1
|
Xiong S. Gut-Microbiota-Driven Lipid Metabolism: Mechanisms and Applications in Swine Production. Metabolites 2025; 15:248. [PMID: 40278377 PMCID: PMC12029090 DOI: 10.3390/metabo15040248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: The gut microbiota plays a pivotal role in host physiology through metabolite production, with lipids serving as essential biomolecules for cellular structure, metabolism, and signaling. This review aims to elucidate the interactions between gut microbiota and lipid metabolism and their implications for enhancing swine production. Methods: We systematically analyzed current literature on microbial lipid metabolism, focusing on mechanistic studies on microbiota-lipid interactions, key regulatory pathways in microbial lipid metabolism, and multi-omics evidence (metagenomic/metabolomic) from swine models. Results: This review outlines the structural and functional roles of lipids in bacterial membranes and examines the influence of gut microbiota on the metabolism of key lipid classes, including cholesterol, bile acids, choline, sphingolipids, and fatty acids. Additionally, we explore the potential applications of microbial lipid metabolism in enhancing swine production performance. Conclusions: Our analysis establishes a scientific framework for microbiota-based strategies to optimize lipid metabolism. The findings highlight potential interventions to improve livestock productivity through targeted manipulation of gut microbial communities.
Collapse
Affiliation(s)
- Shuqi Xiong
- National Key Laboratory of Pig Genetic Improvement and Germplasm Innovation, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| |
Collapse
|
2
|
Ayayee P, Custer G, Clayton JB, Price J, Ramer-Tait A, Larsen T. Assessing gut microbial provisioning of essential amino acids to host in a murine model with reconstituted gut microbiomes. RESEARCH SQUARE 2025:rs.3.rs-6255159. [PMID: 40195995 PMCID: PMC11975013 DOI: 10.21203/rs.3.rs-6255159/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Gut microbial essential amino acid (EAA) provisioning to mammalian hosts remains a critical yet poorly understood aspect of host-microbe nutritional interactions, with significant implications for human and animal health. To investigate microbial EAA contributions in mice with reconstituted gut microbiomes, we analyzed stable carbon isotopes (13C) of six EAAs across multiple organs. Germ-free (GF) mice fed a high-protein diet (18%) were compared to conventionalized (CVZ) mice fed a low-protein diet (10%) following fecal microbiota transplantation 30 days prior and a 20-day dietary intervention. We found no evidence for microbial EAA contributions to host tissues, with 13C-EAA fingerprinting revealing nearly identical patterns between GF and CVZ organs. Both groups maintained their expected microbiome statuses, with CVZ gut microbiota dominated by Firmicutes and Bacteroidetes phyla. These findings raise important questions about the functional capacities of reconstituted gut microbiomes. Future studies should investigate longer adaptation periods, varied dietary protein levels, and complementary analytical techniques to better understand the context-dependent nature of microbial EAA provisioning in mammalian hosts.
Collapse
Affiliation(s)
- Paul Ayayee
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
| | - Gordon Custer
- Department of Natural Sciences, University of Maryland Eastern Shore, Princess Anne, MD, USA
| | - Jonathan B. Clayton
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Jeff Price
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Amanda Ramer-Tait
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Thomas Larsen
- Max Planck Institute of Geoanthropology, Jena, Germany
- Institute for Prehistoric and Protohistoric Archaeology, University of Kiel, Kiel, Germany
| |
Collapse
|
3
|
Wang Y, Bai M, Peng Q, Li L, Tian F, Guo Y, Jing C. Angiogenesis, a key point in the association of gut microbiota and its metabolites with disease. Eur J Med Res 2024; 29:614. [PMID: 39710789 DOI: 10.1186/s40001-024-02224-5] [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: 10/09/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024] Open
Abstract
The gut microbiota is a complex and dynamic ecosystem that plays a crucial role in human health and disease, including obesity, diabetes, cardiovascular diseases, neurodegenerative diseases, inflammatory bowel disease, and cancer. Chronic inflammation is a common feature of these diseases and is closely related to angiogenesis (the process of forming new blood vessels), which is often dysregulated in pathological conditions. Inflammation potentially acts as a central mediator. This abstract aims to elucidate the connection between the gut microbiota and angiogenesis in various diseases. The gut microbiota influences angiogenesis through various mechanisms, including the production of metabolites that directly or indirectly affect vascularization. For example, short-chain fatty acids (SCFAs) such as butyrate, propionate, and acetate are known to regulate immune responses and inflammation, thereby affecting angiogenesis. In the context of cardiovascular diseases, the gut microbiota promotes atherosclerosis and vascular dysfunction by producing trimethylamine N-oxide (TMAO) and other metabolites that promote inflammation and endothelial dysfunction. Similarly, in neurodegenerative diseases, the gut microbiota may influence neuroinflammation and the integrity of the blood-brain barrier, thereby affecting angiogenesis. In cases of fractures and wound healing, the gut microbiota promotes angiogenesis by activating inflammatory responses and immune effects, facilitating the healing of tissue damage. In cancer, the gut microbiota can either inhibit or promote tumor growth and angiogenesis, depending on the specific bacterial composition and their metabolites. For instance, some bacteria can activate inflammasomes, leading to the production of inflammatory factors that alter the tumor immune microenvironment and activate angiogenesis-related signaling pathways, affecting tumor angiogenesis and metastasis. Some bacteria can directly interact with tumor cells, activating angiogenesis-related signaling pathways. Diet, as a modifiable factor, significantly influences angiogenesis through diet-derived microbial metabolites. Diet can rapidly alter the composition of the microbiota and its metabolic activity, thereby changing the concentration of microbial-derived metabolites and profoundly affecting the host's immune response and angiogenesis. For example, a high animal protein diet promotes the production of pro-atherogenic metabolites like TMAO, activating inflammatory pathways and interfering with platelet function, which is associated with the severity of coronary artery plaques, peripheral artery disease, and cardiovascular diseases. A diet rich in dietary fiber promotes the production of SCFAs, which act as ligands for cell surface or intracellular receptors, regulating various biological processes, including inflammation, tissue homeostasis, and immune responses, thereby influencing angiogenesis. In summary, the role of the gut microbiota in angiogenesis is multifaceted, playing an important role in disease progression by affecting various biological processes such as inflammation, immune responses, and multiple signaling pathways. Diet-derived microbial metabolites play a crucial role in linking the gut microbiota and angiogenesis. Understanding the complex interactions between diet, the gut microbiota, and angiogenesis has the potential to uncover novel therapeutic targets for managing these conditions. Therefore, interventions targeting the gut microbiota and its metabolites, such as through fecal microbiota transplantation (FMT) and the application of probiotics to alter the composition of the gut microbiota and enhance the production of beneficial metabolites, present a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yan Wang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Mingshuai Bai
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Qifan Peng
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Feng Tian
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Ying Guo
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Changqing Jing
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
4
|
Thompson SC, Beliakoff R, Garrett TJ, Gonzalez CF, Lorca GL. Erucic acid utilization by Lactobacillus johnsonii N6.2. Front Microbiol 2024; 15:1476958. [PMID: 39654680 PMCID: PMC11625735 DOI: 10.3389/fmicb.2024.1476958] [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: 08/06/2024] [Accepted: 11/06/2024] [Indexed: 12/12/2024] Open
Abstract
A multivariate nutritional analysis indicated that the consumption of erucic acid-rich food, a fatty acid (FA) found primarily in rapeseed and mustard oil, was positively correlated with higher counts of lactic acid bacteria (LAB). Furthermore, we showed Lactobacillus johnsonii N6.2, as well as other species of LAB tested from the former Lactobacillus genus, were able to efficiently use erucic acid (EA) as the source of FA. In this work, we identified significant changes induced in the FA profiles of L. johnsonii cultured with EA as the source of FA. We performed global transcriptomics to identify genes and pathways involved in EA utilization. It was found that L. johnsonii incorporates external fatty acids via a FakA/FakB and the plsX/plsY/plsC pathway for phosphatidic acid synthesis. It was found that cells grown in MRS with EA (MRS-E) significantly upregulated fakB2 and fakB4 when compared to cells grown in standard MRS with tween 80 as the source of FA. Additionally, in MRS-E, L. johnsonii N6.2 induced the expression of plsY2, plsC2 and plsC4 while the expression of pslX was constitutive during short term EA exposure. LC-MS analyses revealed that L. johnsonii N6.2 rapidly incorporates EA and synthesizes a variety of long chain fatty acids, including the health-relevant omega-9 monounsaturated fatty acids such as nervonic and gondoic acids.
Collapse
Affiliation(s)
- Sharon C. Thompson
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Reagan Beliakoff
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Timothy J. Garrett
- Department of Pathology, Immunology and Laboratory of Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Claudio F. Gonzalez
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Graciela L. Lorca
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| |
Collapse
|
5
|
Zhong Z, Mu X, Lang H, Wang Y, Jiang Y, Liu Y, Zeng Q, Xia S, Zhang B, Wang Z, Wang X, Zheng H. Gut symbiont-derived anandamide promotes reward learning in honeybees by activating the endocannabinoid pathway. Cell Host Microbe 2024; 32:1944-1958.e7. [PMID: 39419026 DOI: 10.1016/j.chom.2024.09.013] [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: 03/10/2024] [Revised: 07/28/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024]
Abstract
Polyunsaturated fatty acids (PUFAs) are dietary components participating in neurotransmission and cell signaling. Pollen is a source of PUFAs for honeybees, and disruptions in dietary PUFAs reduce the cognitive performance of honeybees. We reveal that gut bacteria of honeybees contribute to fatty acid metabolism, impacting reward learning. Gut bacteria possess Δ-6 desaturases that mediate fatty acid elongation and compensate for the absence of honeybee factors required for fatty acid metabolism. Colonization with Gilliamella apicola, but not a mutant lacking the Δ-6 desaturase FADS2, increases the production of anandamide (AEA), a ligand of the endocannabinoid system, and alters learning and memory. AEA activates the Hymenoptera-specific transient receptor AmHsTRPA in astrocytes, which induces Ca2+ influx and regulates glutamate re-uptake of glial cells to enhance reward learning. These findings illuminate the roles of gut symbionts in host fatty acid metabolism and the impacts of endocannabinoid signaling on the reward system of social insects.
Collapse
Affiliation(s)
- Zhaopeng Zhong
- College of Food Science and Nutritional Engineering, China Agricultural University, 100083 Beijing, China
| | - Xiaohuan Mu
- College of Food Science and Nutritional Engineering, China Agricultural University, 100083 Beijing, China
| | - Haoyu Lang
- College of Food Science and Nutritional Engineering, China Agricultural University, 100083 Beijing, China
| | - Yueyi Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, 100083 Beijing, China
| | - Yanling Jiang
- College of Food Science and Nutritional Engineering, China Agricultural University, 100083 Beijing, China
| | - Yuwen Liu
- College of Food Science and Nutritional Engineering, China Agricultural University, 100083 Beijing, China
| | - Qian Zeng
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Siyuan Xia
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Baotong Zhang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Zilong Wang
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Xiaofei Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, 100083 Beijing, China.
| | - Hao Zheng
- College of Food Science and Nutritional Engineering, China Agricultural University, 100083 Beijing, China.
| |
Collapse
|
6
|
Van Hul M, Cani PD, Petitfils C, De Vos WM, Tilg H, El-Omar EM. What defines a healthy gut microbiome? Gut 2024; 73:1893-1908. [PMID: 39322314 PMCID: PMC11503168 DOI: 10.1136/gutjnl-2024-333378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 08/16/2024] [Indexed: 09/27/2024]
Abstract
The understanding that changes in microbiome composition can influence chronic human diseases and the efficiency of therapies has driven efforts to develop microbiota-centred therapies such as first and next generation probiotics, prebiotics and postbiotics, microbiota editing and faecal microbiota transplantation. Central to microbiome research is understanding how disease impacts microbiome composition and vice versa, yet there is a problematic issue with the term 'dysbiosis', which broadly links microbial imbalances to various chronic illnesses without precision or definition. Another significant issue in microbiome discussions is defining 'healthy individuals' to ascertain what characterises a healthy microbiome. This involves questioning who represents the healthiest segment of our population-whether it is those free from illnesses, athletes at peak performance, individuals living healthily through regular exercise and good nutrition or even elderly adults or centenarians who have been tested by time and achieved remarkable healthy longevity.This review advocates for delineating 'what defines a healthy microbiome?' by considering a broader range of factors related to human health and environmental influences on the microbiota. A healthy microbiome is undoubtedly linked to gut health. Nevertheless, it is very difficult to pinpoint a universally accepted definition of 'gut health' due to the complexities of measuring gut functionality besides the microbiota composition. We must take into account individual variabilities, the influence of diet, lifestyle, host and environmental factors. Moreover, the challenge in distinguishing causation from correlation between gut microbiome and overall health is presented.The review also highlights the resource-heavy nature of comprehensive gut health assessments, which hinders their practicality and broad application. Finally, we call for continued research and a nuanced approach to better understand the intricate and evolving concept of gut health, emphasising the need for more precise and inclusive definitions and methodologies in studying the microbiome.
Collapse
Affiliation(s)
- Matthias Van Hul
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute, Wavre, Belgium
| | - Patrice D Cani
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute, Wavre, Belgium
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Camille Petitfils
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute, Wavre, Belgium
| | - Willem M De Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medizinische Universitat Innsbruck, Innsbruck, Austria
| | - Emad M El-Omar
- Microbiome Research Centre, St George and Sutherland Clinical Campuses, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
7
|
Kim S, Seo SU, Kweon MN. Gut microbiota-derived metabolites tune host homeostasis fate. Semin Immunopathol 2024; 46:2. [PMID: 38990345 PMCID: PMC11239740 DOI: 10.1007/s00281-024-01012-x] [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: 12/13/2023] [Accepted: 03/15/2024] [Indexed: 07/12/2024]
Abstract
The gut microbiota, housing trillions of microorganisms within the gastrointestinal tract, has emerged as a critical regulator of host health and homeostasis. Through complex metabolic interactions, these microorganisms produce a diverse range of metabolites that substantially impact various physiological processes within the host. This review aims to delve into the intricate relationships of gut microbiota-derived metabolites and their influence on the host homeostasis. We will explore how these metabolites affect crucial aspects of host physiology, including metabolism, mucosal integrity, and communication among gut tissues. Moreover, we will spotlight the potential therapeutic applications of targeting these metabolites to restore and sustain host equilibrium. Understanding the intricate interplay between gut microbiota and their metabolites is crucial for developing innovative strategies to promote wellbeing and improve outcomes of chronic diseases.
Collapse
Affiliation(s)
- Seungil Kim
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine / Asan Medical Center, Seoul, Republic of Korea
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Uk Seo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi-Na Kweon
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine / Asan Medical Center, Seoul, Republic of Korea.
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Salsinha AS, Cima A, Araújo-Rodrigues H, Viana S, Reis F, Coscueta ER, Rodríguez-Alcalá LM, Relvas JB, Pintado M. The use of an in vitro fecal fermentation model to uncover the beneficial role of omega-3 and punicic acid in gut microbiota alterations induced by a Western diet. Food Funct 2024; 15:6095-6117. [PMID: 38757812 DOI: 10.1039/d4fo00727a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
The influence of gut microbiota in the onset and development of several metabolic diseases has gained attention over the last few years. Diet plays an essential role in gut microbiota modulation. Western diet (WD), characterized by high-sugar and high-fat consumption, alters gut microbiome composition, diversity index, microbial relative levels, and functional pathways. Despite the promising health effects demonstrated by polyunsaturated fatty acids, their impact on gut microbiota is still overlooked. The effect of Fish oil (omega-3 source) and Pomegranate oil (punicic acid source), and a mixture of both oils in gut microbiota modulation were determined by subjecting the oil samples to in vitro fecal fermentations. Cecal samples from rats from two different dietary groups: a control diet (CD) and a high-fat high-sugar diet (WD), were used as fecal inoculum. 16S amplicon metagenomics sequencing showed that Fish oil + Pomegranate oil from the WD group increased α-diversity. This sample can also increase the relative abundance of the Firmicutes and Bacteroidetes phylum as well as Akkermansia and Blautia, which were affected by the WD consumption. All samples were able to increase butyrate and acetate concentration in the WD group. Moreover, tyrosine concentrations, a precursor for dopamine and norepinephrine, increase in the Fish oil + Pomegranate oil WD sample. GABA, an important neurotransmitter, was also increased in WD samples. These results suggest a potential positive impact of these oils' mixture on gut-brain axis modulation. It was demonstrated, for the first time, the great potential of using a mixture of both Fish and Pomegranate oil to restore the gut microbiota changes associated with WD consumption.
Collapse
Affiliation(s)
- Ana Sofia Salsinha
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto - Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - André Cima
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
| | - Helena Araújo-Rodrigues
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto - Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Sofia Viana
- Coimbra Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra, Coimbra, Portugal
- Instituto Politécnico de Coimbra, Escola Superior de Tecnologia da Saúde de Coimbra, Rua 5 de Outubro - S. Martinho Bispo, Apartado 7006, 3046-854 Coimbra, Portugal
| | - Flávio Reis
- Coimbra Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Ezequiel R Coscueta
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
| | - Luis Miguel Rodríguez-Alcalá
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
| | - João B Relvas
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto - Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Departmento de Biomedicina, Faculdade de Medicina da Universidade do Porto (FMUP), 4200-319 Porto, Portugal
| | - Manuela Pintado
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina -Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
| |
Collapse
|
9
|
Roussel C, Sola M, Lessard-Lord J, Nallabelli N, Généreux P, Cavestri C, Azeggouar Wallen O, Villano R, Raymond F, Flamand N, Silvestri C, Di Marzo V. Human gut microbiota and their production of endocannabinoid-like mediators are directly affected by a dietary oil. Gut Microbes 2024; 16:2335879. [PMID: 38695302 PMCID: PMC11067990 DOI: 10.1080/19490976.2024.2335879] [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: 01/02/2024] [Accepted: 03/25/2024] [Indexed: 05/05/2024] Open
Abstract
Dietary omega-3 polyunsaturated fatty acids (n-3 PUFAs) and the gut microbiome affect each other. We investigated the impact of supplementation with Buglossoides arvensis oil (BO), rich in stearidonic acid (SDA), on the human gut microbiome. Employing the Mucosal Simulator of the Human Intestinal Microbial Ecosystem (M-SHIME), we simulated the ileal and ascending colon microbiomes of four donors. Our results reveal two distinct microbiota clusters influenced by BO, exhibiting shared and contrasting shifts. Notably, Bacteroides and Clostridia abundance underwent similar changes in both clusters, accompanied by increased propionate production in the colon. However, in the ileum, cluster 2 displayed a higher metabolic activity in terms of BO-induced propionate levels. Accordingly, a triad of bacterial members involved in propionate production through the succinate pathway, namely Bacteroides, Parabacteroides, and Phascolarctobacterium, was identified particularly in this cluster, which also showed a surge of second-generation probiotics, such as Akkermansia, in the colon. Finally, we describe for the first time the capability of gut bacteria to produce N-acyl-ethanolamines, and particularly the SDA-derived N-stearidonoyl-ethanolamine, following BO supplementation, which also stimulated the production of another bioactive endocannabinoid-like molecule, commendamide, in both cases with variations across individuals. Spearman correlations enabled the identification of bacterial genera potentially involved in endocannabinoid-like molecule production, such as, in agreement with previous reports, Bacteroides in the case of commendamide. This study suggests that the potential health benefits on the human microbiome of certain dietary oils may be amenable to stratified nutrition strategies and extend beyond n-3 PUFAs to include microbiota-derived endocannabinoid-like mediators.
Collapse
Affiliation(s)
- Charlène Roussel
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec, QC, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
| | - Mathilde Sola
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
| | - Jacob Lessard-Lord
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec, QC, Canada
| | - Nayudu Nallabelli
- Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada
| | - Pamela Généreux
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
| | - Camille Cavestri
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
| | - Oumaima Azeggouar Wallen
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
- Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada
| | - Rosaria Villano
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche (CNR), Pozzuoli (Napoli), Italy
| | - Frédéric Raymond
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec, QC, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
| | - Nicolas Flamand
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
- Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada
| | - Cristoforo Silvestri
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec, QC, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
- Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada
| | - Vincenzo Di Marzo
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Quebec, QC, Canada
- Centre Nutrition, Santé et Société (NUTRISS), INAF Laval University, Quebec, QC, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Laval University, Quebec, QC, Canada
- Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada
| |
Collapse
|
10
|
Wang B, Jadhav V, Odelade A, Chang E, Chang A, Harrison SH, Maldonado-Devincci AM, Graves JL, Han J. High fat diet reveals sex-specific fecal and liver metabolic alterations in C57BL/6J obese mice. Metabolomics 2023; 19:97. [PMID: 37999907 PMCID: PMC11651078 DOI: 10.1007/s11306-023-02059-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 10/18/2023] [Indexed: 11/25/2023]
Abstract
Obesity is a major health concern that poses significant risks for many other diseases, including diabetes, cardiovascular disease, and cancer. Prevalence of these diseases varies by biological sex. This study utilizes a mouse (C57BL/6J) model of obesity to analyze liver and fecal metabolic profiles at various time points of dietary exposure: 5, 9, and 12 months in control or high fat diet (HFD)-exposed mice. Our study discovered that the female HFD group has a more discernable perturbation and set of significant changes in metabolic profiles than the male HFD group. In the female mice, HFD fecal metabolites including pyruvate, aspartate, and glutamate were lower than control diet-exposed mice after both 9th and 12th month exposure time points, while lactate and alanine were significantly downregulated only at the 12th month. Perturbations of liver metabolic profiles were observed in both male and female HFD groups, compared to controls at the 12th month. Overall, the female HFD group showed higher lactate and glutathione levels compared to controls, while the male HFD group showed higher levels of glutamine and taurine compared to controls. These metabolite-based findings in both fecal and liver samples for a diet-induced effect of obesity may help guide future pioneering discoveries relating to the analysis and prevention of obesity in people, especially for females.
Collapse
Affiliation(s)
- Bo Wang
- Department of Chemistry and Chemical Engineering, Florida Institute of Technology, Melbourne, FL, 32901, USA
| | - Vidya Jadhav
- Department of Biology, College of Science and Technology, North Carolina Agricultural and Technical State University, Greensboro, NC, 27411, USA
| | - Anuoluwapo Odelade
- Department of Biology, College of Science and Technology, North Carolina Agricultural and Technical State University, Greensboro, NC, 27411, USA
| | - Evelyn Chang
- Program in Liberal Medical Education, Division of Biology and Medicine, Brown University, Providence, Rhode Island, 02912, USA
| | - Alex Chang
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, 14852, USA
| | - Scott H Harrison
- Department of Biology, College of Science and Technology, North Carolina Agricultural and Technical State University, Greensboro, NC, 27411, USA
| | - Antoinette M Maldonado-Devincci
- Department of Psychology, Hairston College of Health and Human Sciences, North Carolina Agricultural and Technical State University, Greensboro, 27411, USA
| | - Joseph L Graves
- Department of Biology, College of Science and Technology, North Carolina Agricultural and Technical State University, Greensboro, NC, 27411, USA
| | - Jian Han
- Department of Biology, College of Science and Technology, North Carolina Agricultural and Technical State University, Greensboro, NC, 27411, USA.
| |
Collapse
|
11
|
Leyrolle Q, Prado-Perez L, Layé S. The gut-derived metabolites as mediators of the effect of healthy nutrition on the brain. Front Nutr 2023; 10:1155533. [PMID: 37360297 PMCID: PMC10289296 DOI: 10.3389/fnut.2023.1155533] [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: 01/31/2023] [Accepted: 05/10/2023] [Indexed: 06/28/2023] Open
Abstract
Nutrition is now well recognized to be an environmental factor which positively or negatively influences the risk to develop neurological and psychiatric disorders. The gut microbiota has recently been shown to be an important actor mediating the relationship between environmental factors, including nutrition, and brain function. While its composition has been widely studied and associated with the risk of brain diseases, the mechanisms underlying the relationship between the gut and brain diseases remain to be explored. The wide range of bioactive molecules produced by the gut microbiota, called gut-derived metabolites (GDM), represent new players in the gut to brain interactions and become interesting target to promote brain health. The aim of this narrative review is to highlight some GDMs of interest that are produced in response to healthy food consumption and to summarize what is known about their potential effects on brain function. Overall, GDMs represent future useful biomarkers for the development of personalized nutrition. Indeed, their quantification after nutritional interventions is a useful tool to determine individuals' ability to produce microbiota-derived bioactive compounds upon consumption of specific food or nutrients. Moreover, GDMs represent also a new therapeutic approach to counteract the lack of response to conventional nutritional interventions.
Collapse
Affiliation(s)
- Quentin Leyrolle
- NutriNeurO, UMR 1286, Bordeaux INP, INRAE, University of Bordeaux, Bordeaux, France
| | | | | |
Collapse
|
12
|
Scarano A, Laddomada B, Blando F, De Santis S, Verna G, Chieppa M, Santino A. The Chelating Ability of Plant Polyphenols Can Affect Iron Homeostasis and Gut Microbiota. Antioxidants (Basel) 2023; 12:antiox12030630. [PMID: 36978878 PMCID: PMC10045931 DOI: 10.3390/antiox12030630] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
In the past decades, many studies have widely examined the effects of dietary polyphenols on human health. Polyphenols are well known for their antioxidant properties and for their chelating abilities, by which they can be potentially employed in cases of pathological conditions, such as iron overload. In this review, we have highlighted the chelating abilities of polyphenols, which are due to their structural specific sites, and the differences for each class of polyphenols. We have also explored how the dietary polyphenols and their iron-binding abilities can be important in inflammatory/immunomodulatory responses, with a special focus on the involvement of macrophages and dendritic cells, and how they might contribute to reshape the gut microbiota into a healthy profile. This review also provides evidence that the axes “polyphenol–iron metabolism–inflammatory responses” and “polyphenol–iron availability–gut microbiota” have not been very well explored so far, and the need for further investigation to exploit such a potential to prevent or counteract pathological conditions.
Collapse
Affiliation(s)
- Aurelia Scarano
- Institute of Science of Food Production, C.N.R. Unit of Lecce, 73100 Lecce, Italy
| | - Barbara Laddomada
- Institute of Science of Food Production, C.N.R. Unit of Lecce, 73100 Lecce, Italy
| | - Federica Blando
- Institute of Science of Food Production, C.N.R. Unit of Lecce, 73100 Lecce, Italy
| | - Stefania De Santis
- Department of Pharmacy-Pharmaceutical Science, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Giulio Verna
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Marcello Chieppa
- Department of Biological and Environmental Sciences and Technologies (DISTEBA), University of Salento, 73100 Lecce, Italy
- Correspondence: (M.C.); (A.S.)
| | - Angelo Santino
- Institute of Science of Food Production, C.N.R. Unit of Lecce, 73100 Lecce, Italy
- Correspondence: (M.C.); (A.S.)
| |
Collapse
|
13
|
Abstract
Metabolites produced by commensal gut microbes impact host health through their recognition by the immune system and their influence on numerous metabolic pathways. Notably, the gut microbiota can both transform and synthesize lipids as well as break down dietary lipids to generate secondary metabolites with host modulatory properties. Although lipids have largely been consigned to structural roles, particularly in cell membranes, recent research has led to an increased appreciation of their signaling activities, with potential impacts on host health and physiology. This review focuses on studies that highlight the functions of bioactive lipids in mammalian physiology, with a special emphasis on immunity and metabolism.
Collapse
Affiliation(s)
- Eric M Brown
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Jon Clardy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Blavatnik Institute, Boston, MA 02115, USA
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
14
|
Jeong MK, Min BH, Choi YR, Hyun JY, Park HJ, Eom JA, Won SM, Jeong JJ, Oh KK, Gupta H, Ganesan R, Sharma SP, Yoon SJ, Choi MR, Kim DJ, Suk KT. Food and Gut Microbiota-Derived Metabolites in Nonalcoholic Fatty Liver Disease. Foods 2022; 11:2703. [PMID: 36076888 PMCID: PMC9455821 DOI: 10.3390/foods11172703] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/19/2022] [Accepted: 08/31/2022] [Indexed: 11/30/2022] Open
Abstract
Diet and lifestyle are crucial factors that influence the susceptibility of humans to nonalcoholic fatty liver disease (NAFLD). Personalized diet patterns chronically affect the composition and activity of microbiota in the human gut; consequently, nutrition-related dysbiosis exacerbates NAFLD via the gut-liver axis. Recent advances in diagnostic technology for gut microbes and microbiota-derived metabolites have led to advances in the diagnosis, treatment, and prognosis of NAFLD. Microbiota-derived metabolites, including tryptophan, short-chain fatty acid, fat, fructose, or bile acid, regulate the pathophysiology of NAFLD. The microbiota metabolize nutrients, and metabolites are closely related to the development of NAFLD. In this review, we discuss the influence of nutrients, gut microbes, their corresponding metabolites, and metabolism in the pathogenesis of NAFLD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Ki Tae Suk
- Institute for Liver and Digestive Diseases, Hallym University College of Medicine, Chuncheon 24252, Korea
| |
Collapse
|
15
|
Dietary fatty acid metabolism: New insights into the similarities of lipid metabolism in humans and hamsters. FOOD CHEMISTRY: MOLECULAR SCIENCES 2022; 4:100060. [PMID: 35415688 PMCID: PMC8991696 DOI: 10.1016/j.fochms.2021.100060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/25/2021] [Accepted: 11/27/2021] [Indexed: 11/21/2022]
|
16
|
Ranaivo H, Zhang Z, Alligier M, Van Den Berghe L, Sothier M, Lambert-Porcheron S, Feugier N, Cuerq C, Machon C, Neyrinck AM, Seethaler B, Rodriguez J, Roumain M, Muccioli GG, Maquet V, Laville M, Bischoff SC, Walter J, Delzenne NM, Nazare JA. Chitin-glucan supplementation improved postprandial metabolism and altered gut microbiota in subjects at cardiometabolic risk in a randomized trial. Sci Rep 2022; 12:8830. [PMID: 35614185 PMCID: PMC9132890 DOI: 10.1038/s41598-022-12920-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/05/2022] [Indexed: 01/04/2023] Open
Abstract
Chitin-glucan (CG), an insoluble dietary fiber, has been shown to improve cardiometabolic disorders associated with obesity in mice. Its effects in healthy subjects has recently been studied, revealing its interaction with the gut microbiota. In this double-blind, randomized, cross-over, twice 3-week exploratory study, we investigated the impacts of CG on the cardiometabolic profile and gut microbiota composition and functions in 15 subjects at cardiometabolic risk. They consumed as a supplement 4.5 g of CG daily or maltodextrin as control. Before and after interventions, fasting and postprandial metabolic parameters and exhaled gases (hydrogen [H2] and methane [CH4]) were evaluated. Gut microbiota composition (16S rRNA gene sequencing analysis), fecal concentrations of bile acids, long- and short-chain fatty acids (LCFA, SCFA), zonulin, calprotectin and lipopolysaccharide binding protein (LBP) were analyzed. Compared to control, CG supplementation increased exhaled H2 following an enriched-fiber breakfast ingestion and decreased postprandial glycemia and triglyceridemia response to a standardized test meal challenge served at lunch. Of note, the decrease in postprandial glycemia was only observed in subjects with higher exhaled H2, assessed upon lactulose breath test performed at inclusion. CG decreased a family belonging to Actinobacteria phylum and increased 3 bacterial taxa: Erysipelotrichaceae UCG.003, Ruminococcaceae UCG.005 and Eubacterium ventriosum group. Fecal metabolites, inflammatory and intestinal permeability markers did not differ between groups. In conclusion, we showed that CG supplementation modified the gut microbiota composition and improved postprandial glycemic response, an early determinant of cardiometabolic risk. Our results also suggest breath H2 production as a non-invasive parameter of interest for predicting the effectiveness of dietary fiber intervention.
Collapse
Affiliation(s)
- Harimalala Ranaivo
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Pierre-Bénite, France
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France
| | - Zhengxiao Zhang
- College of Food and Biological Engineering, Jimei University, Xiamen, Fujian, China
| | - Maud Alligier
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Pierre-Bénite, France
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France
| | - Laurie Van Den Berghe
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Pierre-Bénite, France
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France
| | - Monique Sothier
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Pierre-Bénite, France
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France
| | - Stéphanie Lambert-Porcheron
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Pierre-Bénite, France
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France
| | - Nathalie Feugier
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Pierre-Bénite, France
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France
| | - Charlotte Cuerq
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France
- Service de Biochimie et Biologie Moléculaire, Unité Médicale Dyslipidémies et Dysfonctions Nutritionnelles et Digestives, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Christelle Machon
- Hospices Civils de Lyon, Service de Biochimie, Centre de Biologie Sud, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Benjamin Seethaler
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Martin Roumain
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Véronique Maquet
- KitoZyme, Parc Industriel des Hauts-Sart, Zone 2, Rue de Milmort 680, 4040, Herstal, Belgium
| | - Martine Laville
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Pierre-Bénite, France
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Jens Walter
- Department of Medicine, and School of Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Julie-Anne Nazare
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, CENS, FCRIN/FORCE Network, Pierre-Bénite, France.
- Univ-Lyon, CarMeN Laboratory, INSERM, INRAE, Université Claude Bernard Lyon-1, 69600, Oullins, France.
| |
Collapse
|
17
|
Gao B, Zeng S, Maccioni L, Shi X, Armando A, Quehenberger O, Zhang X, Stärkel P, Schnabl B. Lipidomics for the Prediction of Progressive Liver Disease in Patients with Alcohol Use Disorder. Metabolites 2022; 12:433. [PMID: 35629937 PMCID: PMC9146183 DOI: 10.3390/metabo12050433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 02/04/2023] Open
Abstract
Alcohol-related liver disease is a public health care burden globally. Only 10-20% of patients with alcohol use disorder have progressive liver disease. This study aimed to identify lipid biomarkers for the early identification of progressive alcohol-related liver disease, which is a key step for early intervention. We performed untargeted lipidomics analysis in serum and fecal samples for a cohort of 49 subjects, including 17 non-alcoholic controls, 16 patients with non-progressive alcohol-related liver disease, and 16 patients with progressive alcohol-related liver disease. The serum and fecal lipidome profiles in the two patient groups were different from that in the controls. Nine lipid biomarkers were identified that were significantly different between patients with progressive liver disease and patients with non-progressive liver disease in both serum and fecal samples. We further built a random forest model to predict progressive alcohol-related liver disease using nine lipid biomarkers. Fecal lipids performed better (Area Under the Curve, AUC = 0.90) than serum lipids (AUC = 0.79). The lipid biomarkers identified are promising candidates for the early identification of progressive alcohol-related liver disease.
Collapse
Affiliation(s)
- Bei Gao
- School of Marine Sciences, Nanjing University of Information Science and Technology, Nanjing 210044, China;
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (S.Z.); (A.A.); (O.Q.)
| | - Suling Zeng
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (S.Z.); (A.A.); (O.Q.)
| | - Luca Maccioni
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Xiaochun Shi
- School of Environmental Science and Engineering, Nanjing University of Information Science and Technology, Nanjing 210044, China;
| | - Aaron Armando
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (S.Z.); (A.A.); (O.Q.)
| | - Oswald Quehenberger
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (S.Z.); (A.A.); (O.Q.)
| | - Xinlian Zhang
- Division of Biostatistics and Bioinformatics, Department of Family Medicine and Public Health, University of California San Diego, La Jolla, CA 92093, USA;
| | - Peter Stärkel
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium;
- St. Luc University Hospital, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (S.Z.); (A.A.); (O.Q.)
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA 92161, USA
| |
Collapse
|
18
|
Lee AH, Manly A, Dong TS. Leveraging the Microbiome for Obesity: Moving From Form to Function. Front Endocrinol (Lausanne) 2022; 13:918923. [PMID: 35873002 PMCID: PMC9300920 DOI: 10.3389/fendo.2022.918923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/13/2022] [Indexed: 11/24/2022] Open
Abstract
Treatment of obesity, an ongoing global epidemic, is challenging, as weight-loss efforts require a multidisciplinary approach addressing both behavioral and biologic needs that are not completely understood. Recent studies of the gut microbiome may provide better insight into the condition, and ultimately serve to advance more effective therapies. Research in this field has shifted from analyzing microbiome compositional differences to investigating functional changes that affect disease pathophysiology and outcome. Bacteria-derived metabolites are a way to bridge compositional changes to functional consequences. Through the production of metabolites, such as short chain fatty acids, tryptophan derivatives and bile acids, and interactions with peripheral and central signaling pathways, the gut microbiome may alter the body's metabolic and behavioral responses to food. Here, we summarize these mechanisms driven by gut-derived metabolites, through which the microbiome is thought to contribute to obesity, as well as review recent investigations of interventions related to these metabolites. Limitations of existing research, primarily due to paucity of causal studies in humans, are also discussed in this review.
Collapse
Affiliation(s)
- Anna H. Lee
- Department of Internal Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- *Correspondence: Anna H. Lee,
| | - Amanda Manly
- Department of Medicine, Garden City Hospital, Garden City, MI, United States
| | - Tien S. Dong
- Department of Gastroenterology, Greater Los Angeles Veterans Affairs, Los Angeles, CA, United States
- Vatche & Tamar Manoukian Division of Digestive Diseases, Los Angeles, CA, United States
| |
Collapse
|
19
|
Larroya A, Pantoja J, Codoñer-Franch P, Cenit MC. Towards Tailored Gut Microbiome-Based and Dietary Interventions for Promoting the Development and Maintenance of a Healthy Brain. Front Pediatr 2021; 9:705859. [PMID: 34277527 PMCID: PMC8280474 DOI: 10.3389/fped.2021.705859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 05/31/2021] [Indexed: 01/07/2023] Open
Abstract
Mental health is determined by a complex interplay between the Neurological Exposome and the Human Genome. Multiple genetic and non-genetic (exposome) factors interact early in life, modulating the risk of developing the most common complex neurodevelopmental disorders (NDDs), with potential long-term consequences on health. To date, the understating of the precise etiology underpinning these neurological alterations, and their clinical management pose a challenge. The crucial role played by diet and gut microbiota in brain development and functioning would indicate that modulating the gut-brain axis may help protect against the onset and progression of mental-health disorders. Some nutritional deficiencies and gut microbiota alterations have been linked to NDDs, suggesting their potential pathogenic implications. In addition, certain dietary interventions have emerged as promising alternatives or adjuvant strategies for improving the management of particular NDDs, at least in particular subsets of subjects. The gut microbiota can be a key to mediating the effects of other exposome factors such as diet on mental health, and ongoing research in Psychiatry and Neuropediatrics is developing Precision Nutrition Models to classify subjects according to a diet response prediction based on specific individual features, including microbiome signatures. Here, we review current scientific evidence for the impact of early life environmental factors, including diet, on gut microbiota and neuro-development, emphasizing the potential long-term consequences on health; and also summarize the state of the art regarding the mechanisms underlying diet and gut microbiota influence on the brain-gut axis. Furthermore, we describe the evidence supporting the key role played by gut microbiota, diet and nutrition in neurodevelopment, as well as the effectiveness of certain dietary and microbiome-based interventions aimed at preventing or treating NDDs. Finally, we emphasize the need for further research to gain greater insight into the complex interplay between diet, gut microbiome and brain development. Such knowledge would help towards achieving tailored integrative treatments, including personalized nutrition.
Collapse
Affiliation(s)
- Ana Larroya
- Microbial Ecology, Nutrition & Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Jorge Pantoja
- Department of Pediatrics, University Hospital De la Plana, Vila-Real, Castellón, Spain
- Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Pilar Codoñer-Franch
- Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
- Department of Pediatrics, Dr. Peset University Hospital, Valencia, Spain
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain
| | - María Carmen Cenit
- Microbial Ecology, Nutrition & Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
- Department of Pediatrics, University Hospital De la Plana, Vila-Real, Castellón, Spain
- Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| |
Collapse
|
20
|
Ávila-Román J, Arreaza-Gil V, Cortés-Espinar AJ, Soliz-Rueda JR, Mulero M, Muguerza B, Arola-Arnal A, Arola L, Torres-Fuentes C. Impact of gut microbiota on plasma oxylipins profile under healthy and obesogenic conditions. Clin Nutr 2021; 40:1475-1486. [PMID: 33743282 DOI: 10.1016/j.clnu.2021.02.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/03/2021] [Accepted: 02/17/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Oxylipins (OXLs) are bioactive lipid metabolites derived from polyunsaturated fatty acids (PUFAs) which act as signaling molecules and are involved in inflammatory processes such as those that occur in obesity. On the other hand, gut microbiota plays an essential role in regulating inflammatory responses. However, little is known about the potential impact of gut bacteria on OXLs metabolism. Thus, the objective of this study was to investigate the effect of gut microbiota dysbiosis on plasma oxylipins profile in healthy and diet-induced obese animals. METHODS Eight-week-old male Wistar rats were fed with either a standard or cafeteria diet (CAF) for 5 weeks and administered an antibiotic cocktail (ABX) in the drinking water (Ampicillin: 1 g/ml, Vancomycin: 0.5 g/ml, Imipenem: 0.25 g/ml) for the last 2 weeks in order to induce gut microbiota dysbiosis. Metabolomics analysis of OXLs in plasma was performed by HPLC-MS analysis. No antibiotic treated animals were included as controls. RESULTS Plasma OXLs profile was significantly altered due to both CAF feeding and ABX administration. ABX effect was more pronounced under obesogenic conditions. Several significant correlations between different bacteria taxa and these lipid mediators were observed. Among these, the positive correlation of Proteobacteria with LTB4, a proinflammatory OXL involved in obesity-related disorders, was especially remarkable. CONCLUSIONS Gut microbiota plays a key role in regulating these lipid metabolites and, therefore, affecting oxylipins-mediated inflammatory processes. These results are the first evidence to our knowledge of gut microbiota impact on OXLs metabolism. Moreover, this can set the basis for developing new obesity markers based on OXLs and gut microbiota profiles.
Collapse
Affiliation(s)
- Javier Ávila-Román
- Universitat Rovira i Virgili, Departament de Bioquimica i Biotecnologia, Nutrigenomics Research Group, 43007, Tarragona, Spain.
| | - Verónica Arreaza-Gil
- Universitat Rovira i Virgili, Departament de Bioquimica i Biotecnologia, Nutrigenomics Research Group, 43007, Tarragona, Spain.
| | - Antonio J Cortés-Espinar
- Universitat Rovira i Virgili, Departament de Bioquimica i Biotecnologia, Nutrigenomics Research Group, 43007, Tarragona, Spain.
| | - Jorge R Soliz-Rueda
- Universitat Rovira i Virgili, Departament de Bioquimica i Biotecnologia, Nutrigenomics Research Group, 43007, Tarragona, Spain.
| | - Miquel Mulero
- Universitat Rovira i Virgili, Departament de Bioquimica i Biotecnologia, Nutrigenomics Research Group, 43007, Tarragona, Spain.
| | - Begoña Muguerza
- Universitat Rovira i Virgili, Departament de Bioquimica i Biotecnologia, Nutrigenomics Research Group, 43007, Tarragona, Spain.
| | - Anna Arola-Arnal
- Universitat Rovira i Virgili, Departament de Bioquimica i Biotecnologia, Nutrigenomics Research Group, 43007, Tarragona, Spain.
| | - Lluís Arola
- Universitat Rovira i Virgili, Departament de Bioquimica i Biotecnologia, Nutrigenomics Research Group, 43007, Tarragona, Spain.
| | - Cristina Torres-Fuentes
- Universitat Rovira i Virgili, Departament de Bioquimica i Biotecnologia, Nutrigenomics Research Group, 43007, Tarragona, Spain.
| |
Collapse
|
21
|
García-Montero C, Fraile-Martínez O, Gómez-Lahoz AM, Pekarek L, Castellanos AJ, Noguerales-Fraguas F, Coca S, Guijarro LG, García-Honduvilla N, Asúnsolo A, Sanchez-Trujillo L, Lahera G, Bujan J, Monserrat J, Álvarez-Mon M, Álvarez-Mon MA, Ortega MA. Nutritional Components in Western Diet Versus Mediterranean Diet at the Gut Microbiota-Immune System Interplay. Implications for Health and Disease. Nutrients 2021; 13:699. [PMID: 33671569 PMCID: PMC7927055 DOI: 10.3390/nu13020699] [Citation(s) in RCA: 212] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/12/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023] Open
Abstract
The most prevalent diseases of our time, non-communicable diseases (NCDs) (including obesity, type 2 diabetes, cardiovascular diseases and some types of cancer) are rising worldwide. All of them share the condition of an "inflammatory disorder", with impaired immune functions frequently caused or accompanied by alterations in gut microbiota. These multifactorial maladies also have in common malnutrition related to physiopathology. In this context, diet is the greatest modulator of immune system-microbiota crosstalk, and much interest, and new challenges, are arising in the area of precision nutrition as a way towards treatment and prevention. It is a fact that the westernized diet (WD) is partly responsible for the increased prevalence of NCDs, negatively affecting both gut microbiota and the immune system. Conversely, other nutritional approaches, such as Mediterranean diet (MD), positively influence immune system and gut microbiota, and is proposed not only as a potential tool in the clinical management of different disease conditions, but also for prevention and health promotion globally. Thus, the purpose of this review is to determine the regulatory role of nutritional components of WD and MD in the gut microbiota and immune system interplay, in order to understand, and create awareness of, the influence of diet over both key components.
Collapse
Affiliation(s)
- Cielo García-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (A.M.G.-L.); (L.P.); (A.J.C.); (N.G.-H.); (J.B.); (J.M.)
| | - Oscar Fraile-Martínez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (A.M.G.-L.); (L.P.); (A.J.C.); (N.G.-H.); (J.B.); (J.M.)
| | - Ana M. Gómez-Lahoz
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (A.M.G.-L.); (L.P.); (A.J.C.); (N.G.-H.); (J.B.); (J.M.)
| | - Leonel Pekarek
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (A.M.G.-L.); (L.P.); (A.J.C.); (N.G.-H.); (J.B.); (J.M.)
| | - Alejandro J. Castellanos
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (A.M.G.-L.); (L.P.); (A.J.C.); (N.G.-H.); (J.B.); (J.M.)
| | - Fernando Noguerales-Fraguas
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (F.N.-F.); (A.A.)
- Department of General Surgery, Príncipe de Asturias Hospital, 28806 Alcalá de Henares, Spain
| | - Santiago Coca
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (A.M.G.-L.); (L.P.); (A.J.C.); (N.G.-H.); (J.B.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.C.); (L.S.-T.)
- University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
| | - Luis G. Guijarro
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.C.); (L.S.-T.)
- Unit of Biochemistry and Molecular Biology (CIBEREHD), Department of System Biology, University of Alcalá, 28801 Alcalá de Henares, Spain;
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (A.M.G.-L.); (L.P.); (A.J.C.); (N.G.-H.); (J.B.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.C.); (L.S.-T.)
- University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
| | - Angel Asúnsolo
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (F.N.-F.); (A.A.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.C.); (L.S.-T.)
| | - Lara Sanchez-Trujillo
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (A.M.G.-L.); (L.P.); (A.J.C.); (N.G.-H.); (J.B.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.C.); (L.S.-T.)
- Service of Pediatric, Hospital Universitario Principe de Asturias, Alcalá de Henares,28806 Madrid, Spain
| | - Guillermo Lahera
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (A.M.G.-L.); (L.P.); (A.J.C.); (N.G.-H.); (J.B.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.C.); (L.S.-T.)
- Psychiatry Service, Center for Biomedical Research in the Mental Health Network, University Hospital Príncipe de Asturias, 28806 Alcalá de Henares, Spain;
| | - Julia Bujan
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (A.M.G.-L.); (L.P.); (A.J.C.); (N.G.-H.); (J.B.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.C.); (L.S.-T.)
- University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
| | - Jorge Monserrat
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (A.M.G.-L.); (L.P.); (A.J.C.); (N.G.-H.); (J.B.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.C.); (L.S.-T.)
- University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (A.M.G.-L.); (L.P.); (A.J.C.); (N.G.-H.); (J.B.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.C.); (L.S.-T.)
- University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine, University Hospital Príncipe de Asturias, (CIBEREHD), 28806 Alcalá de Henares, Spain;
| | - Miguel A. Álvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (A.M.G.-L.); (L.P.); (A.J.C.); (N.G.-H.); (J.B.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.C.); (L.S.-T.)
- University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
- Department of Psychiatry and Medical Psychology, Hospital Universitario Infanta Leonor, 28031 Madrid, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (A.M.G.-L.); (L.P.); (A.J.C.); (N.G.-H.); (J.B.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.C.); (L.S.-T.)
- University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, 28806 Alcalá de Henares, Spain;
| |
Collapse
|
22
|
Serum Acylcarnitines Associated with High Short-Term Mortality in Patients with Alcoholic Hepatitis. Biomolecules 2021; 11:biom11020281. [PMID: 33672832 PMCID: PMC7917657 DOI: 10.3390/biom11020281] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 02/10/2021] [Indexed: 01/11/2023] Open
Abstract
Alcohol-related liver disease is one of the most prevalent liver diseases in the United States. Early stages of alcohol-related liver disease are characterized by accumulation of triglycerides in hepatocytes. Alcoholic hepatitis is a severe form of alcohol-related liver disease associated with significant morbidity and mortality. We sought to identify patients who are at greatest risk of death using serum lipids. First, we performed lipidomics analysis on serum samples collected from 118 patients with alcoholic hepatitis to identify lipid markers that are associated with high risk of death. Next, we performed gene set enrichment analysis on liver transcriptomics data to identify dysregulated lipid metabolism in patients who received liver transplantation. Finally, we built a random forest model to predict 30-day mortality using serum lipids. A total of 277 lipids were annotated in the serum of patients with alcoholic hepatitis, among which 25 were significantly different between patients in the deceased and alive groups. Five chemical clusters were significantly altered between the two groups. In particular, acylcarnitine cluster was enriched in the deceased group. Several hepatic lipid metabolism pathways were dysregulated in patients with alcoholic hepatitis who received liver transplantation. The mRNA expression of genes involved in the fatty acid transport into mitochondria and β-oxidation were also dysregulated. When predicting 30-day mortality in alcoholic hepatitis patients using serum lipids, we found that the area under the curve achieved 0.95. Serum lipids such as acylcarnitines may serve as biomarkers to identify alcoholic hepatitis patients at the greatest risk of death.
Collapse
|
23
|
Neyrinck AM, Rodriguez J, Zhang Z, Seethaler B, Sánchez CR, Roumain M, Hiel S, Bindels LB, Cani PD, Paquot N, Cnop M, Nazare JA, Laville M, Muccioli GG, Bischoff SC, Walter J, Thissen JP, Delzenne NM. Prebiotic dietary fibre intervention improves fecal markers related to inflammation in obese patients: results from the Food4Gut randomized placebo-controlled trial. Eur J Nutr 2021; 60:3159-3170. [PMID: 33544206 PMCID: PMC8354918 DOI: 10.1007/s00394-021-02484-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/08/2021] [Indexed: 02/07/2023]
Abstract
Purpose Inulin-type fructans (ITF) are prebiotic dietary fibre (DF) that may confer beneficial health effects, by interacting with the gut microbiota. We have tested the hypothesis that a dietary intervention promoting inulin intake versus placebo influences fecal microbial-derived metabolites and markers related to gut integrity and inflammation in obese patients. Methods Microbiota (16S rRNA sequencing), long- and short-chain fatty acids (LCFA, SCFA), bile acids, zonulin, and calprotectin were analyzed in fecal samples obtained from obese patients included in a randomized, placebo-controlled trial. Participants received either 16 g/d native inulin (prebiotic n = 12) versus maltodextrin (placebo n = 12), coupled to dietary advice to consume inulin-rich versus inulin-poor vegetables for 3 months, in addition to dietary caloric restriction. Results Both placebo and prebiotic interventions lowered energy and protein intake. A substantial increase in Bifidobacterium was detected after ITF treatment (q = 0.049) supporting our recent data obtained in a larger cohort. Interestingly, fecal calprotectin, a marker of gut inflammation, was reduced upon ITF treatment. Both prebiotic and placebo interventions increased the ratio of tauro-conjugated/free bile acids in feces. Prebiotic treatment did not significantly modify fecal SCFA content but it increased fecal rumenic acid, a conjugated linoleic acid (cis-9, trans-11 CLA) with immunomodulatory properties, that correlated notably to the expansion of Bifidobacterium (p = 0.031; r = 0.052). Conclusions Our study demonstrates that ITF-prebiotic intake during 3 months decreases a fecal marker of intestinal inflammation in obese patients. Our data point to a potential contribution of microbial lipid-derived metabolites in gastro-intestinal dysfunction related to obesity. ClinicalTrials.gov Identifier NCT03852069 (February 22, 2019 retrospectively, registered). Supplementary Information The online version contains supplementary material available at 10.1007/s00394-021-02484-5.
Collapse
Affiliation(s)
- Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, avenue E. Mounier box B1.73.11, B-1200, Brussels, Belgium
| | - Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, avenue E. Mounier box B1.73.11, B-1200, Brussels, Belgium
| | - Zhengxiao Zhang
- Department of Medicine, University of Alberta, Edmonton, Canada
| | - Benjamin Seethaler
- Institute of Nutritional Medicine, University of Hohenheim, Hohenheim, Germany
| | - Cándido Robles Sánchez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, avenue E. Mounier box B1.73.11, B-1200, Brussels, Belgium
| | - Martin Roumain
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Sophie Hiel
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, avenue E. Mounier box B1.73.11, B-1200, Brussels, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, avenue E. Mounier box B1.73.11, B-1200, Brussels, Belgium
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, avenue E. Mounier box B1.73.11, B-1200, Brussels, Belgium.,WELBIO- Walloon Excellence in Life Sciences and Biotechnology, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Nicolas Paquot
- Laboratory of Diabetology, Nutrition and Metabolic Disease, Université de Liège, Liège, Belgium
| | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium.,Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Julie-Anne Nazare
- Rhône-Alpes Research Center for Human Nutrition, Université-Lyon, CarMeN Laboratory, Hospices Civils de Lyon, Lyon, France
| | - Martine Laville
- Rhône-Alpes Research Center for Human Nutrition, Université-Lyon, CarMeN Laboratory, Hospices Civils de Lyon, Lyon, France
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Hohenheim, Germany
| | - Jens Walter
- Department of Medicine, University of Alberta, Edmonton, Canada.,Department of Medicine, and School of Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jean-Paul Thissen
- Pole of Endocrinology, Diabetes and Nutrition, Institut de Recherche Expérimentale et Clinique, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, avenue E. Mounier box B1.73.11, B-1200, Brussels, Belgium.
| |
Collapse
|
24
|
Lamichhane S, Sen P, Alves MA, Ribeiro HC, Raunioniemi P, Hyötyläinen T, Orešič M. Linking Gut Microbiome and Lipid Metabolism: Moving beyond Associations. Metabolites 2021; 11:55. [PMID: 33467644 PMCID: PMC7830997 DOI: 10.3390/metabo11010055] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 02/08/2023] Open
Abstract
Various studies aiming to elucidate the role of the gut microbiome-metabolome co-axis in health and disease have primarily focused on water-soluble polar metabolites, whilst non-polar microbial lipids have received less attention. The concept of microbiota-dependent lipid biotransformation is over a century old. However, only recently, several studies have shown how microbial lipids alter intestinal and circulating lipid concentrations in the host, thus impacting human lipid homeostasis. There is emerging evidence that gut microbial communities play a particularly significant role in the regulation of host cholesterol and sphingolipid homeostasis. Here, we review and discuss recent research focusing on microbe-host-lipid co-metabolism. We also discuss the interplay of human gut microbiota and molecular lipids entering host systemic circulation, and its role in health and disease.
Collapse
Affiliation(s)
- Santosh Lamichhane
- Turku Bioscience Centre, University of Turku and Abo Akademi University, FI-20520 Turku, Finland; (P.S.); (M.A.A.); (H.C.R.); (P.R.); (M.O.)
| | - Partho Sen
- Turku Bioscience Centre, University of Turku and Abo Akademi University, FI-20520 Turku, Finland; (P.S.); (M.A.A.); (H.C.R.); (P.R.); (M.O.)
- School of Medical Sciences, Orebro University, 702 81 Orebro, Sweden
| | - Marina Amaral Alves
- Turku Bioscience Centre, University of Turku and Abo Akademi University, FI-20520 Turku, Finland; (P.S.); (M.A.A.); (H.C.R.); (P.R.); (M.O.)
| | - Henrique C. Ribeiro
- Turku Bioscience Centre, University of Turku and Abo Akademi University, FI-20520 Turku, Finland; (P.S.); (M.A.A.); (H.C.R.); (P.R.); (M.O.)
| | - Peppi Raunioniemi
- Turku Bioscience Centre, University of Turku and Abo Akademi University, FI-20520 Turku, Finland; (P.S.); (M.A.A.); (H.C.R.); (P.R.); (M.O.)
| | | | - Matej Orešič
- Turku Bioscience Centre, University of Turku and Abo Akademi University, FI-20520 Turku, Finland; (P.S.); (M.A.A.); (H.C.R.); (P.R.); (M.O.)
- School of Medical Sciences, Orebro University, 702 81 Orebro, Sweden
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China
| |
Collapse
|
25
|
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: 149] [Impact Index Per Article: 37.3] [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
|
26
|
Bacterial Metabolites of Human Gut Microbiota Correlating with Depression. Int J Mol Sci 2020; 21:ijms21239234. [PMID: 33287416 PMCID: PMC7730936 DOI: 10.3390/ijms21239234] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/23/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023] Open
Abstract
Depression is a global threat to mental health that affects around 264 million people worldwide. Despite the considerable evolution in our understanding of the pathophysiology of depression, no reliable biomarkers that have contributed to objective diagnoses and clinical therapy currently exist. The discovery of the microbiota-gut-brain axis induced scientists to study the role of gut microbiota (GM) in the pathogenesis of depression. Over the last decade, many of studies were conducted in this field. The productions of metabolites and compounds with neuroactive and immunomodulatory properties among mechanisms such as the mediating effects of the GM on the brain, have been identified. This comprehensive review was focused on low molecular weight compounds implicated in depression as potential products of the GM. The other possible mechanisms of GM involvement in depression were presented, as well as changes in the composition of the microbiota of patients with depression. In conclusion, the therapeutic potential of functional foods and psychobiotics in relieving depression were considered. The described biomarkers associated with GM could potentially enhance the diagnostic criteria for depressive disorders in clinical practice and represent a potential future diagnostic tool based on metagenomic technologies for assessing the development of depressive disorders.
Collapse
|
27
|
Abstract
As human populations spread across the world, they adapted genetically to local conditions. So too did the resident microorganism communities that everyone carries with them. However, the collective influence of the diverse and dynamic community of resident microbes on host evolution is poorly understood. The taxonomic composition of the microbiota varies among individuals and displays a range of sometimes redundant functions that modify the physicochemical environment of the host and may alter selection pressures. Here we review known human traits and genes for which the microbiota may have contributed or responded to changes in host diet, climate, or pathogen exposure. Integrating host–microbiota interactions in human adaptation could offer new approaches to improve our understanding of human health and evolution.
Collapse
Affiliation(s)
- Taichi A. Suzuki
- Department of Microbiome Science, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Ruth E. Ley
- Department of Microbiome Science, Max Planck Institute for Developmental Biology, Tübingen, Germany
| |
Collapse
|
28
|
Rodriguez J, Neyrinck AM, Zhang Z, Seethaler B, Nazare JA, Robles Sánchez C, Roumain M, Muccioli GG, Bindels LB, Cani PD, Maquet V, Laville M, Bischoff SC, Walter J, Delzenne NM. Metabolite profiling reveals the interaction of chitin-glucan with the gut microbiota. Gut Microbes 2020; 12:1810530. [PMID: 32893709 PMCID: PMC7524357 DOI: 10.1080/19490976.2020.1810530] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Dietary fibers are considered beneficial nutrients for health. Current data suggest that their interaction with the gut microbiota largely contributes to their physiological effects. In this context, chitin-glucan (CG) improves metabolic disorders associated with obesity in mice, but its effect on gut microbiota has never been evaluated in humans. This study explores the effect of a 3-week intervention with CG supplementation in healthy individuals on gut microbiota composition and bacterial metabolites. CG was given to healthy volunteers (n = 15) for three weeks as a supplement (4.5 g/day). Food diary, visual analog and Bristol stool form scales and a "quality of life" survey were analyzed. Among gut microbiota-derived metabolites, bile acids (BA), long- and short-chain fatty acids (LCFA, SCFA) profiling were assessed in stool samples. The gut microbiota (primary outcome) was analyzed by Illumina sequencing. A 3-week supplementation with CG is well tolerated in healthy humans. CG induces specific changes in the gut microbiota composition, with Eubacterium, Dorea and Roseburia genera showing the strongest regulation. In addition, CG increased bacterial metabolites in feces including butyric, iso-valeric, caproic and vaccenic acids. No major changes were observed for the fecal BA profile following CG intervention. In summary, our work reveals new potential bacterial genera and gut microbiota-derived metabolites characterizing the interaction between an insoluble dietary fiber -CG- and the gut microbiota.
Collapse
Affiliation(s)
- Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Audrey M. Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Zhengxiao Zhang
- Department of Medicine, University of Alberta, Edmonton, Canada
| | - Benjamin Seethaler
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Julie-Anne Nazare
- Rhône-Alpes Research Center for Human Nutrition, Université-Lyon, CarMeN Laboratory, Hospices Civils de Lyon, Lyon, France
| | - Cándido Robles Sánchez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Martin Roumain
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Giulio G. Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Laure B. Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Patrice D. Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium,WELBIO- Walloon Excellence in Life Sciences and BIOtechnology, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | | | - Martine Laville
- Rhône-Alpes Research Center for Human Nutrition, Université-Lyon, CarMeN Laboratory, Hospices Civils de Lyon, Lyon, France
| | - Stephan C. Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Jens Walter
- Department of Agricultural, Food & Nutritional Science and Department of Biological Sciences, University of Alberta, Edmonton, Canada,APC Microbiome Ireland, Department of Medicine, and School of Microbiology, University College Cork, Cork, Ireland
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium,CONTACT Nathalie M. Delzenne Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, B-1200Brussels, Belgium
| |
Collapse
|
29
|
Tsukidate T, Li Q, Hang HC. Nuclear Receptor Chemical Reporter Enables Domain-Specific Analysis of Ligands in Mammalian Cells. ACS Chem Biol 2020; 15:2324-2330. [PMID: 32909738 DOI: 10.1021/acschembio.0c00432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The characterization of specific metabolite-protein interactions is important in chemical biology and drug discovery. For example, nuclear receptors (NRs) are a family of ligand-activated transcription factors that regulate diverse physiological processes in animals and are key targets for therapeutic development. However, the identification and characterization of physiological ligands for many NRs remains challenging, because of limitations in domain-specific analysis of ligand binding in cells. To address these limitations, we developed a domain-specific covalent chemical reporter for peroxisome proliferator-activated receptors (PPARs) and demonstrated its utility to screen and characterize the potency of candidate NR ligands in live cells. These studies demonstrate targeted and domain-specific chemical reporters provide excellent tools to evaluate endogenous and exogenous (diet, microbiota, therapeutics) ligands of PPARs in mammalian cells, as well as additional protein targets for further investigation.
Collapse
Affiliation(s)
- Taku Tsukidate
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York, New York 10065, United States
| | - Qiang Li
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York, New York 10065, United States
| | - Howard C. Hang
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York, New York 10065, United States
- Departments of Immunology and Microbiology, Chemistry, Scripps Research, La Jolla, California 92037, United States
| |
Collapse
|
30
|
García-Burgos M, Moreno-Fernández J, Alférez MJ, Díaz-Castro J, López-Aliaga I. New perspectives in fermented dairy products and their health relevance. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104059] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
31
|
He X, McClorry S, Hernell O, Lönnerdal B, Slupsky CM. Digestion of human milk fat in healthy infants. Nutr Res 2020; 83:15-29. [PMID: 32987285 DOI: 10.1016/j.nutres.2020.08.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 08/02/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023]
Abstract
Lipid digestion is critical for infant development, and yet, the interconnection between lipid digestion and the microbiota is largely understudied. This review focuses on digestion of the human milk fat globule and summarizes the current understanding of the mechanisms underlying this process in infants. We first discuss the partial hydrolysis of milk fat in the stomach, which leads to rearrangement of lipid droplets, creating a lipid-water interface necessary for duodenal lipolysis. In the first few months of life, secretion of pancreatic triglyceride lipase, phospholipase A2, and bile salts is immature. The dominant lipases aiding fat digestion in the newborn small intestine are therefore pancreatic lipase-related protein 2 and bile salt-stimulated lipase from both the exocrine pancreas and milk. We summarize the interaction between ionic fatty acids and cations to form insoluble fatty acid soaps and how it is influenced by various factors, including cation availability, pH, and bile salt concentration, as well as saturation and chain length of fatty acids. We further argue that the formation of the soap complex does not contribute to lipid bioavailability. Next, the possible roles that the gut microbiota plays in lipid digestion and absorption are discussed. Finally, we provide a perspective on how the manufacturing process of infant formula and dairy products may alter the physical properties and structure of lipid droplets, thereby altering the rate of lipolysis.
Collapse
Affiliation(s)
- Xuan He
- Department of Nutrition, Davis, One Shields Ave, Davis, CA 95616, USA; Department of Food Science and Technology, University of California, Davis, One Shields Ave, Davis, CA 95616, USA
| | - Shannon McClorry
- Department of Nutrition, Davis, One Shields Ave, Davis, CA 95616, USA
| | - Olle Hernell
- Department of Clinical Sciences, Pediatrics, Umeå University, SE 901 85 Umeå, Sweden
| | - Bo Lönnerdal
- Department of Nutrition, Davis, One Shields Ave, Davis, CA 95616, USA
| | - Carolyn M Slupsky
- Department of Nutrition, Davis, One Shields Ave, Davis, CA 95616, USA; Department of Food Science and Technology, University of California, Davis, One Shields Ave, Davis, CA 95616, USA.
| |
Collapse
|
32
|
Machate DJ, Figueiredo PS, Marcelino G, Guimarães RDCA, Hiane PA, Bogo D, Pinheiro VAZ, de Oliveira LCS, Pott A. Fatty Acid Diets: Regulation of Gut Microbiota Composition and Obesity and Its Related Metabolic Dysbiosis. Int J Mol Sci 2020; 21:E4093. [PMID: 32521778 PMCID: PMC7312778 DOI: 10.3390/ijms21114093] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 02/06/2023] Open
Abstract
Long-term high-fat dietary intake plays a crucial role in the composition of gut microbiota in animal models and human subjects, which affect directly short-chain fatty acid (SCFA) production and host health. This review aims to highlight the interplay of fatty acid (FA) intake and gut microbiota composition and its interaction with hosts in health promotion and obesity prevention and its related metabolic dysbiosis. The abundance of the Bacteroidetes/Firmicutes ratio, as Actinobacteria and Proteobacteria species are associated with increased SCFA production, reported high-fat diet rich in medium-chain fatty acids (MCFAs), monounsaturated fatty acids (MUFAs), and n-3 polyunsaturated fatty acids (PUFAs) as well as low-fat diets rich in long-chain fatty acids (LCFAs). SCFAs play a key role in health promotion and prevention and, reduction and reversion of metabolic syndromes in the host. Furthermore, in this review, we discussed the type of fatty acids and their amount, including the administration time and their interplay with gut microbiota and its results about health or several metabolic dysbioses undergone by hosts.
Collapse
Affiliation(s)
- David Johane Machate
- Graduate Program in Biotechnology and Biodiversity in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; (D.J.M.); (A.P.)
| | - Priscila Silva Figueiredo
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; (P.S.F.); (G.M.); (P.A.H.); (D.B.); (V.A.Z.P.)
| | - Gabriela Marcelino
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; (P.S.F.); (G.M.); (P.A.H.); (D.B.); (V.A.Z.P.)
| | - Rita de Cássia Avellaneda Guimarães
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; (P.S.F.); (G.M.); (P.A.H.); (D.B.); (V.A.Z.P.)
| | - Priscila Aiko Hiane
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; (P.S.F.); (G.M.); (P.A.H.); (D.B.); (V.A.Z.P.)
| | - Danielle Bogo
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; (P.S.F.); (G.M.); (P.A.H.); (D.B.); (V.A.Z.P.)
| | - Verônica Assalin Zorgetto Pinheiro
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; (P.S.F.); (G.M.); (P.A.H.); (D.B.); (V.A.Z.P.)
| | | | - Arnildo Pott
- Graduate Program in Biotechnology and Biodiversity in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul, Campo Grande 79079-900, Brazil; (D.J.M.); (A.P.)
| |
Collapse
|
33
|
Peng M, Tabashsum Z, Anderson M, Truong A, Houser AK, Padilla J, Akmel A, Bhatti J, Rahaman SO, Biswas D. Effectiveness of probiotics, prebiotics, and prebiotic-like components in common functional foods. Compr Rev Food Sci Food Saf 2020; 19:1908-1933. [PMID: 33337097 DOI: 10.1111/1541-4337.12565] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/18/2020] [Accepted: 03/30/2020] [Indexed: 12/18/2022]
Abstract
The bioactive ingredients in commonly consumed foods include, but are not limited to, prebiotics, prebiotic-like components, probiotics, and postbiotics. The bioactive ingredients in functional foods have also been associated with beneficial effects on human health. For example, they aid in shaping of gut microflora and promotion of immunity. These functional components also contribute in preventing serious diseases such as cardiovascular malfunction and tumorigenesis. However, the specific mechanisms of these positive influences on human health are still under investigation. In this review, we aim to emphasize the major contents of probiotics, prebiotics, and prebiotic-like components commonly found in consumable functional foods, and we present an overview of direct and indirect benefits they provide on human health. The major contributors are certain families of metabolites, specifically short-chain fatty acids and polyunsaturated fatty acids produced by probiotics, and prebiotics, or prebiotic-like components such as flavonoids, polyphenols, and vitamins that are found in functional foods. These functional ingredients in foods influence the gut microbiota by stimulating the growth of beneficial microbes and the production of beneficial metabolites that, in turn, have direct benefits to the host, while also providing protection from pathogens and maintaining a balanced gut ecosystem. The complex interactions that arise among functional food ingredients, human physiology, the gut microbiota, and their respective metabolic pathways have been found to minimize several factors that contribute to the incidence of chronic disease, such as inflammation oxidative stress.
Collapse
Affiliation(s)
- Mengfei Peng
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Zajeba Tabashsum
- Biological Sciences Program - Molecular and Cellular Biology, University of Maryland, College Park, Maryland
| | - Mary Anderson
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Andy Truong
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Ashley K Houser
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Joselyn Padilla
- Biological Sciences Program - Molecular and Cellular Biology, University of Maryland, College Park, Maryland
| | - Ahlam Akmel
- Biological Sciences Program - Molecular and Cellular Biology, University of Maryland, College Park, Maryland
| | - Jacob Bhatti
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Shaik O Rahaman
- Department of Nutrition and Food Sciences, University of Maryland, College Park, Maryland
| | - Debabrata Biswas
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland.,Biological Sciences Program - Molecular and Cellular Biology, University of Maryland, College Park, Maryland.,Center for Food Safety and Security Systems, University of Maryland, College Park, Maryland
| |
Collapse
|
34
|
Jamar G, Ribeiro DA, Pisani LP. High-fat or high-sugar diets as trigger inflammation in the microbiota-gut-brain axis. Crit Rev Food Sci Nutr 2020; 61:836-854. [PMID: 32267169 DOI: 10.1080/10408398.2020.1747046] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Giovana Jamar
- Programa de Pós-Graduação Interdisciplinar em Ciências da Saúde, Universidade Federal de São Paulo, Santos, SP, Brazil
- Laboratório de Nutrição e Fisiologia Endócrina (LaNFE), Universidade Federal de São Paulo, Santos, SP, Brazil
| | - Daniel Araki Ribeiro
- Departamento de Biociências, Instituto de Saúde e Sociedade, Universidade Federal de São Paulo, Santos, SP, Brazil
| | - Luciana Pellegrini Pisani
- Laboratório de Nutrição e Fisiologia Endócrina (LaNFE), Universidade Federal de São Paulo, Santos, SP, Brazil
- Departamento de Biociências, Instituto de Saúde e Sociedade, Universidade Federal de São Paulo, Santos, SP, Brazil
| |
Collapse
|
35
|
Todorov H, Kollar B, Bayer F, Brandão I, Mann A, Mohr J, Pontarollo G, Formes H, Stauber R, Kittner JM, Endres K, Watzer B, Nockher WA, Sommer F, Gerber S, Reinhardt C. α-Linolenic Acid-Rich Diet Influences Microbiota Composition and Villus Morphology of the Mouse Small Intestine. Nutrients 2020; 12:nu12030732. [PMID: 32168729 PMCID: PMC7146139 DOI: 10.3390/nu12030732] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 12/14/2022] Open
Abstract
α-Linolenic acid (ALA) is well-known for its anti-inflammatory activity. In contrast, the influence of an ALA-rich diet on intestinal microbiota composition and its impact on small intestine morphology are not fully understood. In the current study, we kept adult C57BL/6J mice for 4 weeks on an ALA-rich or control diet. Characterization of the microbial composition of the small intestine revealed that the ALA diet was associated with an enrichment in Prevotella and Parabacteroides. In contrast, taxa belonging to the Firmicutes phylum, including Lactobacillus, Clostridium cluster XIVa, Lachnospiraceae and Streptococcus, had significantly lower abundance compared to control diet. Metagenome prediction indicated an enrichment in functional pathways such as bacterial secretion system in the ALA group, whereas the two-component system and ALA metabolism pathways were downregulated. We also observed increased levels of ALA and its metabolites eicosapentanoic and docosahexanoic acid, but reduced levels of arachidonic acid in the intestinal tissue of ALA-fed mice. Furthermore, intestinal morphology in the ALA group was characterized by elongated villus structures with increased counts of epithelial cells and reduced epithelial proliferation rate. Interestingly, the ALA diet reduced relative goblet and Paneth cell counts. Of note, high-fat Western-type diet feeding resulted in a comparable adaptation of the small intestine. Collectively, our study demonstrates the impact of ALA on the gut microbiome and reveals the nutritional regulation of gut morphology.
Collapse
Affiliation(s)
- Hristo Todorov
- Institute for Developmental Biology and Neurobiology, Faculty of Biology and Center for Computational Sciences in Mainz, Johannes Gutenberg-University Mainz, Staudingerweg 9, 55128 Mainz, Germany; (H.T.); (S.G.)
- Fresenius Kabi Deutschland GmbH, Borkenberg 14, 61440 Oberursel, Germany
| | - Bettina Kollar
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (B.K.); (I.B.); (A.M.); (J.M.); (G.P.)
| | - Franziska Bayer
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (B.K.); (I.B.); (A.M.); (J.M.); (G.P.)
| | - Inês Brandão
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (B.K.); (I.B.); (A.M.); (J.M.); (G.P.)
- Centro de Apoio Tecnológico Agro Alimentar (CATAA), Zona Industrial de Castelo Branco, Rua A, 6000-459 Castelo Branco, Portugal
| | - Amrit Mann
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (B.K.); (I.B.); (A.M.); (J.M.); (G.P.)
| | - Julia Mohr
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (B.K.); (I.B.); (A.M.); (J.M.); (G.P.)
| | - Giulia Pontarollo
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (B.K.); (I.B.); (A.M.); (J.M.); (G.P.)
| | - Henning Formes
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (B.K.); (I.B.); (A.M.); (J.M.); (G.P.)
| | - Roland Stauber
- Nanobiomedicine, University Medical Center Mainz, Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany;
| | - Jens M. Kittner
- Medical Department 2 (Gastroenterology, Hepatology, Pneumology, Endocrinology) Klinikum Darmstadt GmbH, Grafenstr. 9, 64283 Darmstadt, Germany;
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Bernhard Watzer
- Metabolomics Core Facility, Philipps-University, 35043 Marburg, Germany;
| | - Wolfgang Andreas Nockher
- Institute of Laboratory Medicine and Pathobiochemistry, Philipps-University, 35043 Marburg, Germany;
| | - Felix Sommer
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel, 24105 Kiel, Germany;
| | - Susanne Gerber
- Institute for Developmental Biology and Neurobiology, Faculty of Biology and Center for Computational Sciences in Mainz, Johannes Gutenberg-University Mainz, Staudingerweg 9, 55128 Mainz, Germany; (H.T.); (S.G.)
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (B.K.); (I.B.); (A.M.); (J.M.); (G.P.)
- German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, 55131 Mainz, Germany
- Correspondence: ; Tel.: +49-6131-17-8280
| |
Collapse
|
36
|
Neyrinck AM, Rodriguez J, Vinoy S, Maquet V, Walter J, Bischoff SC, Laville M, Delzenne NM. The FiberTAG project: Tagging dietary fibre intake by measuring biomarkers related to the gut microbiota and their interest for health. NUTR BULL 2020; 45:59-65. [PMID: 32194343 PMCID: PMC7074038 DOI: 10.1111/nbu.12416] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/18/2019] [Accepted: 01/02/2020] [Indexed: 12/13/2022]
Abstract
The scientific rationale for dietary fibre intake recommendations comes from the recognition of their benefits for health based on studies first published many years ago. It remains unclear which are the key physiological effects generated by dietary fibre in view of the diversity of the food components considered as dietary fibre, of the relevance of their classification (soluble and insoluble) and from the recent discoveries putting forward their interactions with the gut microbiota. The project FiberTAG (Joint Programming Initiative 'A Healthy Diet for a Healthy Life' 2017-2020 https://www.fibertag.eu/) aims to establish a set of biomarkers (markers of gut barrier function and bacterial co-metabolites including volatile compounds and lipid derivatives), measured in different biological compartments (faeces, blood or breath) linking dietary fibre intake and gut microbiota-related health effects. The FiberTAG consortium brings together academic and industrial partners from Belgium, France, Germany and Canada to share data and samples obtained from existing as well as new intervention studies in order to evaluate the relevance of such biomarkers. The FiberTAG consortium is currently working on five existing cohorts (prospective observational or nutritional interventions in healthy or obese patients), and a number of new intervention studies to analyse the effect of insoluble dietary fibre (wheat bran and chitin-glucan, provided by the industrial partners) in healthy individuals or in obese patients at high cardiometabolic risk.
Collapse
Affiliation(s)
- A. M. Neyrinck
- Metabolism and Nutrition Research GroupLouvain Drug Research InstituteUCLouvainUniversité Catholique de LouvainBrusselsBelgium
| | - J. Rodriguez
- Metabolism and Nutrition Research GroupLouvain Drug Research InstituteUCLouvainUniversité Catholique de LouvainBrusselsBelgium
| | | | | | - J. Walter
- Department of AgriculturalFood & Nutritional Science and Department of Biological SciencesUniversity of AlbertaEdmontonABCanada
| | - S. C. Bischoff
- Institute of Nutritional MedicineUniversity of HohenheimStuttgartGermany
| | - M. Laville
- Centre de Recherche en Nutrition Humaine Rhône‐AlpesUniv‐LyonUniversité Claude Bernard LyonHospices Civils de LyonCENSFCRIN/FORCE NetworkCarMeN LaboratoryLyonFrance
| | - N. M. Delzenne
- Metabolism and Nutrition Research GroupLouvain Drug Research InstituteUCLouvainUniversité Catholique de LouvainBrusselsBelgium
| |
Collapse
|
37
|
Shrestha N, Sleep SL, Cuffe JSM, Holland OJ, McAinch AJ, Dekker Nitert M, Hryciw DH. Pregnancy and diet-related changes in the maternal gut microbiota following exposure to an elevated linoleic acid diet. Am J Physiol Endocrinol Metab 2020; 318:E276-E285. [PMID: 31846371 DOI: 10.1152/ajpendo.00265.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Dietary intakes of linoleic acid (LA) have increased, including in women of reproductive age. Changes in maternal gut microbiome have been implicated in the metabolic adaptions that occur during pregnancy. We aimed to investigate whether consumption of a diet with elevated LA altered fecal microbiome diversity before and during pregnancy. Female Wistar-Kyoto rats consumed a high-LA diet (HLA: 6.21% of energy) or a low-LA diet (LLA: 1.44% of energy) for 10 wk before mating and during pregnancy. DNA was isolated from fecal samples before pregnancy [embryonic day 0 (E0)], or during pregnancy at E10 and E20. The microbiome composition was assessed with 16S rRNA sequencing. At E0, the beta-diversity of LLA and HLA groups differed with HLA rats having significantly lower abundance of the genera Akkermansia, Peptococcus, Sutterella, and Xo2d06 but higher abundance of Butyricimonas and Coprococcus. Over gestation, in LLA but not HLA rats, there was a reduction in alpha-diversity and an increase in beta-diversity. In the LLA group, the abundance of Akkermansia, Blautia, rc4.4, and Streptococcus decreased over gestation, whereas Coprococcus increased. In the HLA group; only the abundance of Butyricimonas decreased. At E20, there were no differences in alpha- and beta-diversity, and the abundance of Roseburia was significantly increased in the HLA group. In conclusion, consumption of a HLA diet alters gut microbiota composition, as does pregnancy in rats consuming a LLA diet. In pregnancy, consumption of a HLA diet does not alter gut microbiota composition.
Collapse
Affiliation(s)
- Nirajan Shrestha
- School of Medical Science, Griffith University, Southport, Queensland, Australia
| | - Simone L Sleep
- School of Medical Science, Griffith University, Southport, Queensland, Australia
| | - James S M Cuffe
- School of Medical Science, Griffith University, Southport, Queensland, Australia
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Olivia J Holland
- School of Medical Science, Griffith University, Southport, Queensland, Australia
| | - Andrew J McAinch
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
- Australian Institute for Musculoskeletal Science, Victoria University, St. Albans, Victoria, Australia
| | - Marloes Dekker Nitert
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Deanne H Hryciw
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
- School of Environment and Science, Griffith University, Nathan, Queensland, Australia
| |
Collapse
|
38
|
Zheng J, Zhang J, Guo Y, Cui H, Lin A, Hu B, Gao Q, Chen Y, Liu H. Improvement on metabolic syndrome in high fat diet-induced obese mice through modulation of gut microbiota by sangguayin decoction. JOURNAL OF ETHNOPHARMACOLOGY 2020; 246:112225. [PMID: 31509781 DOI: 10.1016/j.jep.2019.112225] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 08/23/2019] [Accepted: 09/07/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Our previous research found that Sangguayin (SGY) deccoction made by four dietary and medicinal plant components (Leaf of Morus alba L., Root of Pueraria lobata (Willd.) Ohwi., Root of Dioscorea opposita Thunb. and Fruit of Momordica charantia L.) showed significant anti-diabetic effects on db/db mice and high fat diet induced obese mice. Nevertheless, it remained unclear what the role of gut microbiota in the hypoglycaemia effects of SGY. AIMS OF THE STUDY This study aimed to examine the beneficial effects of Sangguayin Deccoction against metabolic syndrome and and its regulating role in gut microbiota and hepatic metabolome. MATERIALS AND METHODS C57BL/6J mice were divided to a normal chow diet (NCD), high-fat diet (HFD), and high-fat diet with Sangguayin Decoction (HFD-SGY, oral dose of 250 mg/kg/d) for 16 weeks. Next generation sequencing was applied for analyzing the gut microbial community of colonic contents. Further, untargeted metabolomic analysis based on LC-MS was used for determining the changes of hepatic metabolites. Hepatic genes expression were measured by quantitative PCR. RESULTS SGY supplement decreased blood glucose level and glucose intolerance. Illumina MiSeq sequencing revealed that SGY increased Verrucomicrobia phylum, resulting in a bloom of Akkermansia, and eventually upregulated the contents of Lachoclostridium and Roseburia. Additionally, dietary SGY decreased bacteria including Faecalibaculum, and Blautia. Moreover, the hepatic lipid metabolism was notably altered by SGY treatment. The oxidation of glutamione metabolism idecreasees, production of poly-unsaturated fatty acid (PUFA) got significant increase in liver tissue. The reversion of PUFA metabolism by SGY may act through PPARα mediated Fads1 and Fads2 gene expression. The altered metabolites in liver showed intimate correlatship with modified genera. CONCLUSION Data indicated that SGY reshaped gut microbial structure and improved PUFA metabolism. These functions of SGY may alter hepatic lipid metabolism, conferring preventative effects against high-fat diet induced metabolic syndrome.
Collapse
Affiliation(s)
- Junping Zheng
- College of Basic Medicine, Hubei University of Chinese Medicine, Huangjiahu 1, Wuhan, 430065, China; College of Life Sciences, Wuchang University of Technology, Jiangxia Avenue 16, Wuhan, 430223, China
| | - Jing Zhang
- College of Basic Medicine, Hubei University of Chinese Medicine, Huangjiahu 1, Wuhan, 430065, China
| | - Yanlei Guo
- Chongqing Academy of Chinese Materia Medica, Nanshan Road 34, Chongqing, 400065, China
| | - Hairong Cui
- College of Life Sciences, Wuchang University of Technology, Jiangxia Avenue 16, Wuhan, 430223, China
| | - Aizhen Lin
- Hubei Provincial Hospital of Traditional Chinese Medicine, Huayuanshan 4, Wuhan, 430061, China
| | - Baifei Hu
- College of Basic Medicine, Hubei University of Chinese Medicine, Huangjiahu 1, Wuhan, 430065, China
| | - Qinghua Gao
- College of Basic Medicine, Hubei University of Chinese Medicine, Huangjiahu 1, Wuhan, 430065, China
| | - Yunzhong Chen
- College of Basic Medicine, Hubei University of Chinese Medicine, Huangjiahu 1, Wuhan, 430065, China.
| | - Hongtao Liu
- College of Basic Medicine, Hubei University of Chinese Medicine, Huangjiahu 1, Wuhan, 430065, China; College of Life Sciences, Wuchang University of Technology, Jiangxia Avenue 16, Wuhan, 430223, China.
| |
Collapse
|
39
|
Senizza A, Callegari ML, Senizza B, Minuti A, Rocchetti G, Morelli L, Patrone V. Effects of Linoleic Acid on Gut-Derived Bifidobacterium breve DSM 20213: A Transcriptomic Approach. Microorganisms 2019; 7:microorganisms7120710. [PMID: 31861103 PMCID: PMC6955684 DOI: 10.3390/microorganisms7120710] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/12/2019] [Accepted: 12/16/2019] [Indexed: 02/08/2023] Open
Abstract
Bacterial production of conjugated linoleic acid (CLA) has recently received great attention because of the potential health benefits of this fatty acid. Linoleic acid (LA) can be converted to CLA by several microorganisms, including bifidobacteria, possibly as a detoxification mechanism to avoid the growth inhibition effect of LA. In the present in vitro study, we investigated the gene expression landscape of the intestinal strain Bifidobacterium breve DSM 20213 when exposed to LA. Transcriptomic analysis using RNA-seq revealed that LA induced a multifactorial stress response in the test strain, including upregulation of genes involved in iron uptake and downregulation of genes involved in sugar and oligopeptide transport. We also observed reduced transcription of genes involved in membrane and pili biosynthesis. The upregulation of iron uptake was not related to any putative ability of LA to chelate Fe2+, but was somewhat linked to stress response. Furthermore, we demonstrated that LA increased reactive oxygen species (ROS) production in bacterial cells, activating an oxidative stress response. This response was proved by thioredoxin reductase transcription, and was primarily evident among bacteria cultured in the absence of cysteine. This is the first report of the potential mechanisms involved in bacterial LA transport and stress response in B. breve.
Collapse
Affiliation(s)
- Alice Senizza
- Department for Sustainable Food Process (DiSTAS), Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (M.L.C.); (B.S.); (G.R.); (L.M.)
| | - Maria Luisa Callegari
- Department for Sustainable Food Process (DiSTAS), Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (M.L.C.); (B.S.); (G.R.); (L.M.)
- Biotechnology Research Centre (CRB), via Milano 24, 26100 Cremona, Italy
| | - Biancamaria Senizza
- Department for Sustainable Food Process (DiSTAS), Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (M.L.C.); (B.S.); (G.R.); (L.M.)
| | - Andrea Minuti
- Department of Animal Science, Food and Nutrition (DiANA), Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy;
- Nutrigenomics and Proteomics Research Center (PRONUTRIGEN), Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy
| | - Gabriele Rocchetti
- Department for Sustainable Food Process (DiSTAS), Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (M.L.C.); (B.S.); (G.R.); (L.M.)
| | - Lorenzo Morelli
- Department for Sustainable Food Process (DiSTAS), Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (M.L.C.); (B.S.); (G.R.); (L.M.)
- Biotechnology Research Centre (CRB), via Milano 24, 26100 Cremona, Italy
| | - Vania Patrone
- Department for Sustainable Food Process (DiSTAS), Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy; (A.S.); (M.L.C.); (B.S.); (G.R.); (L.M.)
- Nutrigenomics and Proteomics Research Center (PRONUTRIGEN), Università Cattolica del Sacro Cuore, via Emilia Parmense 84, 29122 Piacenza, Italy
- Correspondence: ; Tel.: +39-0523-599247
| |
Collapse
|
40
|
Zirnheld KH, Warner DR, Warner JB, Hardesty JE, McClain CJ, Kirpich IA. Dietary fatty acids and bioactive fatty acid metabolites in alcoholic liver disease. LIVER RESEARCH 2019. [DOI: 10.1016/j.livres.2019.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
41
|
Zhong S, Zhou Z, Liang Y, Cheng X, Li Y, Teng W, Zhao M, Liu C, Guan M, Zhao C. Targeting strategies for chemotherapy-induced peripheral neuropathy: does gut microbiota play a role? Crit Rev Microbiol 2019; 45:369-393. [PMID: 31106639 DOI: 10.1080/1040841x.2019.1608905] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a progressive, often irreversible condition that produces severe neurological deficits. Emerging data suggest that chemotherapy also exerts detrimental effects on gut microbiota composition and intestinal permeability, contributing to dysbiosis and inflammation. Compared with other complications associated with chemotherapy, such as diarrhoea and mucositis, CIPN is of particular concern because it is the most common reason for terminating or suspending treatment. However, specific and effective curative treatment strategies are lacking. In this review, we provide an update on current preclinical and clinical understandings about the role of gut microbiota in CIPN. The gut microbiota serves as an intersection between the microbiome-gut-brain and the neuroimmune-endocrine axis, forming a complex network that can directly or indirectly affect key components involved in the manifestations of CIPN. Herein, we discuss several potential mechanisms within the context of the networks and summarize alterations in gut microbiome induced by chemotherapeutic drugs, providing great potential for researchers to target pathways associated with the gut microbiome and overcome CIPN.
Collapse
Affiliation(s)
- Shanshan Zhong
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Zhike Zhou
- Department of Geriatrics, The First Hospital of China Medical University , Shenyang , PR China
| | - Yifan Liang
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Xi Cheng
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Yong Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University , Shenyang , PR China
| | - Weiyu Teng
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Mei Zhao
- Department of Cardiology, Shengjing Hospital of China Medical University , Shenyang , PR China
| | - Chang Liu
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Meiting Guan
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| | - Chuansheng Zhao
- Department of Neurology and Stroke Center, The First Hospital of China Medical University , Shenyang , PR China
| |
Collapse
|
42
|
Tao J, Li S, Gan RY, Zhao CN, Meng X, Li HB. Targeting gut microbiota with dietary components on cancer: Effects and potential mechanisms of action. Crit Rev Food Sci Nutr 2019; 60:1025-1037. [PMID: 30632784 DOI: 10.1080/10408398.2018.1555789] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancers are common chronic diseases worldwide and cause severe health burdens. There have been ongoing debates on the role of gut microbiota in the prevention and management of cancers, thus, it is worthwhile to pay high attention to the impacts of gut microbiota on several cancers, such as colon, liver, and breast cancers. In addition, it has been reported that gut microbiota may also affect the efficacy of cancer chemotherapy and immunotherapy. Among all the factors that influence gut microbiota, diet is the most influential and modifiable. The prebiotics, dietary fibers, short-chain fatty acids, and other bioactive compounds are all important dietary components to assist the growth of beneficial microbiota in the gut, which can protect against cancers and promote human health. Their beneficial effects can be due to the fermentation of dietary fibers, the metabolism of phytochemicals, the synthesis of estrogens, and interactions with chemotherapies and immunotherapies. In order to provide updated information of the relationships among dietary components, gut microbiota, and cancer, in this review, we summarize the reciprocal interactions between dietary components and gut microbiota, and highlight the impacts of dietary components on several common cancers by targeting gut microbiota, with the potential mechanisms of actions also intensively discussed. As a result, this review can be very helpful for healthy people as well as cancer patients to prevent or manage cancers via dietary factor-mediated regulation of gut microbiota.
Collapse
Affiliation(s)
- Jun Tao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Sha Li
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ren-You Gan
- Department of Food Science & Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Cai-Ning Zhao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Xiao Meng
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, China.,South China Sea Bioresource Exploitation and Utilization Collaborative Innovation Center, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
43
|
Song M, Chan AT. Environmental Factors, Gut Microbiota, and Colorectal Cancer Prevention. Clin Gastroenterol Hepatol 2019; 17:275-289. [PMID: 30031175 PMCID: PMC6314893 DOI: 10.1016/j.cgh.2018.07.012] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/02/2018] [Accepted: 07/06/2018] [Indexed: 02/07/2023]
Abstract
The substantial burden of colorectal cancer and increasing trend in young adults highlight the importance of lifestyle modification as a complement to screening for colorectal cancer prevention. Several dietary and lifestyle factors have been implicated in the development of colorectal cancer, possibly through the intricate metabolic and inflammatory mechanisms. Likewise, as a key metabolic and immune regulator, the gut microbiota has been recognized to play an important role in colorectal tumorigenesis. Increasing data support that environmental factors are crucial determinants for the gut microbial composition and function, whose alterations induce changes in the host gene expression, metabolic regulation, and local and systemic immune response, thereby influencing cancer development. Here, we review the epidemiologic and mechanistic evidence regarding the links between diet and lifestyle and the gut microbiota in the development of colorectal cancer. We focus on factors for which substantial data support their importance for colorectal cancer and their potential role in the gut microbiota, including overweight and obesity, physical activity, dietary patterns, fiber, red and processed meat, marine omega-3 fatty acid, alcohol, and smoking. We also briefly describe other colorectal cancer-preventive factors for which the links with the gut microbiota have been suggested but remain to be mechanistically characterized, including vitamin D status, dairy consumption, and metformin use. Given limitations in available evidence, we highlight the need for further investigations in the relationship between environmental factors, gut microbiota, and colorectal cancer, which may lead to development and clinical translation of potential microbiota-based strategies for cancer prevention.
Collapse
Affiliation(s)
- Mingyang Song
- Departments of Epidemiology and Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA
| | - Andrew T. Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
44
|
In a pilot study, reduced fatty acid desaturase 1 function was associated with nonalcoholic fatty liver disease and response to treatment in children. Pediatr Res 2018; 84:696-703. [PMID: 30120404 PMCID: PMC6726123 DOI: 10.1038/s41390-018-0132-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 07/09/2018] [Accepted: 07/18/2018] [Indexed: 01/13/2023]
Abstract
BACKGROUND FADS1 gene encodes delta 5 desaturase, a rate-limiting enzyme in the metabolism of n-3 and n-6 polyunsaturated fatty acids (PUFAs). Minor alleles of FADS1 locus polymorphisms are associated with reduced FADS1 expression and intra-hepatic fat accumulation. However, the relationship between FADS1 expression and pediatric nonalcoholic fatty liver disease (NAFLD) risk remains to be explored. METHODS We analyzed FADS1 transcription levels and their association with intra-hepatic fat and histology in children, and we performed pathway enrichment analysis on transcriptomic profiles associated with FADS1 polymorphisms. We also evaluated the weight of FADS1 alleles on the response to combined docosahexaenoic acid, choline, and vitamin E (DHA-CHO-VE) treatment. RESULTS FADS1 mRNA level was significantly and inversely associated with intra-hepatic fat (p = 0.004), degree of steatosis (p = 0.03), fibrosis (p = 0.05), and NASH (p = 0.008) among pediatric livers. Transcriptomics demonstrated a significant enrichment of a number of pathways strongly related to NAFLD (e.g., liver damage, fibrosis, and hepatic stellate cell activation). Compared to children who are common allele homozygotes, children with FADS1 minor alleles had a greater reduction in steatosis, fibrosis, and NAFLD activity score after DHA-CHO-VE. CONCLUSION This study suggests that decreased FADS1 expression may be associated with NAFLD in children but an increased response to DHA-CHO-VE.
Collapse
|
45
|
Garcia C, Guillocheau E, Richard L, Drouin G, Catheline D, Legrand P, Rioux V. Conversion of dietary trans-vaccenic acid to trans11,cis13-conjugated linoleic acid in the rat lactating mammary gland by Fatty Acid Desaturase 3-catalyzed methyl-end Δ13-desaturation. Biochem Biophys Res Commun 2018; 505:385-391. [PMID: 30262139 DOI: 10.1016/j.bbrc.2018.09.132] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 09/20/2018] [Indexed: 01/04/2023]
Abstract
In vitro, the rat Fatty Acid Desaturase 3 (FADS3) gene was shown to code for an enzyme able to catalyze the unexpected Δ13-desaturation of trans-vaccenic acid, producing the trans11,cis13-conjugated linoleic acid (CLA) isomer. FADS3 may therefore be the first methyl-end trans-vaccenate Δ13-desaturase functionally characterized in mammals, but the proof of this concept is so far lacking in vivo. The present study therefore aimed at investigating further the putative in vivo synthesis of trans11,cis13-CLA from dietary trans-vaccenic acid in rodents. During one week of pregnancy and two weeks post-partum, Sprague-Dawley female rats were fed two diets either high (10.0% of fatty acids and 3.8% of energy intake) or low (0.4% of fatty acids and 0.2% of energy intake) in trans-vaccenic acid. The trans11,cis13-CLA was specifically detected, formally identified and reproducibly quantified (0.06% of total fatty acids) in the mammary gland phospholipids of lactating female rats fed the high trans-vaccenic acid-enriched diet. This result was consistent with FADS3 mRNA expression being significantly higher in the lactating mammary gland than in the liver. Although the apparent metabolic conversion is low, this physiological evidence demonstrates the existence of this new pathway described in the lactating mammary gland and establishes the FADS3 enzyme as a reliable mammalian trans-vaccenate Δ13-desaturase in vivo.
Collapse
Affiliation(s)
- Cyrielle Garcia
- Laboratory of Biochemistry and Human Nutrition, Agrocampus Ouest, Rennes, France
| | - Etienne Guillocheau
- Laboratory of Biochemistry and Human Nutrition, Agrocampus Ouest, Rennes, France; French Dairy Interbranch Organization (CNIEL), Technical and Scientific Department, Paris, France
| | - Léo Richard
- Laboratory of Biochemistry and Human Nutrition, Agrocampus Ouest, Rennes, France
| | - Gaëtan Drouin
- Laboratory of Biochemistry and Human Nutrition, Agrocampus Ouest, Rennes, France
| | - Daniel Catheline
- Laboratory of Biochemistry and Human Nutrition, Agrocampus Ouest, Rennes, France
| | - Philippe Legrand
- Laboratory of Biochemistry and Human Nutrition, Agrocampus Ouest, Rennes, France
| | - Vincent Rioux
- Laboratory of Biochemistry and Human Nutrition, Agrocampus Ouest, Rennes, France.
| |
Collapse
|
46
|
Targeting the gut microbiota to influence brain development and function in early life. Neurosci Biobehav Rev 2018; 95:191-201. [PMID: 30195933 DOI: 10.1016/j.neubiorev.2018.09.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 12/16/2022]
Abstract
In the first 2-3 years of life, the gut microbiota of infants quickly becomes diverse and rich. Disruptions in the evolving gut microbiota during this critical developmental period can impact brain development. Communication between the microbiota, gut and brain is driven by hormonal and neural regulation, as well as immune and metabolic pathways, however, our understanding of how the parallel developments that may underlie this communication are limited. In this paper, we review the known associations between the gut microbiota and brain development and brain function in early life, speculate on the potential mechanisms involved in this complex relationship and describe how nutritional intervention can further modulate the microbiota and, ultimately, brain development and function.
Collapse
|
47
|
Fabersani E, Russo M, Marquez A, Abeijón-Mukdsi C, Medina R, Gauffin-Cano P. Modulation of intestinal microbiota and immunometabolic parameters by caloric restriction and lactic acid bacteria. Food Res Int 2018; 124:188-199. [PMID: 31466639 DOI: 10.1016/j.foodres.2018.06.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 06/01/2018] [Accepted: 06/02/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Emanuel Fabersani
- Universidad Nacional de Tucumán, Tucumán, Argentina; Centro de Referencia para Lactobacilos (CERELA)-CONICET, Tucumán, Argentina
| | - Matías Russo
- Centro de Referencia para Lactobacilos (CERELA)-CONICET, Tucumán, Argentina
| | - Antonela Marquez
- Centro de Referencia para Lactobacilos (CERELA)-CONICET, Tucumán, Argentina
| | | | - Roxana Medina
- Universidad Nacional de Tucumán, Tucumán, Argentina; Centro de Referencia para Lactobacilos (CERELA)-CONICET, Tucumán, Argentina
| | - Paola Gauffin-Cano
- Centro de Referencia para Lactobacilos (CERELA)-CONICET, Tucumán, Argentina.
| |
Collapse
|
48
|
Pachikian BD, Druart C, Catry E, Bindels LB, Neyrinck AM, Larondelle Y, Cani PD, Delzenne NM. Implication of trans-11,trans-13 conjugated linoleic acid in the development of hepatic steatosis. PLoS One 2018; 13:e0192447. [PMID: 29389988 PMCID: PMC5794163 DOI: 10.1371/journal.pone.0192447] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 01/23/2018] [Indexed: 12/15/2022] Open
Abstract
SCOPE Conjugated linoleic acids are linoleic acid isomers found in the diet that can also be produced through bacterial metabolism of polyunsaturated fatty acids. Our objective was to evaluate the contribution of fatty acid metabolites produced from polyunsaturated fatty acids by the gut microbiota in vivo to regulation of hepatic lipid metabolism and steatosis. METHODS AND RESULTS In mice with depleted n-3 polyunsaturated fatty acids, we observed an accumulation of trans-11,trans-13 CLA and cis-9,cis-11 conjugated linoleic acids in the liver tissue that were associated with an increased triglyceride content and expression of lipogenic genes. We used an in vitro model to evaluate the impact of these two conjugated linoleic acids on hepatic lipid metabolism. In HepG2 cells, we observed that only trans-11,trans-13 conjugated linoleic acids recapitulated triglyceride accumulation and increased lipogenic gene expression, which is a phenomenon that may implicate the nuclear factors sterol regulatory element binding protein 1c (SREBP-1c) and carbohydrate-responsive element-binding protein (ChREBP). CONCLUSION The trans-11,trans-13 conjugated linoleic acids can stimulate hepatic lipogenesis, which supports the conclusion that gut microbiota and related metabolites should be considered in the treatment of non-alcoholic liver disease.
Collapse
Affiliation(s)
- Barbara D. Pachikian
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Céline Druart
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Emilie Catry
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Laure B. Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Audrey M. Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Yvan Larondelle
- Institut des Sciences de la Vie, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Patrice D. Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- Université catholique de Louvain, WELBIO, WELBIO (Walloon Excellence in Life sciences and BIOtechnology), Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Brussels, Belgium
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- * E-mail:
| |
Collapse
|
49
|
Requena T, Martínez-Cuesta MC, Peláez C. Diet and microbiota linked in health and disease. Food Funct 2018; 9:688-704. [DOI: 10.1039/c7fo01820g] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Diet has shaped microbiota profiles through human evolution.
Collapse
Affiliation(s)
- T. Requena
- Department of Food Biotechnology and Microbiology
- Institute of Food Science Research
- 28049 Madrid
- Spain
| | - M. C. Martínez-Cuesta
- Department of Food Biotechnology and Microbiology
- Institute of Food Science Research
- 28049 Madrid
- Spain
| | - C. Peláez
- Department of Food Biotechnology and Microbiology
- Institute of Food Science Research
- 28049 Madrid
- Spain
| |
Collapse
|
50
|
Song M, Zhang X, Meyerhardt JA, Giovannucci EL, Ogino S, Fuchs CS, Chan AT. Marine ω-3 polyunsaturated fatty acid intake and survival after colorectal cancer diagnosis. Gut 2017; 66:1790-1796. [PMID: 27436272 PMCID: PMC5247396 DOI: 10.1136/gutjnl-2016-311990] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/31/2016] [Accepted: 06/12/2016] [Indexed: 12/08/2022]
Abstract
OBJECTIVE Experimental evidence supports an antineoplastic activity of marine ω-3 polyunsaturated fatty acids (ω-3 PUFAs; including eicosapentaenoic acid, docosahexaenoic acid and docosapentaenoic acid). However, the influence of ω-3 PUFAs on colorectal cancer (CRC) survival is unknown. DESIGN Within the Nurses' Health Study and Health Professionals Follow-up Study, we prospectively studied CRC-specific and overall mortality in a cohort of 1659 patients with CRC according to intake of marine ω-3 PUFAs and its change after diagnosis. RESULTS Higher intake of marine ω-3 PUFAs after CRC diagnosis was associated with lower risk of CRC-specific mortality (p for trend=0.03). Compared with patients who consumed <0.10 g/day of marine ω-3 PUFAs, those consuming at least 0.30 g/day had an adjusted HR for CRC-specific mortality of 0.59 (95% CI 0.35 to 1.01). Patients who increased their marine ω-3 PUFA intake by at least 0.15 g/day after diagnosis had an HR of 0.30 (95% CI 0.14 to 0.64, p for trend <0.001) for CRC deaths, compared with those who did not change or changed their intake by <0.02 g/day. No association was found between postdiagnostic marine ω-3 PUFA intake and all-cause mortality (p for trend=0.47). CONCLUSIONS High marine ω-3 PUFA intake after CRC diagnosis is associated with lower risk of CRC-specific mortality. Increasing consumption of marine ω-3 PUFAs after diagnosis may confer additional benefits to patients with CRC.
Collapse
Affiliation(s)
- Mingyang Song
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA,Division of Gastroenterology, Massachusetts General Hospital, Boston, MA,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Xuehong Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA
| | - Jeffrey A. Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Edward L. Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Shuji Ogino
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Division of MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA
| | - Charles S. Fuchs
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Andrew T. Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA,Division of Gastroenterology, Massachusetts General Hospital, Boston, MA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| |
Collapse
|