most particularly in the field of psychological comorbidities associated with functional bowel disorders like irritable bowel syndrome (IBS) and should present new opportunity for interventions with pro- and prebiotics.
Collapse
Affiliation(s)
- Delphine M. Saulnier
- NIZO Food Research; Ede, The Netherlands,Correspondence to: Delphine M. Saulnier,
| | - Yehuda Ringel
- Department of Medicine; University of North Carolina School of Medicine; Chapel Hill, NC USA
| | - Melvin B. Heyman
- Department of Pediatrics; University of California, San Francisco; San Francisco, CA USA
| | - Jane A. Foster
- Department of Medicine; Farncombe Family Digestive Health Research Institute; McMaster University; Hamilton, ON Canada,Department of Psychiatry and Behavioural Neurosciences; McMaster University; Hamilton, ON Canada
| | - Premysl Bercik
- Department of Medicine; Farncombe Family Digestive Health Research Institute; McMaster University; Hamilton, ON Canada,Department of Psychiatry and Behavioural Neurosciences; McMaster University; Hamilton, ON Canada
| | - Robert J. Shulman
- Department of Pediatrics; Baylor College of Medicine; Houston, TX USA
| | - James Versalovic
- Department of Pathology; Baylor College of Medicine; Houston, TX USA,Texas Children’s Microbiome Center; Texas Children’s Hospital; Houston, TX USA
| | - Elena F. Verdu
- Department of Medicine; Farncombe Family Digestive Health Research Institute; McMaster University; Hamilton, ON Canada
| | - Ted G. Dinan
- Alimentary Pharmabiotic Centre; University College Cork; Cork, Ireland
| | - Gail Hecht
- Department of Medicine; Microbiology/Immunology; University of Illinois, Chicago; Chicago, IL USA
| | | |
Collapse
|
45
|
Barrett E, Fitzgerald P, Dinan TG, Cryan JF, Ross RP, Quigley EM, Shanahan F, Kiely B, Fitzgerald GF, O'Toole PW, Stanton C. Bifidobacterium breve with α-linolenic acid and linoleic acid alters fatty acid metabolism in the maternal separation model of irritable bowel syndrome. PLoS One 2012; 7:e48159. [PMID: 23185248 PMCID: PMC3502412 DOI: 10.1371/journal.pone.0048159] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 09/21/2012] [Indexed: 01/23/2023] Open
Abstract
The aim of this study was to compare the impact of dietary supplementation with a Bifidobacterium breve strain together with linoleic acid & α-linolenic acid, for 7 weeks, on colonic sensitivity and fatty acid metabolism in rats. Maternally separated and non-maternally separated Sprague Dawley rats (n = 15) were orally gavaged with either B. breve DPC6330 (10(9) microorganisms/day) alone or in combination with 0.5% (w/w) linoleic acid & 0.5% (w/w) α-linolenic acid, daily for 7 weeks and compared with trehalose and bovine serum albumin. Tissue fatty acid composition was assessed by gas-liquid chromatography and visceral hypersensitivity was assessed by colorectal distension. Significant differences in the fatty acid profiles of the non-separated controls and maternally separated controls were observed for α-linolenic acid and arachidonic acid in the liver, oleic acid and eicosenoic acid (c11) in adipose tissue, and for palmitoleic acid and docosahexaenoic acid in serum (p<0.05). Administration of B. breve DPC6330 to MS rats significantly increased palmitoleic acid, arachidonic acid and docosahexaenoic acid in the liver, eicosenoic acid (c11) in adipose tissue and palmitoleic acid in the prefrontal cortex (p<0.05), whereas feeding B. breve DPC6330 to non separated rats significantly increased eicosapentaenoic acid and docosapentaenoic acid in serum (p<0.05) compared with the NS un-supplemented controls. Administration of B. breve DPC6330 in combination with linoleic acid and α-linolenic acid to maternally separated rats significantly increased docosapentaenoic acid in the serum (p<0.01) and α-linolenic acid in adipose tissue (p<0.001), whereas feeding B. breve DPC6330 with fatty acid supplementation to non-separated rats significantly increased liver and serum docosapentaenoic acid (p<0.05), and α-linolenic acid in adipose tissue (p<0.001). B. breve DPC6330 influenced host fatty acid metabolism. Administration of B. breve DPC6330 to maternally separated rats significantly modified the palmitoleic acid, arachidonic acid and docosahexaenoic acid contents in tissues. The effect was not observed in non-separated animals.
Collapse
Affiliation(s)
- Eoin Barrett
- Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland
- Teagasc, Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| | - Patrick Fitzgerald
- Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland
| | - Timothy G. Dinan
- Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland
| | - John F. Cryan
- Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland
| | - R. Paul Ross
- Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland
- Teagasc, Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| | - Eamonn M. Quigley
- Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland
| | - Fergus Shanahan
- Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland
| | - Barry Kiely
- Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland
| | - Gerald F. Fitzgerald
- Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland
- Department of Microbiology, University College Cork, Cork, Ireland
| | - Paul W. O'Toole
- Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland
- Department of Microbiology, University College Cork, Cork, Ireland
| | - Catherine Stanton
- Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland
- Teagasc, Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW Interest in the microbiota-gut-brain axis is increasing apace and what was, not so long ago, a hypothetical relationship is emerging as a potentially critical factor in the regulation of intestinal and mental health. Studies are now addressing the neural circuitry and mechanisms underlying the influence of gut bacteria on the central nervous system and behavior. RECENT FINDINGS Gut bacteria influence development of the central nervous systems (CNS) and stress responses. In adult animals, the overall composition of the microbiota or exposure to specific bacterial strains can modulate neural function, peripherally and centrally. Gut bacteria can provide protection from the central effects of infection and inflammation as well as modulate normal behavioral responses. Behavioral effects described to date are largely related to stress and anxiety and an altered hypothalamus-pituitary-adrenal axis response is a common observation in many model systems. The vagus nerve has also emerged as an important means of communicating signals from gut microbes to the CNS. SUMMARY Studies of microbiota-gut-brain communication are providing us with a deeper understanding of the relationship between the gut bacteria and their hosts while also suggesting the potential for microbial-based therapeutic strategies that may aid in the treatment of mood disorders.
Collapse
|
47
|
Usami M, Miyoshi M, Kanbara Y, Aoyama M, Sakaki H, Shuno K, Hirata K, Takahashi M, Ueno K, Hamada Y, Tabata S, Asahara T, Nomoto K. Analysis of fecal microbiota, organic acids and plasma lipids in hepatic cancer patients with or without liver cirrhosis. Clin Nutr 2012; 32:444-51. [PMID: 23068014 DOI: 10.1016/j.clnu.2012.09.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Revised: 09/19/2012] [Accepted: 09/24/2012] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Changes in the microbiota composition are able to affect nutrient absorption and energy metabolism, but there are few human studies. The aims were to analyze fecal constituents quantitatively and compare them with liver dysfunction in hepatic cancer patients and to evaluate the relationships among intestinal microbiota, fecal organic acids and plasma lipid composition. METHODS Fecal samples collected from 46 hepatic cancer patients (with liver cirrhosis, chronic hepatitis or liver fibrosis and normal liver) were evaluated for fecal constituents. Blood organic acid, lipid and fatty acid concentrations were analyzed. RESULTS Fecal microbiota and organic acids showed no significant differences among different liver dysfunction patients. In normal liver patients, fecal Candida was positively correlated with plasma phospholipid while Bifidobacterium was negatively correlated with plasma eicosapentaenoic acid and eicosapentaenoic acid/arachidonic acid ratio (all p < 0.05). In cirrhotic liver patients, positive correlations were noted for Lactobacillus and docosahexaenoic acid and Candida and eicosapentaenoic acid or eicosapentaenoic acid/arachidonic acid ratio (all p < 0.01). It was suggested that intestinal biota affected serum fatty acid metabolism and were modified by liver disorders. CONCLUSIONS Intestinal microbiota and organic acid concentrations in hepatic cancer patients had positive and/or negative correlations with serum lipid levels.
Collapse
Affiliation(s)
- Makoto Usami
- Division of Nutrition and Metabolism, Kobe University Graduate School of Health Sciences, Kobe, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Wall R, Marques TM, O'Sullivan O, Ross RP, Shanahan F, Quigley EM, Dinan TG, Kiely B, Fitzgerald GF, Cotter PD, Fouhy F, Stanton C. Contrasting effects of Bifidobacterium breve NCIMB 702258 and Bifidobacterium breve DPC 6330 on the composition of murine brain fatty acids and gut microbiota. Am J Clin Nutr 2012; 95:1278-87. [PMID: 22492373 DOI: 10.3945/ajcn.111.026435] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND We previously showed that microbial metabolism in the gut influences the composition of bioactive fatty acids in host adipose tissue. OBJECTIVE This study compared the effect of dietary supplementation for 8 wk with human-derived Bifidobacterium breve strains on fat distribution and composition and the composition of the gut microbiota in mice. METHODS C57BL/6 mice (n = 8 per group) received B. breve DPC 6330 or B. breve NCIMB 702258 (10(9) microorganisms) daily for 8 wk or no supplement (controls). Tissue fatty acid composition was assessed by gas-liquid chromatography while 16S rRNA pyrosequencing was used to investigate microbiota composition. RESULTS Visceral fat mass and brain stearic acid, arachidonic acid, and DHA were higher in mice supplemented with B. breve NCIMB 702258 than in mice in the other 2 groups (P < 0.05). In addition, both B. breve DPC 6330 and B. breve NCIMB 702258 supplementation resulted in higher propionate concentrations in the cecum than did no supplementation (P < 0.05). Compositional sequencing of the gut microbiota showed a tendency for greater proportions of Clostridiaceae (25%, 12%, and 18%; P = 0.08) and lower proportions of Eubacteriaceae (3%, 12%, and 13%; P = 0.06) in mice supplemented with B. breve DPC 6330 than in mice supplemented with B. breve NCIMB 702258 and unsupplemented controls, respectively. CONCLUSION The response of fatty acid metabolism to administration of bifidobacteria is strain-dependent, and strain-strain differences are important factors that influence modulation of the gut microbial community by ingested microorganisms.
Collapse
Affiliation(s)
- Rebecca Wall
- Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Cork, Ireland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
O'Shea EF, Cotter PD, Stanton C, Ross RP, Hill C. Production of bioactive substances by intestinal bacteria as a basis for explaining probiotic mechanisms: bacteriocins and conjugated linoleic acid. Int J Food Microbiol 2011; 152:189-205. [PMID: 21742394 DOI: 10.1016/j.ijfoodmicro.2011.05.025] [Citation(s) in RCA: 183] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 05/30/2011] [Accepted: 05/31/2011] [Indexed: 01/01/2023]
Abstract
The mechanisms by which intestinal bacteria achieve their associated health benefits can be complex and multifaceted. In this respect, the diverse microbial composition of the human gastrointestinal tract (GIT) provides an almost unlimited potential source of bioactive substances (pharmabiotics) which can directly or indirectly affect human health. Bacteriocins and fatty acids are just two examples of pharmabiotic substances which may contribute to probiotic functionality within the mammalian GIT. Bacteriocin production is believed to confer producing strains with a competitive advantage within complex microbial environments as a consequence of their associated antimicrobial activity. This has the potential to enable the establishment and prevalence of producing strains as well as directly inhibiting pathogens within the GIT. Consequently, these antimicrobial peptides and the associated intestinal producing strains may be exploited to beneficially influence microbial populations. Intestinal bacteria are also known to produce a diverse array of health-promoting fatty acids. Indeed, certain strains of intestinal bifidobacteria have been shown to produce conjugated linoleic acid (CLA), a fatty acid which has been associated with a variety of systemic health-promoting effects. Recently, the ability to modulate the fatty acid composition of the liver and adipose tissue of the host upon oral administration of CLA-producing bifidobacteria and lactobacilli was demonstrated in a murine model. Importantly, this implies a potential therapeutic role for probiotics in the treatment of certain metabolic and immunoinflammatory disorders. Such examples serve to highlight the potential contribution of pharmabiotic production to probiotic functionality in relation to human health maintenance.
Collapse
Affiliation(s)
- Eileen F O'Shea
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| | | | | | | | | |
Collapse
|
50
|
Bibliography. Neonatology and perinatology. Current world literature. Curr Opin Pediatr 2011; 23:253-7. [PMID: 21412083 DOI: 10.1097/mop.0b013e3283454167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|