51
|
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
|
52
|
Zhang Q, Zhang L, Chen C, Li P, Lu B. The gut microbiota-artery axis: A bridge between dietary lipids and atherosclerosis? Prog Lipid Res 2023; 89:101209. [PMID: 36473673 DOI: 10.1016/j.plipres.2022.101209] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/09/2022]
Abstract
Atherosclerotic cardiovascular disease is one of the major leading global causes of death. Growing evidence has demonstrated that gut microbiota (GM) and its metabolites play a pivotal role in the onset and progression of atherosclerosis (AS), now known as GM-artery axis. There are interactions between dietary lipids and GM, which ultimately affect GM and its metabolites. Given these two aspects, the GM-artery axis may play a mediating role between dietary lipids and AS. Diets rich in saturated fatty acids (SFAs), omega-6 polyunsaturated fatty acids (n-6 PUFAs), industrial trans fatty acids (TFAs), and cholesterol can increase the levels of atherogenic microbes and metabolites, whereas monounsaturated fatty acids (MUFAs), ruminant TFAs, and phytosterols (PS) can increase the levels of antiatherogenic microbes and metabolites. Actually, dietary phosphatidylcholine (PC), sphingomyelin (SM), and omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been demonstrated to affect AS via the GM-artery axis. Therefore, that GM-artery axis acts as a communication bridge between dietary lipids and AS. Herein, we will describe the molecular mechanism of GM-artery axis in AS and discuss the complex interactions between dietary lipids and GM. In particular, we will highlight the evidence and potential mechanisms of dietary lipids affecting AS via GM-artery axis.
Collapse
Affiliation(s)
- Qinjun Zhang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China; Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
| | - Liangxiao Zhang
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wubhan, China
| | - Cheng Chen
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peiwu Li
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wubhan, China
| | - Baiyi Lu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China; Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
53
|
Jayapala HPS, Lim SY. N-3 Polyunsaturated Fatty Acids and Gut Microbiota. Comb Chem High Throughput Screen 2023; 26:892-905. [PMID: 35786331 DOI: 10.2174/1386207325666220701121025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/09/2022] [Accepted: 04/07/2022] [Indexed: 11/22/2022]
Abstract
For several decades, studies have reported that n-3 polyunsaturated fatty acids (PUFAs) play a beneficial role in cardiovascular, immune, cognitive, visual, mental and metabolic health. The mammalian intestine is colonized by microbiota, including bacteria, archaea, viruses, protozoans, and fungi. The composition of the gut microbiota is influenced by long-term dietary habits, disease-associated dysbiosis, and the use of antibiotics. Accumulating evidence suggests a relationship between n-3 PUFAs and the gut microbiota. N-3 PUFAs can alter the diversity and abundance of the gut microbiome, and gut microbiota can also affect the metabolism and absorption of n-3 PUFAs. Changes in the populations of certain gut microbiota can lead to negative effects on inflammation, obesity, and metabolic diseases. An imbalanced consumption of n-3/n-6 PUFAs may lead to gut microbial dysbiosis, in particular, a significant increase in the ratio of Firmicutes to Bacteroidetes, which eventually results in being overweight and obesity. N-3 PUFA deficiency disrupts the microbiota community in metabolic disorders. In addition, accumulating evidence indicates that the interplay between n-3 PUFAs, gut microbiota, and immune reactions helps to maintain the integrity of the intestinal wall and interacts with host immune cells. Supplementation with n-3 PUFAs may be an effective therapeutic measure to restore gut microbiota homeostasis and correct metabolic disturbances associated with modern chronic diseases. In particular, marine extracts from seaweed contain a considerable dry weight of lipids, including n-3 PUFAs such as eicosapentaenoic acid (EPA, C20: 5) and docosahexaenoic acid (DHA, C22: 6). This review describes how gut microbiota function in intestinal health, how n-3 PUFAs interact with the gut microbiota, and the potential of n-3 PUFAs to influence the gut-brain axis, acting through gut microbiota composition.
Collapse
Affiliation(s)
| | - Sun Young Lim
- Division of Convergence on Marine Science, Korea Maritime & Ocean University, Busan, 49112, Korea
| |
Collapse
|
54
|
Daniel N, Le Barz M, Mitchell PL, Varin TV, Julien IB, Farabos D, Pilon G, Gauthier J, Garofalo C, Kang JX, Trottier J, Barbier O, Roy D, Chassaing B, Levy E, Raymond F, Lamaziere A, Flamand N, Silvestri C, Jobin C, Di Marzo V, Marette A. Comparing Transgenic Production to Supplementation of ω-3 PUFA Reveals Distinct But Overlapping Mechanisms Underlying Protection Against Metabolic and Hepatic Disorders. FUNCTION 2022; 4:zqac069. [PMID: 36778746 PMCID: PMC9909367 DOI: 10.1093/function/zqac069] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022] Open
Abstract
We compared endogenous ω-3 PUFA production to supplementation for improving obesity-related metabolic dysfunction. Fat-1 transgenic mice, who endogenously convert exogenous ω-6 to ω-3 PUFA, and wild-type littermates were fed a high-fat diet and a daily dose of either ω-3 or ω-6 PUFA-rich oil for 12 wk. The endogenous ω-3 PUFA production improved glucose intolerance and insulin resistance but not hepatic steatosis. Conversely, ω-3 PUFA supplementation fully prevented hepatic steatosis but failed to improve insulin resistance. Both models increased hepatic levels of ω-3 PUFA-containing 2-monoacylglycerol and N-acylethanolamine congeners, and reduced levels of ω-6 PUFA-derived endocannabinoids with ω-3 PUFA supplementation being more efficacious. Reduced hepatic lipid accumulation associated with the endocannabinoidome metabolites EPEA and DHEA, which was causally demonstrated by lower lipid accumulation in oleic acid-treated hepatic cells treated with these metabolites. While both models induced a significant fecal enrichment of the beneficial Allobaculum genus, mice supplemented with ω-3 PUFA displayed additional changes in the gut microbiota functions with a significant reduction of fecal levels of the proinflammatory molecules lipopolysaccharide and flagellin. Multiple-factor analysis identify that the metabolic improvements induced by ω-3 PUFAs were accompanied by a reduced production of the proinflammatory cytokine TNFα, and that ω-3 PUFA supplementation had a stronger effect on improving the hepatic fatty acid profile than endogenous ω-3 PUFA. While endogenous ω-3 PUFA production preferably improves glucose tolerance and insulin resistance, ω-3 PUFA intake appears to be required to elicit selective changes in hepatic endocannabinoidome signaling that are essential to alleviate high-fat diet-induced hepatic steatosis.
Collapse
Affiliation(s)
| | | | - Patricia L Mitchell
- Quebec Heart and Lung Institute Research Centre, Laval University, Quebec, QC G1V 4G5, Canada,Institute of Nutrition and Functional Foods (INAF), Centre NUTRISS, Quebec, QC G1V 0A6, Canada
| | - Thibault V Varin
- Quebec Heart and Lung Institute Research Centre, Laval University, Quebec, QC G1V 4G5, Canada,Institute of Nutrition and Functional Foods (INAF), Centre NUTRISS, Quebec, QC G1V 0A6, Canada
| | - Isabelle Bourdeau Julien
- Institute of Nutrition and Functional Foods (INAF), Centre NUTRISS, Quebec, QC G1V 0A6, Canada,Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Laval University, Quebec, QC G1V 0A6, Canada
| | - Dominique Farabos
- Saint Antoine Research Center, Sorbonne University INSERM UMR 938; Assistance Publique - Hôpitaux de Paris, Clinical Metabolomics department, Hôpital Saint Antoine, Paris, 75571, France
| | - Geneviève Pilon
- Quebec Heart and Lung Institute Research Centre, Laval University, Quebec, QC G1V 4G5, Canada,Institute of Nutrition and Functional Foods (INAF), Centre NUTRISS, Quebec, QC G1V 0A6, Canada
| | - Josée Gauthier
- Department of Medicine, Department of Infectious Diseases and Immunology, and Department of Anatomy and Cell Physiology, University of Florida, Gainesville FL, 32608, USA
| | - Carole Garofalo
- Department of Nutrition, University of Montreal, Montreal QC H3T 1A8, Canada and Research Centre, Sainte-Justine Hospital, Montreal, QC H3T 1C5, Canada
| | - Jing X Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown MA 02129, USA
| | - Jocelyn Trottier
- Laboratory of Molecular Pharmacology, CHU-Quebec Research Centre, and Faculty of Pharmacy, Laval University, Quebec, QC G1V 0A6, Canada
| | - Olivier Barbier
- Laboratory of Molecular Pharmacology, CHU-Quebec Research Centre, and Faculty of Pharmacy, Laval University, Quebec, QC G1V 0A6, Canada
| | - Denis Roy
- Faculty of Agricultural and Food Sciences, School of Nutrition, Laval University, Quebec, QC G1V 0A6, Canada,Institute of Nutrition and Functional Foods (INAF), Centre NUTRISS, Quebec, QC G1V 0A6, Canada
| | - Benoit Chassaing
- INSERM U1016, Mucosal Microbiota in Chronic Inflammatory Diseases’ Team, CNRS UMR 8104, University of Paris, Paris, 75014, France
| | - Emile Levy
- Department of Nutrition, University of Montreal, Montreal QC H3T 1A8, Canada and Research Centre, Sainte-Justine Hospital, Montreal, QC H3T 1C5, Canada
| | - Frédéric Raymond
- Institute of Nutrition and Functional Foods (INAF), Centre NUTRISS, Quebec, QC G1V 0A6, Canada,Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Laval University, Quebec, QC G1V 0A6, Canada
| | - Antonin Lamaziere
- Saint Antoine Research Center, Sorbonne University INSERM UMR 938; Assistance Publique - Hôpitaux de Paris, Clinical Metabolomics department, Hôpital Saint Antoine, Paris, 75571, France
| | - Nicolas Flamand
- Quebec Heart and Lung Institute Research Centre, Laval University, Quebec, QC G1V 4G5, Canada,Faculty of Medicine, Department of Medicine, Laval University, QC G1V 0A6, Canada,Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Laval University, Quebec, QC G1V 0A6, Canada
| | - Cristoforo Silvestri
- Quebec Heart and Lung Institute Research Centre, Laval University, Quebec, QC G1V 4G5, Canada,Institute of Nutrition and Functional Foods (INAF), Centre NUTRISS, Quebec, QC G1V 0A6, Canada,Faculty of Medicine, Department of Medicine, Laval University, QC G1V 0A6, Canada,Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Laval University, Quebec, QC G1V 0A6, Canada
| | - Christian Jobin
- Department of Medicine, Department of Infectious Diseases and Immunology, and Department of Anatomy and Cell Physiology, University of Florida, Gainesville FL, 32608, USA
| | - Vincenzo Di Marzo
- Quebec Heart and Lung Institute Research Centre, Laval University, Quebec, QC G1V 4G5, Canada,Institute of Nutrition and Functional Foods (INAF), Centre NUTRISS, Quebec, QC G1V 0A6, Canada,Faculty of Medicine, Department of Medicine, Laval University, QC G1V 0A6, Canada,Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Laval University, Quebec, QC G1V 0A6, Canada,Joint International Research Unit on Chemical and Biomolecular Research on the Microbiome and its Impact on Metabolic Health and Nutrition between Laval University and Consiglio Nazionale delle Ricerche, Institute of Biomolecular Chemistry, Campania, 80078, Italy
| | | |
Collapse
|
55
|
Ali Q, Ma S, Farooq U, Niu J, Li F, Li D, Wang Z, Sun H, Cui Y, Shi Y. Pasture intake protects against commercial diet-induced lipopolysaccharide production facilitated by gut microbiota through activating intestinal alkaline phosphatase enzyme in meat geese. Front Immunol 2022; 13:1041070. [PMID: 36569878 PMCID: PMC9774522 DOI: 10.3389/fimmu.2022.1041070] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction Diet strongly affects gut microbiota composition, and gut bacteria can influence the intestinal barrier functions and systemic inflammation through metabolic endotoxemia. In-house feeding system (IHF, a low dietary fiber source) may cause altered cecal microbiota composition and inflammatory responses in meat geese via increased endotoxemia (lipopolysaccharides) with reduced intestinal alkaline phosphatase (ALP) production. The effects of artificial pasture grazing system (AGF, a high dietary fiber source) on modulating gut microbiota architecture and gut barrier functions have not been investigated in meat geese. Therefore, this study aimed to investigate whether intestinal ALP could play a critical role in attenuating reactive oxygen species (ROS) generation and ROS facilitating NF-κB pathway-induced systemic inflammation in meat geese. Methods The impacts of IHF and AGF systems on gut microbial composition via 16 sRNA sequencing were assessed in meat geese. The host markers analysis through protein expression of serum and cecal tissues, hematoxylin and eosin (H&E) staining, localization of NF-қB and Nrf2 by immunofluorescence analysis, western blotting analysis of ALP, and quantitative PCR of cecal tissues was evaluated. Results and Discussion In the gut microbiota analysis, meat geese supplemented with pasture showed a significant increase in commensal microbial richness and diversity compared to IHF meat geese demonstrating the antimicrobial, antioxidant, and anti-inflammatory ability of the AGF system. A significant increase in intestinal ALP-induced Nrf2 signaling pathway was confirmed representing LPS dephosphorylation mediated TLR4/MyD88 induced ROS reduction mechanisms in AGF meat geese. Further, the correlation analysis of top 44 host markers with gut microbiota showed that artificial pasture intake protected gut barrier functions via reducing ROS-mediated NF-κB pathway-induced gut permeability, systemic inflammation, and aging phenotypes. In conclusion, the intestinal ALP functions to regulate gut microbial homeostasis and barrier function appear to inhibit pro-inflammatory cytokines by reducing LPS-induced ROS production in AGF meat geese. The AGF system may represent a novel therapy to counteract the chronic inflammatory state leading to low dietary fiber-related diseases in animals.
Collapse
Affiliation(s)
- Qasim Ali
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China
| | - Sen Ma
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Henan Agricultural University, Zhengzhou, Henan, China,Henan Herbage Engineering Technology Research Center, Henan Agricultural University, Zhengzhou, Henan, China
| | - Umar Farooq
- Department of Poultry Science, University of Agriculture Faisalabad, Toba Tek Singh, Pakistan
| | - Jiakuan Niu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China
| | - Fen Li
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China
| | - Defeng Li
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Henan Agricultural University, Zhengzhou, Henan, China,Henan Herbage Engineering Technology Research Center, Henan Agricultural University, Zhengzhou, Henan, China
| | - Zhichang Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Henan Agricultural University, Zhengzhou, Henan, China,Henan Herbage Engineering Technology Research Center, Henan Agricultural University, Zhengzhou, Henan, China
| | - Hao Sun
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Henan Agricultural University, Zhengzhou, Henan, China,Henan Herbage Engineering Technology Research Center, Henan Agricultural University, Zhengzhou, Henan, China
| | - Yalei Cui
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Henan Agricultural University, Zhengzhou, Henan, China,Henan Herbage Engineering Technology Research Center, Henan Agricultural University, Zhengzhou, Henan, China
| | - Yinghua Shi
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan, China,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Henan Agricultural University, Zhengzhou, Henan, China,Henan Herbage Engineering Technology Research Center, Henan Agricultural University, Zhengzhou, Henan, China,*Correspondence: Yinghua Shi,
| |
Collapse
|
56
|
Marathe SJ, Snider MA, Flores-Torres AS, Dubin PJ, Samarasinghe AE. Human matters in asthma: Considering the microbiome in pulmonary health. Front Pharmacol 2022; 13:1020133. [PMID: 36532717 PMCID: PMC9755222 DOI: 10.3389/fphar.2022.1020133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/15/2022] [Indexed: 07/25/2023] Open
Abstract
Microbial communities form an important symbiotic ecosystem within humans and have direct effects on health and well-being. Numerous exogenous factors including airborne triggers, diet, and drugs impact these established, but fragile communities across the human lifespan. Crosstalk between the mucosal microbiota and the immune system as well as the gut-lung axis have direct correlations to immune bias that may promote chronic diseases like asthma. Asthma initiation and pathogenesis are multifaceted and complex with input from genetic, epigenetic, and environmental components. In this review, we summarize and discuss the role of the airway microbiome in asthma, and how the environment, diet and therapeutics impact this low biomass community of microorganisms. We also focus this review on the pediatric and Black populations as high-risk groups requiring special attention, emphasizing that the whole patient must be considered during treatment. Although new culture-independent techniques have been developed and are more accessible to researchers, the exact contribution the airway microbiome makes in asthma pathogenesis is not well understood. Understanding how the airway microbiome, as a living entity in the respiratory tract, participates in lung immunity during the development and progression of asthma may lead to critical new treatments for asthma, including population-targeted interventions, or even more effective administration of currently available therapeutics.
Collapse
Affiliation(s)
- Sandesh J. Marathe
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Division of Pulmonology, Allergy-Immunology, and Sleep, Memphis, TN, United States
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN, United States
| | - Mark A. Snider
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Division of Emergency Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Armando S. Flores-Torres
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN, United States
| | - Patricia J. Dubin
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Division of Pulmonology, Allergy-Immunology, and Sleep, Memphis, TN, United States
| | - Amali E. Samarasinghe
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Division of Pulmonology, Allergy-Immunology, and Sleep, Memphis, TN, United States
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN, United States
| |
Collapse
|
57
|
Xu H, Jurado-Fasoli L, Ortiz-Alvarez L, Osuna-Prieto FJ, Kohler I, Di X, Vilchez-Vargas R, Link A, Plaza-Díaz J, Gil A, Rensen PCN, Ruiz JR, Martinez-Tellez B. Plasma Levels of Omega-3 and Omega-6 Derived Oxylipins Are Associated with Fecal Microbiota Composition in Young Adults. Nutrients 2022; 14:nu14234991. [PMID: 36501021 PMCID: PMC9736377 DOI: 10.3390/nu14234991] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Pre-clinical studies suggest that circulating oxylipins, i.e., the oxidation products of polyunsaturated fatty acids (PUFAs), modulate gut microbiota composition in mice, but there is no information available in humans. Therefore, this study aimed to investigate the relationship between omega-3 and omega-6 derived oxylipins plasma levels and fecal microbiota composition in a cohort of young adults. 80 young adults (74% women; 21.9 ± 2.2 years old) were included in this cross-sectional study. Plasma levels of oxylipins were measured using liquid chromatography-tandem mass spectrometry. Fecal microbiota composition was analyzed by V3-V4 16S rRNA gene sequencing. We observed that plasma levels of omega-3 derived oxylipins were positively associated with the relative abundance of Clostridium cluster IV genus (Firmicutes phylum; rho ≥ 0.415, p ≤ 0.009) and negatively associated with the relative abundance of Sutterella genus (Proteobacteria phylum; rho ≥ -0.270, p ≤ 0.041), respectively. Moreover, plasma levels of omega-6 derived oxylipins were negatively associated with the relative abundance of Acidaminococcus and Phascolarctobacterium genera (Firmicutes phylum; all rho ≥ -0.263, p ≤ 0.024), as well as Sutterella, Succinivibrio, and Gemmiger genera (Proteobacteria phylum; all rho ≥ -0.263, p ≤ 0.024). Lastly, the ratio between omega-6 and omega-3 oxylipins plasma levels was negatively associated with the relative abundance of Clostridium cluster IV genus (Firmicutes phylum; rho = -0.334, p = 0.004) and Butyricimonas genus (Bacteroidetes phylum; rho = -0.292, p = 0.014). In conclusion, our results show that the plasma levels of omega-3 and omega-6 derived oxylipins are associated with the relative abundance of specific fecal bacteria genera.
Collapse
Affiliation(s)
- Huiwen Xu
- PROFITH (PROmoting FITness and Health through Physical Activity) Research Group, Sport and Health University Research Institute (iMUDS), Department of Physical and Sports Education, Faculty of Sport Sciences, University of Granada, 18071 Granada, Spain
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
| | - Lucas Jurado-Fasoli
- PROFITH (PROmoting FITness and Health through Physical Activity) Research Group, Sport and Health University Research Institute (iMUDS), Department of Physical and Sports Education, Faculty of Sport Sciences, University of Granada, 18071 Granada, Spain
- Department of Physiology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| | - Lourdes Ortiz-Alvarez
- PROFITH (PROmoting FITness and Health through Physical Activity) Research Group, Sport and Health University Research Institute (iMUDS), Department of Physical and Sports Education, Faculty of Sport Sciences, University of Granada, 18071 Granada, Spain
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
| | - Francisco J. Osuna-Prieto
- PROFITH (PROmoting FITness and Health through Physical Activity) Research Group, Sport and Health University Research Institute (iMUDS), Department of Physical and Sports Education, Faculty of Sport Sciences, University of Granada, 18071 Granada, Spain
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, 18071 Granada, Spain
| | - Isabelle Kohler
- Division of BioAnalytical Chemistry, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands
- Center for Analytical Sciences Amsterdam, 1081 Amsterdam, The Netherlands
| | - Xinyu Di
- Department of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, 2300 Leiden, The Netherlands
| | - Ramiro Vilchez-Vargas
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke-University Magdeburg, 39106 Magdeburg, Germany
| | - Alexander Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke-University Magdeburg, 39106 Magdeburg, Germany
| | - Julio Plaza-Díaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
- Instituto de Investigación Biosanitaria, ibs.Granada, 18014 Granada, Spain
| | - Angel Gil
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria, ibs.Granada, 18014 Granada, Spain
- CIBEROBN, Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition, Carlos III Health Institute, 28029 Madrid, Spain
- Institute of Nutrition and Food Technology, Center of Biomedical Research, University of Granada, 18071 Granada, Spain
| | - Patrick C. N. Rensen
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 Leiden, The Netherlands
| | - Jonatan R. Ruiz
- PROFITH (PROmoting FITness and Health through Physical Activity) Research Group, Sport and Health University Research Institute (iMUDS), Department of Physical and Sports Education, Faculty of Sport Sciences, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria, ibs.Granada, 18014 Granada, Spain
- Correspondence: (J.R.R.); (B.M.-T.)
| | - Borja Martinez-Tellez
- PROFITH (PROmoting FITness and Health through Physical Activity) Research Group, Sport and Health University Research Institute (iMUDS), Department of Physical and Sports Education, Faculty of Sport Sciences, University of Granada, 18071 Granada, Spain
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2300 Leiden, The Netherlands
- Department of Education, Faculty of Education Sciences and SPORT Research Group (CTS-1024), CERNEP Research Center, University of Almería, 04120 Almería, Spain
- Correspondence: (J.R.R.); (B.M.-T.)
| |
Collapse
|
58
|
Vergne-Salle P, Salle L, Fressinaud-Marie AC, Descamps-Deplas A, Montestruc F, Bonnet C, Bertin P. Diet and Disease Activity in Patients with Axial Spondyloarthritis: SpondyloArthritis and NUTrition Study (SANUT). Nutrients 2022; 14:nu14224730. [PMID: 36432416 PMCID: PMC9695957 DOI: 10.3390/nu14224730] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
Axial Spondyloarthritis (axSpA) patients with inflamed intestines have higher SpA activity. Diets that modulate microbiota may influence inflammation and SpA activity. Today, data concerning the impact of diet on SpA activity are scarce. SANUT was a single-center, noninterventional, cohort study that assessed dietetic profiles associated with SpA activity in axSpA. Demographic, clinical, SpA-related, quality of life (QoL), fatigue, physical activity, and dietary data were collected. SpA activity was assessed by Ankylosing Spondylitis Disease Activity Score (ASDAS) and by Bath Ankylosing Spondylitis Disease Activity Index (BASDAI). We assessed whether high SpA activity was associated with nutriment consumption. Between 12 February 2018 and 12 February 2020, 278 patients participated. High SpA activity, as measured by ASDAS and BASDAI, was significantly associated with higher body mass index and waist circumference, negative HLA-B27, lower QoL, higher fatigue, and higher digestive-symptom scores. Furthermore, high SpA activity, as measured by BASDAI, was associated with female sex, smoking status, patients who were not actively employed, reduced physical activity, and high intake of ultra-transformed foods, while high SpA activity, as measured by ASDAS, was associated with low intake of omega-3 PUFAs and fiber. Therefore, low intakes of omega-3 PUFAs and fiber, and high intake of ultra-transformed foods, are associated with high SpA activity.
Collapse
Affiliation(s)
- Pascale Vergne-Salle
- Rheumatology Department, University Hospital of Limoges and Laboratory PEIRENE UR 22722 Institut OmegaHealth, 87042 Limoges, France
- Correspondence:
| | - Laurence Salle
- Endocrinology Department, University Hospital of Limoges and Inserm U1094, EpiMaCT—Epidemiology of Chronic Diseases in Tropical Zone, 87042 Limoges, France
| | - Anne Catherine Fressinaud-Marie
- Rheumatology Department, University Hospital of Limoges and Laboratory PEIRENE UR 22722 Institut OmegaHealth, 87042 Limoges, France
| | - Adeline Descamps-Deplas
- Rheumatology Department, University Hospital of Limoges and Laboratory PEIRENE UR 22722 Institut OmegaHealth, 87042 Limoges, France
| | | | - Christine Bonnet
- Rheumatology Department, University Hospital of Limoges and Laboratory PEIRENE UR 22722 Institut OmegaHealth, 87042 Limoges, France
| | - Philippe Bertin
- Rheumatology Department, University Hospital of Limoges and Laboratory PEIRENE UR 22722 Institut OmegaHealth, 87042 Limoges, France
| |
Collapse
|
59
|
Lee D, Lee VMY, Hur SK. Manipulation of the diet-microbiota-brain axis in Alzheimer's disease. Front Neurosci 2022; 16:1042865. [PMID: 36408394 PMCID: PMC9672822 DOI: 10.3389/fnins.2022.1042865] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Several studies investigating the pathogenesis of Alzheimer's disease have identified various interdependent constituents contributing to the exacerbation of the disease, including Aβ plaque formation, tau protein hyperphosphorylation, neurofibrillary tangle accumulation, glial inflammation, and the eventual loss of proper neural plasticity. Recently, using various models and human patients, another key factor has been established as an influential determinant in brain homeostasis: the gut-brain axis. The implications of a rapidly aging population and the absence of a definitive cure for Alzheimer's disease have prompted a search for non-pharmaceutical tools, of which gut-modulatory therapies targeting the gut-brain axis have shown promise. Yet multiple recent studies examining changes in human gut flora in response to various probiotics and environmental factors are limited and difficult to generalize; whether the state of the gut microbiota in Alzheimer's disease is a cause of the disease, a result of the disease, or both through numerous feedback loops in the gut-brain axis, remains unclear. However, preliminary findings of longitudinal studies conducted over the past decades have highlighted dietary interventions, especially Mediterranean diets, as preventative measures for Alzheimer's disease by reversing neuroinflammation, modifying the intestinal and blood-brain barrier (BBB), and addressing gut dysbiosis. Conversely, the consumption of Western diets intensifies the progression of Alzheimer's disease through genetic alterations, impaired barrier function, and chronic inflammation. This review aims to support the growing body of experimental and clinical data highlighting specific probiotic strains and particular dietary components in preventing Alzheimer's disease via the gut-brain axis.
Collapse
Affiliation(s)
- Daniel Lee
- Middleton High School, Middleton, WI, United States
| | - Virginia M-Y. Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Seong Kwon Hur
- Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA, United States
| |
Collapse
|
60
|
Dang DX, Kim IH. Coated refined fish oil supplementation improves growth performance and meat quality in finishing pigs. Livest Sci 2022. [DOI: 10.1016/j.livsci.2022.105099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
61
|
Hao L, Nie YH, Chen CY, Li XY, Kaliannan K, Kang JX. Omega-3 Polyunsaturated Fatty Acids Protect against High-Fat Diet-Induced Morphological and Functional Impairments of Brown Fat in Transgenic Fat-1 Mice. Int J Mol Sci 2022; 23:ijms231911903. [PMID: 36233205 PMCID: PMC9570395 DOI: 10.3390/ijms231911903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/28/2022] [Accepted: 10/01/2022] [Indexed: 11/17/2022] Open
Abstract
The role of omega-3 polyunsaturated fatty acids (n-3 PUFAs) in the regulation of energy homeostasis remains poorly understood. In this study, we used a transgenic fat-1 mouse model, which can produce n-3 PUFAs endogenously, to investigate how n-3 PUFAs regulate the morphology and function of brown adipose tissue (BAT). We found that high-fat diet (HFD) induced a remarkable morphological change in BAT, characterized by “whitening” due to large lipid droplet accumulation within BAT cells, associated with obesity in wild-type (WT) mice, whereas the changes in body fat mass and BAT morphology were significantly alleviated in fat-1 mice. The expression of thermogenic markers and lypolytic enzymes was significantly higher in fat-1 mice than that in WT mice fed with HFD. In addition, fat-1 mice had significantly lower levels of inflammatory markers in BAT and lipopolysaccharide (LPS) in plasma compared with WT mice. Furthermore, fat-1 mice were resistant to LPS-induced suppression of UCP1 and PGC-1 expression and lipid deposits in BAT. Our data has demonstrated that high-fat diet-induced obesity is associated with impairments of BAT morphology (whitening) and function, which can be ameliorated by elevated tissue status of n-3 PUFAs, possibly through suppressing the effects of LPS on inflammation and thermogenesis.
Collapse
Affiliation(s)
- Lei Hao
- Laboratory for Lipid Medicine and Technology (LLMT), Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
- Department of Nursing and Allied Health Professions, Indiana University of Pennsylvania, Indiana, PA 15705, USA
| | - Yong-Hui Nie
- Laboratory for Lipid Medicine and Technology (LLMT), Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Chih-Yu Chen
- Laboratory for Lipid Medicine and Technology (LLMT), Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Xiang-Yong Li
- Laboratory for Lipid Medicine and Technology (LLMT), Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Kanakaraju Kaliannan
- Laboratory for Lipid Medicine and Technology (LLMT), Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Jing X. Kang
- Laboratory for Lipid Medicine and Technology (LLMT), Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
- Correspondence: ; Tel.: +1-(617)-726-8509; Fax: +1-(617)-726-6144
| |
Collapse
|
62
|
Vivekanandan L, Singaravel S, Thangavel S. Favorable Aspects of Silymarin in Linezolid Treatment Against Diabetic
Methicillin-Resistant Staphylococcus aureus (MRSA) Infected Rats. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180819666220411080831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Linezolid used for diabetic methicillin-resistant Staphylococcus aureus (MRSA)
infections is limited due to hepatotoxicity, lactic acidosis, anemia, and oxidative stress induced by diabetes
and linezolid therapy. Silymarin is a hepatoprotective, antioxidant, antibacterial, and antidiabetic.
Objective:
The research investigated the role of silymarin in linezolid treatment against MRSA-infected
diabetic rats.
Methods:
Type 2 diabetes mellitus (T2DM) was induced by a high-fat diet (58% calories fat) for 2 weeks,
followed by a single intraperitoneal injection of streptozotocin (STZ) 35 mg/kg into Wistar rats. The diabetic
rats were rendered neutropenic and subcutaneously injected with 106 CFU/ml of MRSA. Linezolid
and silymarin were administered orally at a dose of 50 mg/kg twice daily for 14 days. The bacterial
load/abscess, hematological, biochemical, enzymatic parameters, antioxidants, and histopathological studies
were performed on the 42nd day.
Results:
The MRSA was confirmed by PCR assay. The minimum inhibitory concentration of linezolid
was found to be 0.5-2 μg/ml. The linezolid treated MRSA infected diabetic rats showed 9.69 x 103 CFU /
abscess bacterial count, decreased intestinal alkaline phosphatase (IAP), RBC, antioxidants, elevated lactate,
and liver markers than diabetic rats. The silymarin treatment showed a decrease in the bacterial count
(2.98 x 103 CFU / abscess), serum lactate, liver markers, increased IAP levels, and antioxidants in linezolid
treated diabetic infected rats.
Conclusion:
The research concluded that silymarin could be a better herbal therapeutic agent that attenuated
diabetic and linezolid induced complications in MRSA-infected diabetic rats.
Collapse
Affiliation(s)
- Lalitha Vivekanandan
- Department of Pharmacology, Nandha College of Pharmacy and Research Institute, Erode 638052, Tamilnadu, India
| | - Sengottuvelu Singaravel
- Department of Pharmacology, Nandha College of Pharmacy and Research Institute, Erode 638052, Tamilnadu, India
| | - Sivakumar Thangavel
- Department of Chemistry, Nandha College of Pharmacy and Research Institute, Erode 638052, Tamilnadu, India
| |
Collapse
|
63
|
Roussel C, Anunciação Braga Guebara S, Plante PL, Desjardins Y, Di Marzo V, Silvestri C. Short-term supplementation with ω-3 polyunsaturated fatty acids modulates primarily mucolytic species from the gut luminal mucin niche in a human fermentation system. Gut Microbes 2022; 14:2120344. [PMID: 36109831 PMCID: PMC9481098 DOI: 10.1080/19490976.2022.2120344] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Consumption of omega-3 polyunsaturated fatty acids (ω-3 PUFAs) eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) provides multifaceted health benefits. Recent studies suggest that ω-3 PUFAs modulate the gut microbiota by enhancing health-promoting bacteria, such as the mucin specialist Akkermansia muciniphila. However, these prebiotic properties have been poorly investigated and direct effects on the gut microbiome have never been explored dynamically across gut regions and niches (lumen vs. mucus-associated microbiota). Thus, we studied the effects of 1 week EPA- and DHA-enriched ω-3 fish-oil supplementation on the composition and functionality of the human microbiome in a Mucosal Simulator of the Human Intestinal Microbial Ecosystem (M-SHIME®). Gut microbial communities derived from one individual harvested in two different seasons were tested in duplicate. Luminal and outer mucus-associated microbiota of the ileum, ascending, transverse and descending colons were cultivated over 28 d from fecal inoculates and supplemented with ω-3 PUFAs for the last 7 d. We show that ω-3 PUFA supplementation modulates the microbiota in a gut region- and niche-dependent fashion. The outer mucus-associated microbiota displayed a higher resilience than the luminal mucin habitat to ω-3 PUFAs, with a remarkable blooming of Akkermansia muciniphila in opposition to a decrease of Firmicutes-mucolytic bacteria. The ω-3 PUFAs also induced a gradual and significant depletion of non-mucolytic Clostridia members in luminal habitats. Finally, increased concentrations of the short chain fatty acids (SCFA) propionate in colon regions at the end of the supplementation was associated positively with the bloom of Akkermansia muciniphila and members of the Desulfovibrionia class.
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, CRIUCPQ Laval University, Quebec, QC, Canada
| | - Sara Anunciação Braga Guebara
- 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, CRIUCPQ Laval University, Quebec, QC, Canada
| | - Pier-Luc Plante
- 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, CRIUCPQ Laval University, Quebec, QC, Canada
| | - Yves Desjardins
- 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
| | - 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, CRIUCPQ Laval University, Quebec, QC, Canada,Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada,CONTACT Vincenzo Di Marzo Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, 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, CRIUCPQ Laval University, Quebec, QC, Canada,Faculty of Medicine, Department of Medicine, Laval University, Quebec, QC, Canada,Cristoforo Silvestri Faculty of Medicine, Department of Medicine Laval University, Quebec, QC, Canada
| |
Collapse
|
64
|
Cirulli F, De Simone R, Musillo C, Ajmone-Cat MA, Berry A. Inflammatory Signatures of Maternal Obesity as Risk Factors for Neurodevelopmental Disorders: Role of Maternal Microbiota and Nutritional Intervention Strategies. Nutrients 2022; 14:nu14153150. [PMID: 35956326 PMCID: PMC9370669 DOI: 10.3390/nu14153150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/18/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Obesity is a main risk factor for the onset and the precipitation of many non-communicable diseases. This condition, which is associated with low-grade chronic systemic inflammation, is of main concern during pregnancy leading to very serious consequences for the new generations. In addition to the prominent role played by the adipose tissue, dysbiosis of the maternal gut may also sustain the obesity-related inflammatory milieu contributing to create an overall suboptimal intrauterine environment. Such a condition here generically defined as “inflamed womb” may hold long-term detrimental effects on fetal brain development, increasing the vulnerability to mental disorders. In this review, we will examine the hypothesis that maternal obesity-related gut dysbiosis and the associated inflammation might specifically target fetal brain microglia, the resident brain immune macrophages, altering neurodevelopmental trajectories in a sex-dependent fashion. We will also review some of the most promising nutritional strategies capable to prevent or counteract the effects of maternal obesity through the modulation of inflammation and oxidative stress or by targeting the maternal microbiota.
Collapse
Affiliation(s)
- Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; or
- Correspondence: (F.C.); (A.B.)
| | - Roberta De Simone
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (R.D.S.); (M.A.A.-C.)
| | - Chiara Musillo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; or
- PhD Program in Behavioral Neuroscience, Department of Psychology, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Antonietta Ajmone-Cat
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (R.D.S.); (M.A.A.-C.)
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; or
- Correspondence: (F.C.); (A.B.)
| |
Collapse
|
65
|
Bilal M, Ashraf S, Zhao X. Dietary Component-Induced Inflammation and Its Amelioration by Prebiotics, Probiotics, and Synbiotics. Front Nutr 2022; 9:931458. [PMID: 35938108 PMCID: PMC9354043 DOI: 10.3389/fnut.2022.931458] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022] Open
Abstract
A balanced diet with many dietary components maintains immune homeostasis directly by interacting with innate and adaptive immune components or indirectly through gut microbiota and their metabolites. Dietary components may inhibit pro-inflammatory mediators and promote anti-inflammatory functions or vice versa. Western diets with imbalanced dietary components skew the immune balance toward pro-inflammation and induce intestinal inflammation, consequently leading to many intestinal and systemic inflammatory diseases like ulcerative colitis, Crohn's disease, irritable bowel syndrome, cardiovascular problems, obesity, and diabetes. The dietary component-induced inflammation is usually chronic in nature and frequently caused or accompanied by alterations in gut microbiota. Therefore, microbiome-targeted therapies such as probiotics, prebiotics and synbiotics hold great potentials to amend immune dysregulation and gut dysbiosis, preventing and treating intestinal and systemic inflammatory diseases. Probiotics, prebiotics and synbioitcs are progressively being added to foods and beverages, with claims of health benefits. However, the underlining mechanisms of these interventions for preventing and treating dietary component-induced inflammation are still not very clear. In addition, possibly ineffective or negative consequences of some probiotics, prebiotics and synbiotics call for stringent testing and regulation. Here, we will first briefly review inflammation, in terms of its types and the relationship between different dietary components and immune responses. Then, we focus on current knowledge about the direct and indirect effects of probiotics, prebiotics and synbiotics on intestinal and systemic inflammation. Understanding how probiotics, prebiotics and synbiotics modulate the immune system and gut microbiota will improve our strategies for preventing and treating dietary component-induced intestinal inflammation and inflammatory diseases.
Collapse
|
66
|
Dwaib HS, AlZaim I, Ajouz G, Eid AH, El-Yazbi A. Sex Differences in Cardiovascular Impact of Early Metabolic Impairment: Interplay between Dysbiosis and Adipose Inflammation. Mol Pharmacol 2022; 102:481-500. [PMID: 34732528 DOI: 10.1124/molpharm.121.000338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/23/2021] [Indexed: 11/22/2022] Open
Abstract
The evolving view of gut microbiota has shifted toward describing the colonic flora as a dynamic organ in continuous interaction with systemic physiologic processes. Alterations of the normal gut bacterial profile, known as dysbiosis, has been linked to a wide array of pathologies. Of particular interest is the cardiovascular-metabolic disease continuum originating from positive energy intake and high-fat diets. Accumulating evidence suggests a role for sex hormones in modulating the gut microbiome community. Such a role provides an additional layer of modulation of the early inflammatory changes culminating in negative metabolic and cardiovascular outcomes. In this review, we will shed the light on the role of sex hormones in cardiovascular dysfunction mediated by high-fat diet-induced dysbiosis, together with the possible involvement of insulin resistance and adipose tissue inflammation. Insights into novel therapeutic interventions will be discussed as well. SIGNIFICANCE STATEMENT: Increasing evidence implicates a role for dysbiosis in the cardiovascular complications of metabolic dysfunction. This minireview summarizes the available data on the sex-based differences in gut microbiota alterations associated with dietary patterns leading to metabolic impairment. A role for a differential impact of adipose tissue inflammation across sexes in mediating the cardiovascular detrimental phenotype following diet-induced dysbiosis is proposed. Better understanding of this pathway will help introduce early approaches to mitigate cardiovascular deterioration in metabolic disease.
Collapse
Affiliation(s)
- Haneen S Dwaib
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| | - Ghina Ajouz
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| | - Ahmed El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine (H.S.D., I.A., G.A., A.E.-Y.), Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences (H.S.D.), American University of Beirut, Beirut, Lebanon; Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon (I.A.); Department of Basic Medical Sciences, College of Medicine (A.H.E.), Biomedical and Pharmaceutical Research Unit, QU Health (A.H.E.), Qatar University, Doha, Qatar; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (A.E.-Y.); and Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt (A.E.-Y.)
| |
Collapse
|
67
|
Carneiro PV, Montenegro NDA, Lana A, Amato AA, Santos GM. Lipids from gut microbiota: pursuing a personalized treatment. Trends Mol Med 2022; 28:631-643. [PMID: 35739018 DOI: 10.1016/j.molmed.2022.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 10/18/2022]
Abstract
The discovery of microbiome metabolites has enlivened the field of fecal transplantation for therapeutic purposes. However, the transfer of pathogenic living organisms was recently observed to limit its therapeutic potential by increasing the risk of infection. Lipids produced by gut microbiota enter the circulation and control many phenotypic changes associated with microbiota composition. Fecal lipids significantly impact the regulation of several cell signaling pathways, including inflammation. Focusing on these molecules, we review how bioactive gut microbiota-associated lipids affect cellular functioning and clinical outcome. Here, we interrogate whether the gut microbiota can be considered a cutting-edge biotechnological tool for rapid metabolic engineering of meaningful lipids to offer a novel personalized therapy.
Collapse
Affiliation(s)
- Pamela V Carneiro
- Laboratório de Farmacologia Molecular, Universidade de Brasília, Brasília, Brasil
| | | | - Addison Lana
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, USA
| | - Angelica A Amato
- Laboratório de Farmacologia Molecular, Universidade de Brasília, Brasília, Brasil
| | - Guilherme M Santos
- Laboratório de Farmacologia Molecular, Universidade de Brasília, Brasília, Brasil.
| |
Collapse
|
68
|
Zhang H, Liu X, Zheng Y, Zha X, Elsabagh M, Zhang Y, Ma Y, Loor JJ, Wang M, Wang H. Effects of the maternal gut microbiome and gut-placental axis on melatonin efficacy in alleviating cadmium-induced fetal growth restriction. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 237:113550. [PMID: 35487173 DOI: 10.1016/j.ecoenv.2022.113550] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 06/14/2023]
Abstract
Cadmium (Cd) is a major environmental stressor that induces fetal growth restriction (FGR). Also, changes in gut microbiome diversity-which can be modulated positively by melatonin (Mel) have implications on fetal development and placental functions. Therefore, this study aimed to explore whether the role of Mel in counteracting the Cd-induced FGR by regulating placental barrier injury, endoplasmic reticulum stress (ERS) and mitophagy in pregnant mice is mediated-in part- via the gut microbiota modulations. Pregnant mice were intraperitoneally injected with CdCl2 (5 mg/kg) and Mel (5 mg/kg) once daily, respectively, at the same time from gestational day (GD) 8 to GD18, and then the maternal colon and placental tissues were collected for detection. To investigate the inner relationship between intestinal flora and the protection of Mel on FGR caused by Cd, gut microbiota transplantation (GMT) was carried out from GD0 to GD18 after the removal of intestinal microbiota by antibiotics. Results indicated that Mel relieved barrier injury, ERS and mitophagy in the placenta, and reversed the maternal gut microbiota dysbiosis. The GMT approach suggested a role of intestinal microbiota in placental barrier injury, ERS and mitophagy induced by Cd. Overall, the results highlighted that the intestinal microbiota and gut-placental axis play a central role in the protective effect of Mel against Cd-induced FGR.
Collapse
Affiliation(s)
- Hao Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, PR China
| | - Xiaoyun Liu
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, PR China
| | - Yi Zheng
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, PR China
| | - Xia Zha
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, PR China
| | - Mabrouk Elsabagh
- Department of Animal Production and Technology, Faculty of Agricultural Sciences and Technologies, Niğde Ömer Halisdemir University, Nigde 51240, Turkey; Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Ying Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, PR China
| | - Yi Ma
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, PR China
| | - Juan J Loor
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana 61801, USA
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, PR China
| | - Hongrong Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, PR China.
| |
Collapse
|
69
|
Kaliannan K, Donnell SO, Murphy K, Stanton C, Kang C, Wang B, Li XY, Bhan AK, Kang JX. Decreased Tissue Omega-6/Omega-3 Fatty Acid Ratio Prevents Chemotherapy-Induced Gastrointestinal Toxicity Associated with Alterations of Gut Microbiome. Int J Mol Sci 2022; 23:5332. [PMID: 35628140 PMCID: PMC9140600 DOI: 10.3390/ijms23105332] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 02/04/2023] Open
Abstract
Gastrointestinal toxicity (GIT) is a debilitating side effect of Irinotecan (CPT-11) and limits its clinical utility. Gut dysbiosis has been shown to mediate this side effect of CPT-11 by increasing gut bacterial β-glucuronidase (GUSB) activity and impairing the intestinal mucosal barrier (IMB). We have recently shown the opposing effects of omega-6 (n-6) and omega-3 (n-3) polyunsaturated fatty acids (PUFA) on the gut microbiome. We hypothesized that elevated levels of tissue n-3 PUFA with a decreased n-6/n-3 PUFA ratio would reduce CPT-11-induced GIT and associated changes in the gut microbiome. Using a unique transgenic mouse (FAT-1) model combined with dietary supplementation experiments, we demonstrate that an elevated tissue n-3 PUFA status with a decreased n-6/n-3 PUFA ratio significantly reduces CPT-11-induced weight loss, bloody diarrhea, gut pathological changes, and mortality. Gut microbiome analysis by 16S rRNA gene sequencing and QIIME2 revealed that improvements in GIT were associated with the reduction in the CPT-11-induced increase in both GUSB-producing bacteria (e.g., Enterobacteriaceae) and GUSB enzyme activity, decrease in IMB-maintaining bacteria (e.g., Bifidobacterium), IMB dysfunction and systemic endotoxemia. These results uncover a host-microbiome interaction approach to the management of drug-induced gut toxicity. The prevention of CPT-11-induced gut microbiome changes by decreasing the tissue n-6/n-3 PUFA ratio could be a novel strategy to prevent chemotherapy-induced GIT.
Collapse
Affiliation(s)
- Kanakaraju Kaliannan
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; (K.K.); (B.W.); (X.-Y.L.)
| | - Shane O. Donnell
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (S.O.D.); (C.S.)
- Teagasc Moorepark Food Research Centre, Fermoy, P61 C996 Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland;
| | - Kiera Murphy
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland;
| | - Catherine Stanton
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (S.O.D.); (C.S.)
- Teagasc Moorepark Food Research Centre, Fermoy, P61 C996 Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland;
| | - Chao Kang
- Department of Nutrition, The General Hospital of Western Theater Command, Chengdu 610000, China;
| | - Bin Wang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; (K.K.); (B.W.); (X.-Y.L.)
| | - Xiang-Yong Li
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; (K.K.); (B.W.); (X.-Y.L.)
| | - Atul K. Bhan
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA;
| | - Jing X. Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA; (K.K.); (B.W.); (X.-Y.L.)
| |
Collapse
|
70
|
Olendzki B, Bucci V, Cawley C, Maserati R, McManus M, Olednzki E, Madziar C, Chiang D, Ward DV, Pellish R, Foley C, Bhattarai S, McCormick BA, Maldonado-Contreras A. Dietary manipulation of the gut microbiome in inflammatory bowel disease patients: Pilot study. Gut Microbes 2022; 14:2046244. [PMID: 35311458 PMCID: PMC8942410 DOI: 10.1080/19490976.2022.2046244] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Diet is a modifiable, noninvasive, inexpensive behavior that is crucial in shaping the intestinal microbiome. A microbiome "imbalance" or dysbiosis in inflammatory bowel disease (IBD) is linked to inflammation. Here, we aim to define the impact of specific foods on bacterial species commonly depleted in patients with IBD to better inform dietary treatment. We performed a single-arm, pre-post intervention trial. After a baseline period, a dietary intervention with the IBD-Anti-Inflammatory Diet (IBD-AID) was initiated. We collected stool and blood samples and assessed dietary intake throughout the study. We applied advanced computational approaches to define and model complex interactions between the foods reported and the microbiome. A dense dataset comprising 553 dietary records and 340 stool samples was obtained from 22 participants. Consumption of prebiotics, probiotics, and beneficial foods correlated with increased abundance of Clostridia and Bacteroides, commonly depleted in IBD cohorts. We further show that specific foods categorized as prebiotics or adverse foods are correlated to levels of cytokines in serum (i.e., GM-CSF, IL-6, IL-8, TNF-alpha) that play a central role in IBD pathogenesis. By using robust predictive analytics, this study represents the first steps to detangle diet-microbiome and diet-immune interactions to inform personalized nutrition for patients suffering from dysbiosis-related IBD.
Collapse
Affiliation(s)
- Barbara Olendzki
- Department of Population and Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Vanni Bucci
- Department of Microbiology and Physiological Systems and Program of Microbiome Dynamics. University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Caitlin Cawley
- Department of Microbiology and Physiological Systems and Program of Microbiome Dynamics. University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Rene Maserati
- Department of Microbiology and Physiological Systems and Program of Microbiome Dynamics. University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Margaret McManus
- Center for Clinical and Translational Science, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Effie Olednzki
- Center for Applied Nutrition, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Camilla Madziar
- Department of Population and Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - David Chiang
- Department of Medicine,University of Massachusetts Medical SchoolWorcester, Massachusetts, USA
| | - Doyle V. Ward
- Department of Microbiology and Physiological Systems and Program of Microbiome Dynamics. University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Randall Pellish
- UMass Memorial Medical Center University Campus, Department of Gastroenterology
| | - Christine Foley
- Department of Population and Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Shakti Bhattarai
- Department of Microbiology and Physiological Systems and Program of Microbiome Dynamics. University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Beth A. McCormick
- Department of Microbiology and Physiological Systems and Program of Microbiome Dynamics. University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Ana Maldonado-Contreras
- Department of Microbiology and Physiological Systems and Program of Microbiome Dynamics. University of Massachusetts Medical School, Worcester, Massachusetts, USA,CONTACT Ana Maldonado-Contreras Department of Microbiology and Physiological Systems and Program of Microbiome Dynamics, 368 Plantation Street, Albert Sherman Center, Office AS.81045, Worcester, Massachusetts, 01605, Worcester, Massachusetts, USA
| |
Collapse
|
71
|
Differential modulations of lauric acid and its glycerides on High fat diet-induced metabolic disorders and gut microbiota dysbiosis. Food Res Int 2022; 157:111437. [DOI: 10.1016/j.foodres.2022.111437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 11/18/2022]
|
72
|
Mundula T, Russo E, Curini L, Giudici F, Piccioni A, Franceschi F, Amedei A. Chronic systemic low-grade inflammation and modern lifestyle: the dark role of gut microbiota on related diseases with a focus on pandemic COVID-19. Curr Med Chem 2022; 29:5370-5396. [PMID: 35524667 DOI: 10.2174/0929867329666220430131018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/01/2022] [Accepted: 02/23/2022] [Indexed: 12/12/2022]
Abstract
Inflammation is a physiological, beneficial and auto-limiting response of the host to alarming stimuli. Conversely, a chronic systemic low-grade inflammation (CSLGI), known as a long-time persisting condition, causes organs and host tissues' damage, representing a major risk for chronic diseases. Currently, a worldwide a high incidence of inflammatory chronic diseases is observed, often linked to the lifestyle-related changes occurred in the last decade's society. The mains lifestyle-related factors are a proinflammatory diet, psychological stress, tobacco smoking, alcohol abuse, physical inactivity, and finally indoor living and working with its related consequences such as indoor pollution, artificial light exposure and low vitamin D production. Recent scientific evidences found that gut microbiota (GM) has a main role in shaping the host's health, particularly as CSLGI mediator. As a matter of facts, based on the last discoveries regarding the remarkable GM activity, in this manuscript we focused on the elements of actual lifestyle that influence the composition and function of intestinal microbial community, in order to elicit the CSLGI and its correlated pathologies. In this scenario, we provide a broad review of the interplay between modern lifestyle, GM and CSLGI with a special focus on the COVID symptoms and emerging long-COVID syndrome.
Collapse
Affiliation(s)
- Tiziana Mundula
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Edda Russo
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lavinia Curini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Giudici
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Andrea Piccioni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Franceschi
- Emergency Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
73
|
Wu H, Wang Y, Li H, Meng L, Zheng N, Wang J. Protective Effect of Alkaline Phosphatase Supplementation on Infant Health. Foods 2022; 11:foods11091212. [PMID: 35563935 PMCID: PMC9101100 DOI: 10.3390/foods11091212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/13/2022] [Accepted: 04/13/2022] [Indexed: 12/03/2022] Open
Abstract
Alkaline phosphatase (ALP) is abundant in raw milk. Because of its high heat resistance, ALP negative is used as an indicator of successful sterilization. However, pasteurized milk loses its immune protection against allergy. Clinically, ALP is also used as an indicator of organ diseases. When the activity of ALP in blood increases, it is considered that diseases occur in viscera and organs. Oral administration or injecting ALP will not cause harm to the body and has a variety of probiotic effects. For infants with low immunity, ALP intake is a good prebiotic for protecting the infant’s intestine from potential pathogenic bacteria. In addition, ALP has a variety of probiotic effects for any age group, including prevention and treatment intestinal diseases, allergies, hepatitis, acute kidney injury (AKI), diabetes, and even the prevention of aging. The prebiotic effects of alkaline phosphatase on the health of infants and consumers and the content of ALP in different mammalian raw milk are summarized. The review calls on consumers and manufacturers to pay more attention to ALP, especially for infants with incomplete immune development. ALP supplementation is conducive to the healthy growth of infants.
Collapse
Affiliation(s)
- Haoming Wu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yang Wang
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China;
| | - Huiying Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lu Meng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Zheng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Correspondence: ; Tel.: +86-10-62816069
| | - Jiaqi Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (H.W.); (H.L.); (L.M.); (J.W.)
- Laboratory of Quality and Safety Risk Assessment for Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
74
|
Wei Z, Li F, Pi G. Association Between Gut Microbiota and Osteoarthritis: A Review of Evidence for Potential Mechanisms and Therapeutics. Front Cell Infect Microbiol 2022; 12:812596. [PMID: 35372125 PMCID: PMC8966131 DOI: 10.3389/fcimb.2022.812596] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a multifactorial joint disease characterized by degeneration of articular cartilage, which leads to joints pain, disability and reduced quality of life in patients with OA. Interpreting the potential mechanisms underlying OA pathogenesis is crucial to the development of new disease modifying treatments. Although multiple factors contribute to the initiation and progression of OA, gut microbiota has gradually been regarded as an important pathogenic factor in the development of OA. Gut microbiota can be regarded as a multifunctional “organ”, closely related to a series of immune, metabolic and neurological functions. This review summarized research evidences supporting the correlation between gut microbiota and OA, and interpreted the potential mechanisms underlying the correlation from four aspects: immune system, metabolism, gut-brain axis and gut microbiota modulation. Future research should focus on whether there are specific gut microbiota composition or even specific pathogens and the corresponding signaling pathways that contribute to the initiation and progression of OA, and validate the potential of targeting gut microbiota for the treatment of patients with OA.
Collapse
Affiliation(s)
| | - Feng Li
- *Correspondence: Feng Li, ; Guofu Pi,
| | - Guofu Pi
- *Correspondence: Feng Li, ; Guofu Pi,
| |
Collapse
|
75
|
Preventing Bacterial Translocation in Patients with Leaky Gut Syndrome: Nutrition and Pharmacological Treatment Options. Int J Mol Sci 2022; 23:ijms23063204. [PMID: 35328624 PMCID: PMC8949204 DOI: 10.3390/ijms23063204] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/14/2022] [Accepted: 03/14/2022] [Indexed: 12/11/2022] Open
Abstract
Leaky gut syndrome is a medical condition characterized by intestinal hyperpermeability. Since the intestinal barrier is one of the essential components maintaining homeostasis along the gastrointestinal tract, loss of its integrity due to changes in bacterial composition, decreased expression levels of tight junction proteins, and increased concentration of pro-inflammatory cytokines may lead to intestinal hyperpermeability followed by the development of gastrointestinal and non-gastrointestinal diseases. Translocation of microorganisms and their toxic metabolites beyond the gastrointestinal tract is one of the fallouts of the leaky gut syndrome. The presence of intestinal bacteria in sterile tissues and distant organs may cause damage due to chronic inflammation and progression of disorders, including inflammatory bowel diseases, liver cirrhosis, and acute pancreatitis. Currently, there are no medical guidelines for the treatment or prevention of bacterial translocation in patients with the leaky gut syndrome; however, several studies suggest that dietary intervention can improve barrier function and restrict bacteria invasion. This review contains current literature data concerning the influence of diet, dietary supplements, probiotics, and drugs on intestinal permeability and bacterial translocation.
Collapse
|
76
|
Pasupuleti RR, Tsai PC, Ponnusamy VK, Chen NC. Green sample pre-treatment technique coupled with UHPLC-MS/MS for the rapid biomonitoring of dietary poly-unsaturated (omega) fatty acids to predict health risks. CHEMOSPHERE 2022; 291:132685. [PMID: 34743796 DOI: 10.1016/j.chemosphere.2021.132685] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 09/05/2021] [Accepted: 10/23/2021] [Indexed: 06/13/2023]
Abstract
Polyunsaturated fatty acids (PUFAs) consumption indicates beneficial effects on cardiovascular disease (CVD) and physiological processes in humans. However, the inappropriate ratio of omega-(ω)-PUFA levels in human blood is considered as raising the risk of CVD. Therefore, monitoring dietary ω-FAs in human serum is vital for early diagnosis for individuals to predict CVD risk. This work reports a fast green sample pre-treatment protocol for sensitive and simultaneous monitoring of ω-3-FAs and ω-6-FAs in serum by novel in-syringe-based ultrasonication-assisted alkaline hydrolysis coupled with vortex-induced liquid-liquid microextraction (IS-USAH-VI-LLME) technique connected with UHPLC-MS/MS analysis. Factors affecting extraction recoveries of ten ω-PUFAs by the presented method were well-studied. ω-3 and ω-6 PUFAs demonstrated excellent linearities between the concentrations between 0.1-10,000 ng mL-1 with good regression coefficients between 0.9910-0.9997. The detection and quantification limits were between 0.05-0.35 and 0.16-1.07 ng mL-1, demonstrating that the presented method is highly sensitive and versatile. The precision of the technique was <8.2% that deemed acceptable in clinical analysis. Further, the proposed method was applied for ω-PUFAs analysis in human blood samples, and spiked recoveries showed between 80.32-119.34% with <9.82% precision. Results proved that the developed method is green, sensitive, and reliable to simultaneously determine ten ω-PUFAs in human blood samples for clinical diagnosis applications for predicting health hazards.
Collapse
Affiliation(s)
- Raghavendra Rao Pasupuleti
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan
| | - Pei-Chien Tsai
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan
| | - Vinoth Kumar Ponnusamy
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan; Research Center for Environmental Medicine, Kaohsiung Medical University (KMU), Kaohsiung City, 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital (KMUH), Kaohsiung City, 807, Taiwan; Department of Chemistry, National Sun Yat-sen University (NSYSU), Kaohsiung City, 804, Taiwan; Program of Aquatic Science and Technology, College of Hydrosphere Science, National Kaohsiung University of Science and Technology (NKUST), Kaohsiung City, 811, Taiwan.
| | - Nai-Ching Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung City, 833, Taiwan.
| |
Collapse
|
77
|
Sanchez JMS, DePaula-Silva AB, Libbey JE, Fujinami RS. Role of diet in regulating the gut microbiota and multiple sclerosis. Clin Immunol 2022; 235:108379. [PMID: 32156562 PMCID: PMC7483914 DOI: 10.1016/j.clim.2020.108379] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/18/2019] [Accepted: 03/06/2020] [Indexed: 02/06/2023]
Affiliation(s)
| | | | | | - Robert S. Fujinami
- Corresponding author at: University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT 84112, USA. (R.S. Fujinami)
| |
Collapse
|
78
|
Riedel S, Pheiffer C, Johnson R, Louw J, Muller CJF. Intestinal Barrier Function and Immune Homeostasis Are Missing Links in Obesity and Type 2 Diabetes Development. Front Endocrinol (Lausanne) 2022; 12:833544. [PMID: 35145486 PMCID: PMC8821109 DOI: 10.3389/fendo.2021.833544] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
Noncommunicable diseases, such as type 2 diabetes (T2D), place a burden on healthcare systems worldwide. The rising prevalence of obesity, a major risk factor for T2D, is mainly attributed to the adoption of Westernized diets and lifestyle, which cause metabolic dysfunction and insulin resistance. Moreover, diet may also induce changes in the microbiota composition, thereby affecting intestinal immunity. The critical role of intestinal immunity and intestinal barrier function in the development of T2D is increasingly acknowledged, however, limited studies have investigated the link between intestinal function and metabolic disease. In this review, studies reporting specific roles of the intestinal immune system and intestinal epithelial cells (IECs) in metabolic disease are highlighted. Innate chemokine signaling, eosinophils, immunoglobulin A (IgA), T helper (Th) 17 cells and their cytokines were associated with obesity and/or dysregulated glucose homeostasis. Intestinal epithelial cells (IECs) emerged as critical modulators of obesity and glucose homeostasis through their effect on lipopolysaccharide (LPS) signaling and decontamination. Furthermore, IECs create a link between microbial metabolites and whole-body metabolic function. Future in depth studies of the intestinal immune system and IECs may provide new opportunities and targets to develop treatments and prevention strategies for obesity and T2D.
Collapse
Affiliation(s)
- Sylvia Riedel
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
| | - Carmen Pheiffer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
- Department of Obstetrics and Gynaecology, University of Pretoria, Pretoria, South Africa
| | - Rabia Johnson
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
| | - Johan Louw
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Christo J. F. Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| |
Collapse
|
79
|
The Influence of Gut Microbiota on Neurogenesis: Evidence and Hopes. Cells 2022; 11:cells11030382. [PMID: 35159192 PMCID: PMC8834402 DOI: 10.3390/cells11030382] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
Adult neurogenesis (i.e., the life-long generation of new neurons from undifferentiated neuronal precursors in the adult brain) may contribute to brain repair after damage, and participates in plasticity-related processes including memory, cognition, mood and sensory functions. Among the many intrinsic (oxidative stress, inflammation, and ageing), and extrinsic (environmental pollution, lifestyle, and diet) factors deemed to impact neurogenesis, significant attention has been recently attracted by the myriad of saprophytic microorganismal communities inhabiting the intestinal ecosystem and collectively referred to as the gut microbiota. A growing body of evidence, mainly from animal studies, reveal the influence of microbiota and its disease-associated imbalances on neural stem cell proliferative and differentiative activities in brain neurogenic niches. On the other hand, the long-claimed pro-neurogenic activity of natural dietary compounds endowed with antioxidants and anti-inflammatory properties (such as polyphenols, polyunsaturated fatty acids, or pro/prebiotics) may be mediated, at least in part, by their action on the intestinal microflora. The purpose of this review is to summarise the available information regarding the influence of the gut microbiota on neurogenesis, analyse the possible underlying mechanisms, and discuss the potential implications of this emerging knowledge for the fight against neurodegeneration and brain ageing.
Collapse
|
80
|
Changes in Metabolic Regulation and the Microbiota Composition after Supplementation with Different Fatty Acids in db/db Mice. INTERNATIONAL JOURNAL OF FOOD SCIENCE 2022; 2022:3336941. [PMID: 35036426 PMCID: PMC8759926 DOI: 10.1155/2022/3336941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 12/15/2021] [Indexed: 12/20/2022]
Abstract
Introduction The effects of fatty acids on health vary and depend on the type, amount, and route of consumption. EPA and DHA have a defined role in health, unlike coconut oil. Objective The aim was to investigate the changes in metabolic regulation and the composition of the culture-dependent microbiota after supplementation with different fatty acids in db/db mice. Material and Methods. We were using 32 8-week-old db/db mice, supplemented for eight weeks with EPA/DHA derived from microalgae as well as coconut oil. The lipid, hormonal profiles, and composition of the culture-dependent microbiota and the phylogenetic analysis based on the 16S rRNA gene sequencing were determined for identification of the intestinal microbiota. Results Enriched diet with EPA/DHA reduced TNF-α, C-peptide, insulin resistance, resistin, and the plasma atherogenic index, but increased TC, LDL-c, VLDL-c, and TG without changes in HDL-c. Coconut oil raised the HDL-c, GIP, and TNF-α, with TG, insulin resistance, adiponectin, and C-peptide reduced. Conclusion The most abundant microbial populations were Firmicutes and the least Proteobacteria. EPA/DHA derived from microalgae contributes to improving the systemic inflammatory status, but depressed the diversity of the small intestine microbiota. Coconut oil only decreased the C-peptide, raising TNF-α, with an unfavorable hormonal and lipid profile.
Collapse
|
81
|
Beneficial effects of eicosapentaenoic acid on the metabolic profile of obese female mice entails upregulation of HEPEs and increased abundance of enteric Akkermansia muciniphila. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159059. [PMID: 34619367 PMCID: PMC8627244 DOI: 10.1016/j.bbalip.2021.159059] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 08/30/2021] [Accepted: 09/24/2021] [Indexed: 01/03/2023]
Abstract
Eicosapentaenoic acid (EPA) ethyl esters are of interest given their clinical approval for lowering circulating triglycerides and cardiometabolic disease risk. EPA ethyl esters prevent metabolic complications driven by a high fat diet in male mice; however, their impact on female mice is less studied. Herein, we first investigated how EPA influences the metabolic profile of female C57BL/6J mice consuming a high fat diet. EPA lowered murine fat mass accumulation, potentially through increased biosynthesis of 8-hydroxyeicosapentaenoic acid (HEPE), as revealed by mass spectrometry and cell culture studies. EPA also reversed the effects of a high fat diet on circulating levels of insulin, glucose, and select inflammatory/metabolic markers. Next, we studied if the improved metabolic profile of obese mice consuming EPA was associated with a reduction in the abundance of key gut Gram-negative bacteria that contribute toward impaired glucose metabolism. Using fecal 16S-ribosomal RNA gene sequencing, we found EPA restructured the gut microbiota in a time-dependent manner but did not lower the levels of key Gram-negative bacteria. Interestingly, EPA robustly increased the abundance of the Gram-negative Akkermansia muciniphila, which controls glucose homeostasis. Finally, predictive functional profiling of microbial communities revealed EPA-mediated reversal of high fat diet-associated changes in a wide range of genes related to pathways such as Th-17 cell differentiation and PI3K-Akt signaling. Collectively, these results show that EPA ethyl esters prevent some of the deleterious effects of a high fat diet in female mice, which may be mediated mechanistically through 8-HEPE and the upregulation of intestinal Akkermansia muciniphila.
Collapse
|
82
|
Chataigner M, Lucas C, Di Miceli M, Pallet V, Laye S, Mehaignerie A, Bouvret E, Dinel AL, Joffre C. Dietary Fish Hydrolysate Improves Memory Performance Through Microglial Signature Remodeling During Aging. Front Nutr 2021; 8:750292. [PMID: 34888336 PMCID: PMC8650686 DOI: 10.3389/fnut.2021.750292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/01/2021] [Indexed: 12/28/2022] Open
Abstract
Brain aging is characterized by a chronic low-grade inflammation, which significantly impairs cognitive function. Microglial cells, the immunocompetent cells of the brain, present a different phenotype, switching from a homeostatic signature (M0) to a more reactive phenotype called “MGnD” (microglial neurodegenerative phenotype), leading to a high production of pro-inflammatory cytokines. Furthermore, microglial cells can be activated by age-induced gut dysbiosis through the vagus nerve or the modulation of the peripheral immune system. Nutrients, in particular n-3 long chain polyunsaturated fatty acids (LC-PUFAs) and low molecular weight peptides, display powerful immunomodulatory properties, and can thus prevent age-related cognitive decline. The objective of this study was to investigate the effects of n-3 LC-PUFAs and low molecular weight peptides contained in a marine by-product-derived hydrolysate on microglial phenotypes and intestinal permeability and their consequences on cognition in mice. We demonstrated that the hydrolysate supplementation for 8 weeks prevented short- and long-term memory decline during aging. These observations were linked to the modulation of microglial signature. Indeed, the hydrolysate supplementation promoted homeostatic microglial phenotype by increasing TGF-β1 expression and stimulated phagocytosis by increasing Clec7a expression. Moreover, the hydrolysate supplementation promoted anti-inflammatory intestinal pathway and tended to prevent intestinal permeability alteration occurring during aging. Therefore, the fish hydrolysate appears as an interesting candidate to prevent cognitive decline during aging.
Collapse
Affiliation(s)
- Mathilde Chataigner
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France.,Abyss Ingredients, Caudan, France
| | - Céline Lucas
- NutriBrain Research and Technology Transfer, NutriNeuro, Bordeaux, France
| | - Mathieu Di Miceli
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France.,Worcester Biomedical Research Group, School of Science and the Environment, University of Worcester, Worcester, United Kingdom
| | - Véronique Pallet
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | - Sophie Laye
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| | | | | | - Anne-Laure Dinel
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France.,NutriBrain Research and Technology Transfer, NutriNeuro, Bordeaux, France
| | - Corinne Joffre
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, Bordeaux, France
| |
Collapse
|
83
|
Ren M, Li H, Fu Z, Li Q. Succession Analysis of Gut Microbiota Structure of Participants from Long-Lived Families in Hechi, Guangxi, China. Microorganisms 2021; 9:microorganisms9122524. [PMID: 34946126 PMCID: PMC8703768 DOI: 10.3390/microorganisms9122524] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/28/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota structure has been proposed to be involved in longevity. In this study, trajectories of age-related changes in gut microbiota were analyzed by comparing the gut microbiota composition from long-lived families. A specific bacterial community pattern and signature taxa of long-lived people were found in long-lived families, such as the enrichment of Enterobacteriaceae in all age groups and the higher abundances of Christensenellaceae, Verrucomicrobiaceae, Porphyromonadaceae, Rikenellaceae, Mogibacteriaceae, and Odoribacteraceae in long-lived elderly and the positive correlation between them. The cumulative abundance of the core microbiota was approximately stable along with age, but the genera and species in the core microbiota were rearranged with age, especially in Ruminococcaceae and Lachnospiraceae. Compared with the control group, the proportions of Lachnospiraceae, Roseburia, and Blautia were significantly higher in participants from the long-lived village, but their abundances gradually decreased along with age. Based on functional predictions, the proportions of pathways related to short-chain fatty acid metabolism, amino acid metabolism, and lipoic acid metabolism were significantly higher in the long-lived elderly compared with the offspring group. The trajectory of gut microbiota composition along with age in participants from long-lived families might reveal potential health-promoting metabolic characteristics, which could play an important role in healthy aging.
Collapse
|
84
|
Musazadeh V, Dehghan P, Saleh-Ghadimi S, Abbasalizad Farhangi M. Omega 3-rich Camelina sativa oil in the context of a weight loss program improves glucose homeostasis, inflammation and oxidative stress in patients with NAFLD: A randomised placebo-controlled clinical trial. Int J Clin Pract 2021; 75:e14744. [PMID: 34423525 DOI: 10.1111/ijcp.14744] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Over the past few years, the benefits of omega-3 fatty acids have been reported in the management of non-alcoholic fatty liver disease (NAFLD) complications.This study evaluated the effects of Camelina sativa oil (CSO) supplementation as one of the richest dietary sources of omega-3 fatty acids on glucose homeostasis,inflammation, metabolic endotoxemia, and oxidative stress in NAFLD patients. METHODS A total of 46 patients with NAFLD were allocated to either an intervention (20 g/d CSO) or placebo (20 g/d sunflower oil) group receiving a calorie-restricted diet for 12 weeks. Fasting plasma levels of glycemic indices, hs-CRP, lipopolysaccharide (LPS), antioxidant enzymes activity, total antioxidant capacity (TAC), malondialdehyde (MDA), 8-iso-prostaglandin F2α (8-iso-PGF2α), and uric acid were measured at baseline and post-intervention. RESULTS The CSO supplementation led to significant differences in insulin concentration, homeostasis model assessment of insulin resistance (HOMA-IR), hs-CRP, LPS, TAC, superoxide dismutase (SOD) activity, MDA and 8-iso-PGF2α between the two groups at end of the study (ANCOVA, P < .05). Hs-CRP decreased significantly in both groups (pair-t-test, P < .05). Insulin concentration, quantitative insulin sensitivity check index, LPS, TAC, SOD, glutathione peroxidase activity, MDA and 8-iso-PGF2α changed significantly only in CSO group (P < .05). CONCLUSION These findings indicate that CSO may improve glycemia, inflammation, metabolic endotoxemia, and oxidative stress status in patients with NAFLD.
Collapse
Affiliation(s)
- Vali Musazadeh
- Student Research Committee, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Dehghan
- Nutrition Research Center, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sevda Saleh-Ghadimi
- Nutrition Research Center, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdieh Abbasalizad Farhangi
- Nutrition Research Center, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
85
|
Abramov VM, Kosarev IV, Priputnevich TV, Machulin AV, Abashina TN, Chikileva IO, Donetskova AD, Takada K, Melnikov VG, Vasilenko RN, Khlebnikov VS, Samoilenko VA, Nikonov IN, Sukhikh GT, Uversky VN, Karlyshev AV. S-layer protein 2 of vaginal Lactobacillus crispatus 2029 enhances growth, differentiation, VEGF production and barrier functions in intestinal epithelial cell line Caco-2. Int J Biol Macromol 2021; 189:410-419. [PMID: 34437917 DOI: 10.1016/j.ijbiomac.2021.08.150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022]
Abstract
We have previously demonstrated the ability of the human vaginal strain Lactobacillus crispatus 2029 (LC2029) for strong adhesion to cervicovaginal epithelial cells, expression of the surface layer protein 2 (Slp2), and antagonistic activity against urogenital pathogens. Slp2 forms regular two-dimensional structure around the LC2029 cells,which is secreted into the medium and inhibits intestinal pathogen-induced activation of caspase-9 and caspase-3 in the human intestinal Caco-2 cells. Here, we elucidated the effects of soluble Slp2 on adhesion of proteobacteria pathogens inducing necrotizing enterocolitis (NEC), such as Escherichia coli ATCC E 2348/69, E. coli ATCC 31705, Salmonella Enteritidis ATCC 13076, Campylobacter jejuni ATCC 29428, and Pseudomonas aeruginosa ATCC 27853 to Caco-2 cells, as well as on growth promotion, differentiation, vascular endothelial growth factor (VEGF) production, and intestinal barrier function of Caco-2 cell monolayers. Slp2 acts as anti-adhesion agent for NEC-inducing proteobacteria, promotes growth of immature Caco-2 cells and their differentiation, and enhances expression and functional activity of sucrase, lactase, and alkaline phosphatase. Slp2 stimulates VEGF production, decreases paracellular permeability, and increases transepithelial electrical resistance, strengthening barrier function of Caco-2 cell monolayers. These data support the important role of Slp2 in the early postnatal development of the human small intestine enterocytes.
Collapse
Affiliation(s)
- Vyacheslav M Abramov
- Institute of Immunological Engineering, Lyubuchany 142380, Moscow Region, Russia; Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, Moscow 117997, Russia
| | - Igor V Kosarev
- Institute of Immunological Engineering, Lyubuchany 142380, Moscow Region, Russia; Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, Moscow 117997, Russia
| | - Tatiana V Priputnevich
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, Moscow 117997, Russia
| | - Andrey V Machulin
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", Pushchino 142290, Moscow Region, Russia
| | - Tatiana N Abashina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", Pushchino 142290, Moscow Region, Russia
| | - Irina O Chikileva
- Institute of Immunological Engineering, Lyubuchany 142380, Moscow Region, Russia; Laboratory of Cell Immunity, Blokhin National Research, Center of Oncology Ministry of Health RF, Moscow 115478, Russia
| | | | - Kazuhide Takada
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Vyacheslav G Melnikov
- Gabrichevsky Moscow Research Institute of Epidemiology and Microbiology, Federal Service for Supervision of Consumer Rights Protection and Human Welfare, Moscow 152212, Russia
| | - Raisa N Vasilenko
- Institute of Immunological Engineering, Lyubuchany 142380, Moscow Region, Russia
| | | | - Vladimir A Samoilenko
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", Pushchino 142290, Moscow Region, Russia
| | - Ilya N Nikonov
- Federal State Education Institution of Higher Professional Education Moscow State Academy of Veterinary Medicine and Biotechnology named after K.I. Skryabin, Moscow 109472, Russia
| | - Gennady T Sukhikh
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, Moscow 117997, Russia
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Andrey V Karlyshev
- Department of Science, Engineering and Computing, Kingston University London, Kingston upon Thames KT1 2EE, UK
| |
Collapse
|
86
|
Varesi A, Deumer US, Ananth S, Ricevuti G. The Emerging Role of Gut Microbiota in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS): Current Evidence and Potential Therapeutic Applications. J Clin Med 2021; 10:jcm10215077. [PMID: 34768601 PMCID: PMC8584653 DOI: 10.3390/jcm10215077] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/11/2022] Open
Abstract
The well-known symptoms of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) are chronic pain, cognitive dysfunction, post-exertional malaise and severe fatigue. Another class of symptoms commonly reported in the context of ME/CFS are gastrointestinal (GI) problems. These may occur due to comorbidities such as Crohn's disease or irritable bowel syndrome (IBS), or as a symptom of ME/CFS itself due to an interruption of the complex interplay between the gut microbiota (GM) and the host GI tract. An altered composition and overall decrease in diversity of GM has been observed in ME/CFS cases compared to controls. In this review, we reflect on genetics, infections, and other influences that may factor into the alterations seen in the GM of ME/CFS individuals, we discuss consequences arising from these changes, and we contemplate the therapeutic potential of treating the gut to alleviate ME/CFS symptoms holistically.
Collapse
Affiliation(s)
- Angelica Varesi
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
- Almo Collegio Borromeo, 27100 Pavia, Italy
- Correspondence: (A.V.); (G.R.)
| | - Undine-Sophie Deumer
- Department of Biological Sciences, Faculty of Natural Sciences and Mathematics, University of Cologne, 50674 Cologne, Germany;
| | - Sanjana Ananth
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK;
| | - Giovanni Ricevuti
- Department of Drug Sciences, School of Pharmacy, University of Pavia, 27100 Pavia, Italy
- Correspondence: (A.V.); (G.R.)
| |
Collapse
|
87
|
Intestinal Microbiota as a Contributor to Chronic Inflammation and Its Potential Modifications. Nutrients 2021; 13:nu13113839. [PMID: 34836095 PMCID: PMC8618457 DOI: 10.3390/nu13113839] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota is a crucial factor in maintaining homeostasis. The presence of commensal microorganisms leads to the stimulation of the immune system and its maturation. In turn, dysbiosis with an impaired intestinal barrier leads to accelerated contact of microbiota with the host’s immune cells. Microbial structural parts, i.e., pathogen-associated molecular patterns (PAMPs), such as flagellin (FLG), peptidoglycan (PGN), lipoteichoic acid (LTA), and lipopolysaccharide (LPS), induce inflammation via activation of pattern recognition receptors. Microbial metabolites can also develop chronic low-grade inflammation, which is the cause of many metabolic diseases. This article aims to systematize information on the influence of microbiota on chronic inflammation and the benefits of microbiota modification through dietary changes, prebiotics, and probiotic intake. Scientific research indicates that the modification of the microbiota in various disease states can reduce inflammation and improve the metabolic profile. However, since there is no pattern for a healthy microbiota, there is no optimal way to modify it. The methods of influencing microbiota should be adapted to the type of dysbiosis. Although there are studies on the microbiota and its effects on inflammation, this subject is still relatively unknown, and more research is needed in this area.
Collapse
|
88
|
Serek P, Oleksy-Wawrzyniak M. The Effect of Bacterial Infections, Probiotics and Zonulin on Intestinal Barrier Integrity. Int J Mol Sci 2021; 22:11359. [PMID: 34768787 PMCID: PMC8583036 DOI: 10.3390/ijms222111359] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
The intestinal barrier plays an extremely important role in maintaining the immune homeostasis of the gut and the entire body. It is made up of an intricate system of cells, mucus and intestinal microbiota. A complex system of proteins allows the selective permeability of elements that are safe and necessary for the proper nutrition of the body. Disturbances in the tightness of this barrier result in the penetration of toxins and other harmful antigens into the system. Such events lead to various digestive tract dysfunctions, systemic infections, food intolerances and autoimmune diseases. Pathogenic and probiotic bacteria, and the compounds they secrete, undoubtedly affect the properties of the intestinal barrier. The discovery of zonulin, a protein with tight junction regulatory activity in the epithelia, sheds new light on the understanding of the role of the gut barrier in promoting health, as well as the formation of diseases. Coincidentally, there is an increasing number of reports on treatment methods that target gut microbiota, which suggests that the prevention of gut-barrier defects may be a viable approach for improving the condition of COVID-19 patients. Various bacteria-intestinal barrier interactions are the subject of this review, aiming to show the current state of knowledge on this topic and its potential therapeutic applications.
Collapse
Affiliation(s)
- Paweł Serek
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Monika Oleksy-Wawrzyniak
- Department of Pharmaceutical Microbiology and Parasitology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| |
Collapse
|
89
|
Hosomi R, Matsudo A, Sugimoto K, Shimono T, Kanda S, Nishiyama T, Yoshida M, Fukunaga K. Dietary Eicosapentaenoic Acid and Docosahexaenoic Acid Ethyl Esters Influence the Gut Microbiota and Bacterial Metabolites in Rats. J Oleo Sci 2021; 70:1469-1480. [PMID: 34497184 DOI: 10.5650/jos.ess21189] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Dietary fish oil containing eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) has been reported to affect the diversity and composition of gut microbiota and bacterial metabolites. However, few reports have focused on the effects of EPA and DHA on gut microbiota diversity and bacterial metabolites. This study evaluated the effects of dietary EPA-ethyl ester (EE) and DHA-EE on steroid metabolism, gut microbiota, and bacterial metabolites in Wistar rats. Male rats were fed the experimental diets containing 5% (w/w) soybean oil-EE (SOY diet), EPA-EE (EPA diet), and DHA-EE (DHA diet) for four weeks. The lipid contents in the serum and liver, mRNA expression levels in the liver, and the diversity, composition, and metabolites of the gut microbiota were evaluated. The EPA and DHA diets decreased serum and liver cholesterol contents compared to the SOY diet. In addition, there were no significant changes in gene expression levels related to steroid metabolism in the liver between the EPA and DHA groups. Rats fed the DHA diet had lower microbiota diversity indices, such as Simpson and Shannon indices, than rats fed the SOY and EPA diets. In addition, rats fed EPA and DHA had significant differences in the relative abundance of microbiota at the genus level, such as Phascolarctobacterium, Turicibacter, and [Eubacterium]. Therefore, it was concluded that EPA and DHA have different effects on the diversity and composition of gut microbiota under the experimental conditions employed herein.
Collapse
Affiliation(s)
- Ryota Hosomi
- Department of Life Science and Biotechnology, Kansai University
| | - Anna Matsudo
- Department of Life Science and Biotechnology, Kansai University
| | - Koki Sugimoto
- Department of Life Science and Biotechnology, Kansai University
| | - Takaki Shimono
- Department of Hygiene and Public Health, Kansai Medical University
| | - Seiji Kanda
- Department of Hygiene and Public Health, Kansai Medical University
| | | | | | - Kenji Fukunaga
- Department of Life Science and Biotechnology, Kansai University
| |
Collapse
|
90
|
Zhuang P, Li H, Jia W, Shou Q, Zhu Y, Mao L, Wang W, Wu F, Chen X, Wan X, Wu Y, Liu X, Li Y, Zhu F, He L, Chen J, Zhang Y, Jiao J. Eicosapentaenoic and docosahexaenoic acids attenuate hyperglycemia through the microbiome-gut-organs axis in db/db mice. MICROBIOME 2021; 9:185. [PMID: 34507608 PMCID: PMC8434703 DOI: 10.1186/s40168-021-01126-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/08/2021] [Indexed: 05/04/2023]
Abstract
BACKGROUND Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have been suggested to prevent the development of metabolic disorders. However, their individual role in treating hyperglycemia and the mechanism of action regarding gut microbiome and metabolome in the context of diabetes remain unclear. RESULTS Supplementation of DHA and EPA attenuated hyperglycemia and insulin resistance without changing body weight in db/db mice while the ameliorative effect appeared to be more pronounced for EPA. DHA/EPA supplementation reduced the abundance of the lipopolysaccharide-containing Enterobacteriaceae whereas elevated the family Coriobacteriaceae negatively correlated with glutamate level, genera Barnesiella and Clostridium XlVa associated with bile acids production, beneficial Bifidobacterium and Lactobacillus, and SCFA-producing species. The gut microbiome alterations co-occurred with the shifts in the metabolome, including glutamate, bile acids, propionic/butyric acid, and lipopolysaccharide, which subsequently relieved β cell apoptosis, suppressed hepatic gluconeogenesis, and promoted GLP-1 secretion, white adipose beiging, and insulin signaling. All these changes appeared to be more evident for EPA. Furthermore, transplantation with DHA/EPA-mediated gut microbiota mimicked the ameliorative effect of DHA/EPA on glucose homeostasis in db/db mice, together with similar changes in gut metabolites. In vitro, DHA/EPA treatment directly inhibited the growth of Escherichia coli (Family Enterobacteriaceae) while promoted Coriobacterium glomerans (Family Coriobacteriaceae), demonstrating a causal effect of DHA/EPA on featured gut microbiota. CONCLUSIONS DHA and EPA dramatically attenuated hyperglycemia and insulin resistance in db/db mice, which was mediated by alterations in gut microbiome and metabolites linking gut to adipose, liver and pancreas. These findings shed light into the gut-organs axis as a promising target for restoring glucose homeostasis and also suggest a better therapeutic effect of EPA for treating diabetes. Video abstract.
Collapse
Affiliation(s)
- Pan Zhuang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Haoyu Li
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Wei Jia
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Qiyang Shou
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310005, Zhejiang, China
| | - Ya'er Zhu
- Analysis Center of Agrobiology and Environmental Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Lei Mao
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Wenqiao Wang
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Fei Wu
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Xiaoqian Chen
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Xuzhi Wan
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Yuqi Wu
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Xiaohui Liu
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Yin Li
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Fanghuan Zhu
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Lilin He
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Jingnan Chen
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Yu Zhang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| | - Jingjing Jiao
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition of Affiliated Second Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
91
|
Li J, Wan Y, Zheng Z, Zhang H, Li Y, Guo X, Li K, Li D. Maternal n-3 polyunsaturated fatty acids restructure gut microbiota of offspring mice and decrease their susceptibility to mammary gland cancer. Food Funct 2021; 12:8154-8168. [PMID: 34291263 DOI: 10.1039/d1fo00906k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Our previous studies have revealed that a maternal diet rich in n-3 polyunsaturated fatty acids (PUFAs) is associated with decreased mammary cancer risk in offspring. However, the underlying mechanism remains unclear. The present study aimed to investigate the possible mechanism by which maternal n-3 PUFAs decrease the mammary cancer risk of offspring in terms of gut microbiota. C57BL/6 pregnant mice were fed a control standard chow (CON), fish oil supplemented diet (n-3 Sup-FO), flaxseed oil supplemented diet (n-3 Sup-FSO) or n-3 PUFA deficient diet (n-3 Def) (n = 10) throughout gestation and lactation. After weaning, all offspring were fed a AIN-93G diet. The tumor incidence and volume were significantly increased in n-3 Def offspring compared with the other groups. Maternal n-3 PUFA supplementation resulted in a significantly increased α-diversity of the gut microbiota in n-3 Sup-FO and n-3 Sup-FSO offspring compared with that in n-3 Def offspring. The relative abundances of Akkermansia, Lactobacillus and Mucispirillum observed in adult offspring of both the n-3 Sup-FO and n-3 Sup-FSO groups were higher than those observed in the control group, whereas the maternal n-3 Def diet was associated with decreased abundances of Lactobacillus, Bifidobacterium and Barnesiella in 7-week-old offspring. The levels of the pro-inflammatory factors IL-1β, IL-6 and TNF-α were significantly lower in n-3 PUFA supplemented offspring than in n-3 Def offspring. In addition, the abundance of Mucispirillum was positively associated with the concentration of the anti-inflammatory factor IL-10, whereas the abundances of Bifidobacterium and Akkermansia were negatively associated with IL-1β and IL-6, respectively. Based on the bacterial composition of the gut microbiota, metabolites were predicted and the results showed that arachidonic acid metabolism and the MAPK signaling pathways were more enriched, while the butyric acid metabolic pathway was less enriched in offspring of the n-3 Def group than in those of the other three groups. Our findings suggest that decreased pro-inflammatory factors and changed gut microbiota are associated with the protective effects of maternal n-3 PUFAs against offspring's mammary tumorigenesis.
Collapse
Affiliation(s)
- Jiaomei Li
- Institute of Nutrition and Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China.
| | | | | | | | | | | | | | | |
Collapse
|
92
|
White B, Sterrett JD, Grigoryan Z, Lally L, Heinze JD, Alikhan H, Lowry CA, Perez LJ, DeSipio J, Phadtare S. Characterization of gut microbiome and metabolome in Helicobacter pylori patients in an underprivileged community in the United States. World J Gastroenterol 2021; 27:5575-5594. [PMID: 34588753 PMCID: PMC8433610 DOI: 10.3748/wjg.v27.i33.5575] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/02/2021] [Accepted: 08/13/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori), a bacterium that infects approximately half of the world’s population, is associated with various gastrointestinal diseases, including peptic ulcers, non-ulcer dyspepsia, gastric adenocarcinoma, and gastric lymphoma. As the burden of antibiotic resistance increases, the need for new adjunct therapies designed to facilitate H. pylori eradication and reduce negative distal outcomes associated with infection has become more pressing. Characterization of the interactions between H. pylori, the fecal microbiome, and fecal fatty acid metabolism, as well as the mechanisms underlying these interactions, may offer new therapeutic approaches.
AIM To characterize the gut microbiome and metabolome in H. pylori patients in a socioeconomically challenged and underprivileged inner-city community.
METHODS Stool samples from 19 H. pylori patients and 16 control subjects were analyzed. 16S rRNA gene sequencing was performed on normalized pooled amplicons using the Illumina MiSeq System using a MiSeq reagent kit v2. Alpha and beta diversity analyses were performed in QIIME 2. Non-targeted fatty acid analysis of the samples was carried out using gas chromatography-mass spectrometry, which measures the total content of 30 fatty acids in stool after conversion into their corresponding fatty acid methyl esters. Multi-dimensional scaling (MDS) was performed on Bray-Curtis distance matrices created from both the metabolomics and microbiome datasets and a Procrustes test was performed on the metabolomics and microbiome MDS coordinates.
RESULTS Fecal microbiome analysis showed that alpha diversity was lowest in H. pylori patients over 40 years of age compared to control subjects of similar age group. Beta diversity analysis of the samples revealed significant differences in microbial community structure between H. pylori patients and control subjects across all ages. Thirty-eight and six taxa had lower and higher relative abundance in H. pylori patients, respectively. Taxa that were enriched in H. pylori patients included Atopobium, Gemellaceae, Micrococcaceae, Gemellales and Rothia (R. mucilaginosa). Notably, relative abundance of the phylum Verrucomicrobia was decreased in H. pylori patients compared to control subjects. Procrustes analysis showed a significant relationship between the microbiome and metabolome datasets. Stool samples from H. pylori patients showed increases in several fatty acids including the polyunsaturated fatty acids (PUFAs) 22:4n6, 22:5n3, 20:3n6 and 22:2n6, while decreases were noted in other fatty acids including the PUFA 18:3n6. The pattern of changes in fatty acid concentration correlated to the Bacteroidetes:Firmicutes ratio determined by 16S rRNA gene analysis.
CONCLUSION This exploratory study demonstrates H. pylori-associated changes to the fecal microbiome and fecal fatty acid metabolism. Such changes may have implications for improving eradication rates and minimizing associated negative distal outcomes.
Collapse
Affiliation(s)
- Brian White
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| | - John D Sterrett
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, United States
| | - Zoya Grigoryan
- Department of Internal Medicine, Lenox Hill Hospital, NYC, NY 10075, United States
| | - Lauren Lally
- Department of Internal Medicine, Thomas Jefferson University Hospital, Philadelphia, PA 19107, United States
| | - Jared D Heinze
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, United States
| | - Hyder Alikhan
- Department of Biological Sciences, Rowan University, Glassboro, NJ 08028, United States
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, United States
| | - Lark J Perez
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ 08028, United States
| | - Joshua DeSipio
- Department of Gastroenterology, Cooper University Hospital, Camden, NJ 08103, United States
| | - Sangita Phadtare
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| |
Collapse
|
93
|
Sears B, Saha AK. Dietary Control of Inflammation and Resolution. Front Nutr 2021; 8:709435. [PMID: 34447777 PMCID: PMC8382877 DOI: 10.3389/fnut.2021.709435] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/13/2021] [Indexed: 12/18/2022] Open
Abstract
The healing of any injury requires a dynamic balance of initiation and resolution of inflammation. This hypothesis-generating review presents an overview of the various nutrients that can act as signaling agents to modify the metabolic responses essential for the optimal healing of injury-induced inflammation. In this hypothesis-generating review, we describe a defined nutritional program consisting of an integrated interaction of a calorie-restricted anti-inflammatory diet coupled with adequate levels of omega-3 fatty acids and sufficient levels of dietary polyphenols that can be used in clinical trials to treat conditions associated with insulin resistance. Each dietary intervention works in an orchestrated systems-based approach to reduce, resolve, and repair the tissue damage caused by any inflammation-inducing injury. The orchestration of these specific nutrients and their signaling metabolites to facilitate healing is termed the Resolution Response. The final stage of the Resolution Response is the activation of intracellular 5' adenosine monophosphate-activated protein kinase (AMPK), which is necessary to repair tissue damaged by the initial injury-induced inflammation. The dietary optimization of the Resolution Response can be personalized to the individual by using standard blood markers. Once each of those markers is in their appropriate ranges, activation of intracellular AMPK will be facilitated. Finally, we outline how the resulting activation of AMPK will affect a diverse number of other intercellular signaling systems leading to an extended healthspan.
Collapse
Affiliation(s)
- Barry Sears
- Inflammation Research Foundation, Peabody, MA, United States
| | | |
Collapse
|
94
|
Nana G, Mitra S, Watson H, Young C, Wood HM, Perry SL, Race AD, Quirke P, Toogood GJ, Loadman PM, Hull MA. Luminal Bioavailability of Orally Administered ω-3 PUFAs in the Distal Small Intestine, and Associated Changes to the Ileal Microbiome, in Humans with a Temporary Ileostomy. J Nutr 2021; 151:2142-2152. [PMID: 34036331 PMCID: PMC8349127 DOI: 10.1093/jn/nxab113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/22/2021] [Accepted: 03/29/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Oral administration of purified omega-3 (ω-3) PUFAs is associated with changes to the fecal microbiome. However, it is not known whether this effect is associated with increased PUFA concentrations in the gut. OBJECTIVES We investigated the luminal bioavailability of oral ω-3 PUFAs (daily dose 1 g EPA and 1g DHA free fatty acid equivalents as triglycerides in soft-gel capsules, twice daily) and changes to the gut microbiome, in the ileum. METHODS Ileostomy fluid (IF) and blood were obtained at baseline, after first capsule dosing (median 2 h), and at a similar time after final dosing on day 28, in 11 individuals (median age 63 y) with a temporary ileostomy. Fatty acids were measured by LC-tandem MS. The ileal microbiome was characterized by 16S rRNA PCR and Illumina sequencing. RESULTS There was a mean 6.0 ± 9.8-fold and 6.6 ± 9.6-fold increase in ileal EPA and DHA concentrations (primary outcome), respectively, at 28 d, which was associated with increased RBC ω-3 PUFA content (P ≤ 0.05). The first oral dose did not increase the ileal ω-3 PUFA concentration except in 4 individuals, who displayed high luminal EPA and DHA concentrations, which reduced to concentrations similar to the overall study population at day 28, suggesting physiological adaptation. Bacteroides, Clostridium, and Streptococcus were abundant bacterial genera in the ileum. Ileal microbiome variability over time and between individuals was large, with no consistent change associated with acute ω-3 PUFA dosing. However, high concentrations of EPA and DHA in IF on day 28 were associated with higher abundance of Bacteroides (r2 > 0.86, P < 0.05) and reduced abundance of other genera, including Actinomyces (r2 > 0.94, P < 0.05). CONCLUSIONS Oral administration of ω-3 PUFAs leads to increased luminal ω-3 PUFA concentrations and changes to the microbiome, in the ileum of individuals with a temporary ileostomy. This study is registered on the ISRCTN registry as ISRCTN14530452.
Collapse
Affiliation(s)
- Gael Nana
- Leeds Institute of Medical Research, St James's University Hospital, University of Leeds, Leeds, United Kingdom
- Department of Hepatobiliary Surgery, St James's University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Suparna Mitra
- Leeds Institute of Medical Research, St James's University Hospital, University of Leeds, Leeds, United Kingdom
| | - Henry Watson
- Leeds Institute of Medical Research, St James's University Hospital, University of Leeds, Leeds, United Kingdom
- Department of Hepatobiliary Surgery, St James's University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Caroline Young
- Leeds Institute of Medical Research, St James's University Hospital, University of Leeds, Leeds, United Kingdom
| | - Henry M Wood
- Leeds Institute of Medical Research, St James's University Hospital, University of Leeds, Leeds, United Kingdom
| | - Sarah L Perry
- Leeds Institute of Medical Research, St James's University Hospital, University of Leeds, Leeds, United Kingdom
| | - Amanda D Race
- Institute of Cancer Therapeutics, University of Bradford, Bradford, United Kingdom
| | - Philip Quirke
- Leeds Institute of Medical Research, St James's University Hospital, University of Leeds, Leeds, United Kingdom
| | - Giles J Toogood
- Department of Hepatobiliary Surgery, St James's University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Paul M Loadman
- Institute of Cancer Therapeutics, University of Bradford, Bradford, United Kingdom
| | - Mark A Hull
- Leeds Institute of Medical Research, St James's University Hospital, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
95
|
Chen S, Xiao Y, Liu Y, Tian X, Wang W, Jiang L, Wu W, Zhang T, Cai W, Wang Y. Fish oil-based lipid emulsion alleviates parenteral nutrition-associated liver diseases and intestinal injury in piglets. JPEN J Parenter Enteral Nutr 2021; 46:709-720. [PMID: 34291472 DOI: 10.1002/jpen.2229] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Thisstudy aimed to investigate the impact of fish oil-based lipid emulsion (FO) on enterohepatic injuries and intestinal microbiota in piglets of parenteral nutrition (PN). METHODS Newborn piglets were divided into three groups, including enteral diet (the controls), PN with 100% FO and PN with medium-chain triglyceride/long-chain triglyceride-based lipid emulsion (MCT/LCT) for 14 days. Serum biochemical indicators, hepatic and intestinal histology, and expression of genes associated with inflammation, oxidative stress, and lipid metabolism were measured. The bile acid (BA) profiles in serum and the taxonomic composition of the gut microbiome in different intestinal segments were analyzed. RESULTS Compared with MCT/LCT-piglets, FO reduced inflammation, promoted fatty acid oxidation, and decreased oxidative stress in the liver. In the intestine, FO decreased intestinal inflammation and intestinal permeability, leading to reduced lipopolysaccharide entry into the blood circulation relative to MCT/LCT-piglets. PN groups have dominant contents of Proteobacteria and Bacteroides, whereas the control group have Firmicutes at the phylum level. FO altered the taxonomic compositions of the gut microbiome in different segments, increased the relative abundance of Bacteroidaceae in ileum, and Rikenellaceae and Ruminococcaceae in the colon. FO treatment shifted BA composition ratio in serum and had a lower ratio of secondary BAs to primary BAs. CONCLUSION FO alleviates PNLAD and intestinal injury by regulating the homeostasis of BAs' enterohepatic circulation and altering microbiota composition in different intestinal segments.
Collapse
Affiliation(s)
- Shanshan Chen
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongtao Xiao
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai, China
| | - Yang Liu
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinbei Tian
- Shanghai Institute for Pediatric Research, Shanghai, China
| | - Weipeng Wang
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lu Jiang
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai, China
| | - Wenjie Wu
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tian Zhang
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Cai
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai, China.,Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Wang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai, China
| |
Collapse
|
96
|
The role of precision nutrition in the modulation of microbial composition and function in people with inflammatory bowel disease. Lancet Gastroenterol Hepatol 2021; 6:754-769. [PMID: 34270915 DOI: 10.1016/s2468-1253(21)00097-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/05/2021] [Accepted: 03/11/2021] [Indexed: 12/18/2022]
Abstract
Inflammatory bowel diseases, principally Crohn's disease and ulcerative colitis, are multifactorial chronic conditions. Alterations in gut microbial patterns partly affect disease onset and severity. Moreover, the evolution of dietary patterns, and their effect on gut microbial behaviour, have been shown to play a crucial role in disease processes. This Viewpoint reviews the role of dietary patterns, their influence on the structure and function of the gut microbiome, and their effects on inflammation and immunity in individuals with inflammatory bowel disease. We also discuss innovative dietary intervention strategies, summarise findings that have been used to develop recommendations for clinical practice, and provide suggestions for the design of future studies for development of precision nutrition in patients with inflammatory bowel disease.
Collapse
|
97
|
Sasaki Y, Honyashiki M, Kinoshita T, Matsui A, Nakashoji A, Inagawa T, Ikezawa S, Yoshimura N, Yamamura R, Amano M, Tomo Y, Tachimori H, Matsuoka YJ, Okubo R. Perilla Oil and Bifidobacterium for Alleviating Fear of Cancer Recurrence in Breast Cancer Survivors: Study Protocol for a Three-Arm Phase II Randomized Controlled Study (POB Study). Methods Protoc 2021; 4:mps4030046. [PMID: 34287371 PMCID: PMC8293445 DOI: 10.3390/mps4030046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 11/25/2022] Open
Abstract
The fear of cancer recurrence (FCR) is the most common and most severe unmet need among cancer survivors. Safe treatments for the FCR that are easily disseminated are greatly needed. Our primary aim is a preliminary evaluation of the efficacy and effect size of perilla oil, which is rich in omega-3 fatty acids, and Bifidobacterium, a probiotic, on FCR in breast cancer survivors after the completion of chemotherapy. This study has been planned as an exploratory clinical study (phase II) and will be conducted as a three-arm, 12-week parallel group, masked-rater randomized controlled trial. Fifteen participants will be randomized with 1:1:1 allocation to receive Bifidobacterium plus perilla oil, Bifidobacterium alone, or no intervention (control). Interventions will end within 12 weeks after the random allocation of each participant. The participants will be outpatients with invasive breast cancer aged 20 years or older whose chemotherapy was completed at least 6 months before registration; hormone therapy may be ongoing. The primary outcome will be severity of FCR at 12 weeks assessed by masked raters using the 4-item Concerns about Recurrence Scale concerning overall fear of recurrence. The study protocol for the current study is registered in the Japan Registry of Clinical Trials (jRCTs031200029).
Collapse
Affiliation(s)
- Yohei Sasaki
- Department of Clinical Epidemiology, Translational Medical Center, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan; (Y.S.); (Y.T.); (H.T.)
| | - Mina Honyashiki
- Department of Psychiatry, Tokyo Medical University, Tokyo 113-8510, Japan;
| | - Takayuki Kinoshita
- Department of Breast Surgery, National Hospital Organization Tokyo Medical Center, Tokyo 187-8551, Japan; (T.K.); (A.M.); (A.N.)
| | - Akira Matsui
- Department of Breast Surgery, National Hospital Organization Tokyo Medical Center, Tokyo 187-8551, Japan; (T.K.); (A.M.); (A.N.)
| | - Ayako Nakashoji
- Department of Breast Surgery, National Hospital Organization Tokyo Medical Center, Tokyo 187-8551, Japan; (T.K.); (A.M.); (A.N.)
| | - Takuma Inagawa
- Department of Psychiatry, National Center Hospital of Neurology and Psychiatry, Tokyo 187-8551, Japan; (T.I.); (S.I.); (N.Y.)
| | - Satoru Ikezawa
- Department of Psychiatry, National Center Hospital of Neurology and Psychiatry, Tokyo 187-8551, Japan; (T.I.); (S.I.); (N.Y.)
| | - Naoki Yoshimura
- Department of Psychiatry, National Center Hospital of Neurology and Psychiatry, Tokyo 187-8551, Japan; (T.I.); (S.I.); (N.Y.)
| | - Ryodai Yamamura
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan;
| | | | - Yui Tomo
- Department of Clinical Epidemiology, Translational Medical Center, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan; (Y.S.); (Y.T.); (H.T.)
| | - Hisateru Tachimori
- Department of Clinical Epidemiology, Translational Medical Center, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan; (Y.S.); (Y.T.); (H.T.)
| | - Yutaka J. Matsuoka
- Division of Health Care Research, Center for Public Health Sciences, National Cancer Center Japan, Tokyo 104-0045, Japan;
- Lifestyle Medicine, Cooperative Graduate Program, The Jikei University Graduate School of Medicine, Tokyo 105-8461, Japan
| | - Ryo Okubo
- Department of Clinical Epidemiology, Translational Medical Center, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan; (Y.S.); (Y.T.); (H.T.)
- Correspondence: ; Tel.: +81-42-341-2712 (ext. 5843)
| |
Collapse
|
98
|
Influences of dietary oils and fats, and the accompanied minor content of components on the gut microbiota and gut inflammation: A review. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.05.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
99
|
Thanabalan A, Kiarie EG. Influence of Feeding Omega-3 Polyunsaturated Fatty Acids to Broiler Breeders on Indices of Immunocompetence, Gastrointestinal, and Skeletal Development in Broiler Chickens. Front Vet Sci 2021; 8:653152. [PMID: 34262961 PMCID: PMC8273488 DOI: 10.3389/fvets.2021.653152] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/01/2021] [Indexed: 01/24/2023] Open
Abstract
Modern broiler chickens are associated with rapid growth rates and superior feed efficiency. However, they are also susceptible to physiological and metabolic disorders (e.g., skin lesions, lameness, sudden death, enteric diseases, myopathies) that exert substantial economic losses to producers. This is further exacerbated by consumer pressure and mandated cessation of production practices such as indiscriminate use of antimicrobial growth promoters. Manipulation of broiler breeder (BB) nutrition and management can influence chick quality, robustness, and resilience to stressors in the production environment. The present review examines the role of feeding BB functional polyunsaturated omega-3 fatty acids (n-3 PUFA) and subsequent impact on the indices of immunocompetence, skeletal, and gastrointestinal (GIT) development in broiler chickens. Research in mammalian and avian models led evidence that perinatal feeding of long chain n-3 PUFA such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) engender transgenerational effects through regulation of a variety of biological processes including development of vital organs such as skeleton, brain and GIT. It is shown that feeding poultry breeders n-3 PUFA decreases inflammatory states and enriches hatching eggs with n-3 PUFA and immunoglobulins. Further evidence also shows that after 15 days of incubation, chicken embryos preferentially utilize long chain n-3 PUFA-critical for optimal cell, tissues, and organ development. Enrichment of n-3 PUFA in newly hatchling tissues reduce proinflammatory eicosanoids with consequences of enhanced bone mineralization. Dietary n-3 PUFA also modulates breeder GIT microbiota with consequences of microbial colonization and succession in chicks. As well, research shows that feeding poultry breeders n-3 PUFA bolsters progeny immunocompetence through enhanced passive immunity and antibody titres against routine vaccination. In conclusion, it appears that chicks may benefit from the incorporation of n-3 PUFA in the breeder diets; however, little attention is paid to fatty acids composition in breeder nutrition. We also highlight gaps in knowledge and future research perspectives.
Collapse
Affiliation(s)
| | - Elijah G Kiarie
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
100
|
Effect of High-Fat Diet on the Intestinal Flora in Letrozole-Induced Polycystic Ovary Syndrome Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6674965. [PMID: 34257691 PMCID: PMC8257354 DOI: 10.1155/2021/6674965] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 05/10/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022]
Abstract
Aim The aim of this study was to explore whether letrozole and high-fat diets (HFD) can induce obese insulin-resistant polycystic ovary syndrome (PCOS) with intestinal flora dysbiosis in a rat model. We compared the changes in the intestinal flora of letrozole-induced rats fed with HFD or normal chow, to explore the effects of HFD and letrozole independently and synergistically on the intestinal flora. Methods Five-week-old female Sprague Dawley (SD) rats were divided into four groups: control (C) group fed with regular diet; L1 group administered with letrozole and fed with regular diet; L2 group received letrozole and fed with HFD; and HFD group fed with HFD. At the end of the experiment, ovarian morphology, hormones, metabolism, oxidative stress, and inflammatory status of all rats were studied. 16S rDNA high-throughput sequencing was used to profile microbial communities, and various multivariate analysis approaches were used to quantitate microbial composition, abundance, and diversity. Results Compared to the C group, the increased plasma fasting insulin and glucose, HOMA-IR, triglyceride, testosterone, and malondialdehyde were significantly higher in the L2 group, while high-density lipoprotein cholesterol was significantly lower in the L1 group and L2 group. The indices of Chao1 and the Abundance-based Coverage Estimator (ACE) (α-diversity) in the L2 and HFD groups were significantly lower than that in the C group. Bray–Curtis dissimilarity based principal coordinate analysis (PCoA) plots and analysis of similarities (ANOSIM) test showed obvious separations between the L2 group and C group, between the HFD group and C group, and between the L2 and HFD groups. At the phylum level, Firmicutes and ratio of Firmicutes and Bacteroidetes (F/B ratio) were increased in the L2 group; Bacteroidetes was decreased in the L2 and HFD groups. No significant differences in bacterial abundance between the C group and L1 group were observed at the phylum level. Based on linear discriminant analysis (LDA) effect size (LEfSe) analysis, the bacterial genera (the relative abundance > 0.1%, LDA > 3, p < 0.05) were selected as candidate bacterial signatures. They showed that the abundance of Vibrio was significantly increased in the L1 group; Bacteroides and Phascolarctobacterium were enriched in the HFD group, and Bacteroides, Phascolarctobacterium, Blautia, Parabacteroides, Akkermansia [Ruminococcus]_torques_group, and Anaerotruncus were enriched in the L2 group. Conclusion The effect of letrozole on intestinal flora was not significant as HFD. HFD could destroy the balance of intestinal flora and aggravate the intestinal flora dysbiosis in PCOS. Letrozole-induced rats fed with HFD have many characteristics like human PCOS, including some metabolic disorders and intestinal flora dysbiosis. The dysbiosis was characterized by an increased Firmicutes/Bacteroidetes ratio, an expansion of Firmicutes, a contraction of Bacteroidetes, and the decreased microbial richness. Beta-diversity also showed significant differences in intestinal microflora, compared with control rats.
Collapse
|