1
|
Zheng PC, Pan XQ, Zhou YJ, Lai KP, Li R, Zhang XX. Unraveling the impact of micro- and nano-sized polymethyl methacrylate on gut microbiota and liver lipid metabolism: Insights from oral exposure studies. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 373:126157. [PMID: 40157484 DOI: 10.1016/j.envpol.2025.126157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/14/2025] [Accepted: 03/27/2025] [Indexed: 04/01/2025]
Abstract
Microplastics, particularly polymethyl methacrylate (PMMA), have emerged as significant environmental pollutants, with growing concerns about their impact on various biological processes. However, the effects of chronic PMMA exposure on hepatic lipid metabolism remain insufficiently studied. This research aimed to examine the consequences of chronic exposure to PMMA particles of different sizes (100 nm and 2 μm) on hepatic lipid metabolism in mice. Female C57BL/6J mice were administered PMMA particles in drinking water over an 8-week period, and the effects on intestinal and liver morphology and function were evaluated. Histopathological analyses, gut microbiota profiling, and serum and liver assays were conducted to assess oxidative stress, lipid metabolism-related biomarkers, and liver metabolomics. The results revealed that PMMA particles accumulated in both the liver and colon, causing liver injury characterized by elevated ALT and AST levels. The exposure also induced oxidative stress by inhibiting the NRF2/HO-1 signaling pathway. Furthermore, PMMA exposure resulted in significant alterations to the gut microbiota and hepatic metabolism. These changes were linked to increased microbial diversity, which impacted cholesterol metabolism through the gut-liver axis. Additionally, the activation of the PI3K/AKT/PPARγ signaling pathway disrupted hepatic lipid metabolism, leading to increased cholesterol synthesis and hepatic lipid accumulation. This study underscores the potential of PMMA to disrupt both hepatic lipid metabolism and gut microbiota composition, suggesting a novel mechanism by which PMMA exposure could contribute to metabolic disorders and liver disease.
Collapse
Affiliation(s)
- Peng Chen Zheng
- College of Basic Medical Sciences, Guilin Medical University, Guilin, 541100, China
| | - Xin Qiang Pan
- College of Basic Medical Sciences, Guilin Medical University, Guilin, 541100, China
| | - Yi Jiong Zhou
- College of Basic Medical Sciences, Guilin Medical University, Guilin, 541100, China
| | - Keng Po Lai
- Key Laboratory of Environmental Pollution and Integrative Omics, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, Guilin, 541100, China; Department of Applied Science, Hong Kong Metropolitan University, Hong Kong, China
| | - Rong Li
- College of Basic Medical Sciences, Guilin Medical University, Guilin, 541100, China; Key Laboratory of Environmental Pollution and Integrative Omics, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, Guilin, 541100, China.
| | - Xiao Xi Zhang
- Key Laboratory of Environmental Pollution and Integrative Omics, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, Guilin, 541100, China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, 541100, China.
| |
Collapse
|
2
|
Xie B, Wu Y, Liu Z, Huang Y, Lu Q, Bian A, Han B, Yan Y, Lai Y, He B, Li Y, Yan F, Yan L, Chen F. Association of dietary and plasma fatty acids with periodontitis: Results from the 2009-2014 National Health and Nutrition Examination Survey and Mendelian randomization study. J Am Dent Assoc 2025; 156:198-210.e28. [PMID: 39864010 DOI: 10.1016/j.adaj.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND The authors aimed to explore the association of fatty acids with periodontitis and its severity and to assess causality using Mendelian randomization (MR) analyses. METHODS Data for participants with complete data were extracted from the 2009-2014 National Health and Nutrition Examination Survey. Weighted logistic regression was used to explore the relationship between dietary fatty acids and periodontitis and its severity. Univariable and multivariable MR analyses were performed to explore the causal association between plasma fatty acids and periodontitis. RESULTS Two types of saturated fatty acids (hexadecanoic C16:0, octadecanoic C18:0) and monounsaturated fatty acids (hexadecenoic C16:1, docosenoic C22:1) and 3 types of polyunsaturated fatty acids (eicosatetraenoic C20:4, eicosapentaenoic C20:5, docosahexaenoic C22:6) were positively associated with periodontitis. Conversely, octadecadienoic (C18:2, a type of polyunsaturated fatty acid), total polyunsaturated fatty acids, and omega-6 fatty acids were negatively associated with periodontitis. Similar association patterns were also found between these fatty acids and the severity of periodontitis. Results of MR analyses revealed that no significant association was found between plasma fatty acids and periodontitis. CONCLUSIONS The authors provided evidence of significant associations between certain fatty acids and periodontitis and its severity, highlighting their contributory role, although the evidence does not support a causal role based on the results of MR-based analyses. PRACTICAL IMPLICATIONS The results of this study support a contributory or inhibitory role of fatty acids in the progression of periodontitis, although not as a direct cause, and underscored the importance of maintaining a balanced dietary lipid profile for periodontal health.
Collapse
|
3
|
Xiong N, Zhang W, Zhang Y, Nie C, Dan H. Association between nutrient intake and inflammatory bowel disease risk: Insights from NHANES data and dose-response analysis. Nutrition 2025; 131:112632. [PMID: 39700661 DOI: 10.1016/j.nut.2024.112632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/15/2024] [Accepted: 10/25/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND The role of dietary and nutritional factors in inflammatory bowel disease (IBD) remains poorly understood, necessitating further investigation. This study aims to explore the association between nutrient intake and the risk of IBD. METHODS This cross-sectional study utilized data from the 2009-2010 NHANES cycle, focusing on individuals with complete 24-hour dietary intake records and clinically assessed IBD information. Nutrient intake was assessed through dietary recalls and supplement data. Associations between nutrient intake and IBD risk were analyzed by propensity score matching (PSM) with balanced baseline characteristics and logistic regression. Dose-response relationships were examined by restricted cubic splines (RCS). Statistical significance was set at P < 0.05, and analyses were conducted using R software. RESULTS The study included 4,072 participants with clinically assessed IBD and complete nutrient intake data. In adjusted analyses, lower intake of vitamin B3, copper, phosphorus, selenium, sodium, and protein below the recommended dietary allowance may increase the risk of developing IBD. Similarly, reduced intake of vitamin B6, vitamin E, and total PUFA is associated with elevated susceptibility to IBD. CONCLUSION This study elucidates the intricate relationship between nutrient intake and the onset of IBD, underscoring the importance of maintaining a balanced diet for gastrointestinal health. These findings emphasize the significance of informed dietary choices in promoting intestinal wellness and potentially reducing the risk of IBD development.
Collapse
Affiliation(s)
- Nuoya Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Wei Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yajie Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Chunlai Nie
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongxia Dan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
4
|
Lammi C, Ottaviano E, Fiore G, Bollati C, d'Adduzio L, Fanzaga M, Ceccarani C, Vizzuso S, Zuccotti G, Borghi E, Verduci E. Effect of docosahexaenoic acid as an anti-inflammatory for Caco-2 cells and modulating agent for gut microbiota in children with obesity (the DAMOCLE study). J Endocrinol Invest 2025; 48:465-481. [PMID: 39186221 PMCID: PMC11785711 DOI: 10.1007/s40618-024-02444-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024]
Abstract
PURPOSE Docosahexaenoic acid (DHA) is a long-chain omega-3 polyunsaturated fatty acid. We investigated the dual health ability of DHA to modulate gut microbiota in children with obesity and to exert anti-inflammatory activity on human intestinal Caco-2 cells. METHODS In a pilot study involving 18 obese children (8-14 years), participants received a daily DHA supplement (500 mg/day) and dietary intervention from baseline (T0) to 4 months (T1), followed by dietary intervention alone from 4 months (T1) to 8 months (T2). Fecal samples, anthropometry, biochemicals and dietary assessment were collected at each timepoint. At preclinical level, we evaluated DHA's antioxidant and anti-inflammatory effects on Caco-2 cells stimulated with Hydrogen peroxide (H2O2) and Lipopolysaccharides (LPS), by measuring also Inducible nitric oxide synthase (iNOS) levels and cytokines, respectively. RESULTS Ten children were included in final analysis. No major changes were observed for anthropometric and biochemical parameters, and participants showed a low dietary compliance at T1 and T2. DHA supplementation restored the Firmicutes/Bacteroidetes ratio that was conserved also after the DHA discontinuation at T2. DHA supplementation drove a depletion in Ruminococcaceae and Dialisteraceae, and enrichment in Bacteroidaceae, Oscillospiraceae, and Akkermansiaceae. At genus level, Allisonella was the most decreased by DHA supplementation. In Caco-2 cells, DHA decreased H2O2-induced reactive oxygen species (ROS) and nitric oxide (NO) production via iNOS pathway modulation. Additionally, DHA modulated proinflammatory (IL-1β, IL-6, IFN-γ, TNF-α) and anti-inflammatory (IL-10) cytokine production in LPS-stimulated Caco-2 cells. CONCLUSION An improvement in gut dysbiosis of children with obesity seems to be triggered by DHA and to continue after discontinuation. The ability to modulate gut microbiota, matches also with an anti-inflammatory effect of DHA on Caco-2 cells.
Collapse
Affiliation(s)
- C Lammi
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | - E Ottaviano
- Department of Health Sciences, University of Milan, 20142, Milan, Italy
| | - G Fiore
- Department of Health Sciences, University of Milan, 20142, Milan, Italy.
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Via Lodovico Castelvetro 32, 20154, Milan, Italy.
| | - C Bollati
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | - L d'Adduzio
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | - M Fanzaga
- Department of Pharmaceutical Sciences, University of Milan, 20133, Milan, Italy
| | - C Ceccarani
- Institute for Biomedical Technologies, CNR, Segrate, Italy
| | - S Vizzuso
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Via Lodovico Castelvetro 32, 20154, Milan, Italy
| | - G Zuccotti
- Department of Pediatrics, Vittore Buzzi Children's Hospital, University of Milan, Via Lodovico Castelvetro 32, 20154, Milan, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, 20157, Milan, Italy
| | - E Borghi
- Department of Health Sciences, University of Milan, 20142, Milan, Italy
| | - E Verduci
- Department of Health Sciences, University of Milan, 20142, Milan, Italy
- Metabolic Diseases Unit, Department of Paediatrics, Vittore Buzzi Children's Hospital, University of Milan, 20157, Milan, Italy
| |
Collapse
|
5
|
Young MG, Straub TJ, Worby CJ, Metsky HC, Gnirke A, Bronson RA, van Dijk LR, Desjardins CA, Matranga C, Qu J, Villicana JB, Azimzadeh P, Kau A, Dodson KW, Schreiber HL, Manson AL, Hultgren SJ, Earl AM. Distinct Escherichia coli transcriptional profiles in the guts of recurrent UTI sufferers revealed by pangenome hybrid selection. Nat Commun 2024; 15:9466. [PMID: 39487120 PMCID: PMC11530686 DOI: 10.1038/s41467-024-53829-7] [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: 07/23/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024] Open
Abstract
Low-abundance members of microbial communities are difficult to study in their native habitats, including Escherichia coli, a minor but common inhabitant of the gastrointestinal tract, and key opportunistic pathogen of the urinary tract. While multi-omic analyses have detailed interactions between uropathogenic Escherichia coli (UPEC) and the bladder mediating urinary tract infection (UTI), little is known about UPEC in its pre-infection reservoir, the gastrointestinal tract, partly due to its low relative abundance (<1%). To sensitively explore the genomes and transcriptomes of diverse gut E. coli, we develop E. coli PanSelect, which uses probes designed to specifically capture E. coli's broad pangenome. We demonstrate its ability to enrich diverse E. coli by orders of magnitude, in a mock community and in human stool from a study investigating recurrent UTI (rUTI). Comparisons of transcriptomes between gut E. coli of women with and without history of rUTI suggest rUTI gut E. coli are responding to increased oxygen and nitrate, suggestive of mucosal inflammation, which may have implications for recurrent disease. E. coli PanSelect is well suited for investigations of in vivo E. coli biology in other low-abundance environments, and the framework described here has broad applicability to other diverse, low-abundance organisms.
Collapse
Affiliation(s)
- Mark G Young
- Infectious Disease & Microbiome Program, Broad Institute, Cambridge, MA, 02142, USA
| | - Timothy J Straub
- Infectious Disease & Microbiome Program, Broad Institute, Cambridge, MA, 02142, USA
| | - Colin J Worby
- Infectious Disease & Microbiome Program, Broad Institute, Cambridge, MA, 02142, USA
| | - Hayden C Metsky
- Infectious Disease & Microbiome Program, Broad Institute, Cambridge, MA, 02142, USA
| | - Andreas Gnirke
- Infectious Disease & Microbiome Program, Broad Institute, Cambridge, MA, 02142, USA
| | - Ryan A Bronson
- Infectious Disease & Microbiome Program, Broad Institute, Cambridge, MA, 02142, USA
| | - Lucas R van Dijk
- Infectious Disease & Microbiome Program, Broad Institute, Cambridge, MA, 02142, USA
- Delft Bioinformatics Lab, Delft University of Technology, Van Mourik Broekmanweg 6, Delft, 2628 XE, The Netherlands
| | | | - Christian Matranga
- Infectious Disease & Microbiome Program, Broad Institute, Cambridge, MA, 02142, USA
| | - James Qu
- Infectious Disease & Microbiome Program, Broad Institute, Cambridge, MA, 02142, USA
| | - Jesús Bazan Villicana
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Philippe Azimzadeh
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew Kau
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Karen W Dodson
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Henry L Schreiber
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Abigail L Manson
- Infectious Disease & Microbiome Program, Broad Institute, Cambridge, MA, 02142, USA
| | - Scott J Hultgren
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Ashlee M Earl
- Infectious Disease & Microbiome Program, Broad Institute, Cambridge, MA, 02142, USA.
| |
Collapse
|
6
|
Winter SE, Bäumler AJ. Gut dysbiosis: Ecological causes and causative effects on human disease. Proc Natl Acad Sci U S A 2023; 120:e2316579120. [PMID: 38048456 PMCID: PMC10722970 DOI: 10.1073/pnas.2316579120] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/02/2023] [Indexed: 12/06/2023] Open
Abstract
The gut microbiota plays a role in many human diseases, but high-throughput sequence analysis does not provide a straightforward path for defining healthy microbial communities. Therefore, understanding mechanisms that drive compositional changes during disease (gut dysbiosis) continues to be a central goal in microbiome research. Insights from the microbial pathogenesis field show that an ecological cause for gut dysbiosis is an increased availability of host-derived respiratory electron acceptors, which are dominant drivers of microbial community composition. Similar changes in the host environment also drive gut dysbiosis in several chronic human illnesses, and a better understanding of the underlying mechanisms informs approaches to causatively link compositional changes in the gut microbiota to an exacerbation of symptoms. The emerging picture suggests that homeostasis is maintained by host functions that control the availability of resources governing microbial growth. Defining dysbiosis as a weakening of these host functions directs attention to the underlying cause and identifies potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Sebastian E. Winter
- Department of Medicine, Division of Infectious Diseases, University of California, Davis, CA95616
- Department of Medical Microbiology and Immunology, University of California, Davis, CA95616
| | - Andreas J. Bäumler
- Department of Medical Microbiology and Immunology, University of California, Davis, CA95616
| |
Collapse
|
7
|
Kayama H, Takeda K. Emerging roles of host and microbial bioactive lipids in inflammatory bowel diseases. Eur J Immunol 2023; 53:e2249866. [PMID: 37191284 DOI: 10.1002/eji.202249866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/11/2023] [Accepted: 05/15/2023] [Indexed: 05/17/2023]
Abstract
The intestinal tract harbors diverse microorganisms, host- and microbiota-derived metabolites, and potentially harmful dietary antigens. The epithelial barrier separates the mucosa, where diverse immune cells exist, from the lumen to avoid excessive immune reactions against microbes and dietary antigens. Inflammatory bowel disease (IBD), such as ulcerative colitis and Crohn's disease, is characterized by a chronic and relapsing disorder of the gastrointestinal tract. Although the precise etiology of IBD is still largely unknown, accumulating evidence suggests that IBD is multifactorial, involving host genetics and microbiota. Alterations in the metabolomic profiles and microbial community are features of IBD. Advances in mass spectrometry-based lipidomic technologies enable the identification of changes in the composition of intestinal lipid species in IBD. Because lipids have a wide range of functions, including signal transduction and cell membrane formation, the dysregulation of lipid metabolism drastically affects the physiology of the host and microorganisms. Therefore, a better understanding of the intimate interactions of intestinal lipids with host cells that are implicated in the pathogenesis of intestinal inflammation might aid in the identification of novel biomarkers and therapeutic targets for IBD. This review summarizes the current knowledge on the mechanisms by which host and microbial lipids control and maintain intestinal health and diseases.
Collapse
Affiliation(s)
- Hisako Kayama
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- WPI, Osaka University, Suita, Osaka, Japan
- Institute for Advanced Co-Creation Studies, Osaka University, Suita, Osaka, Japan
| | - Kiyoshi Takeda
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- WPI, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Center for Infection Disease Education and Research, Osaka University, Suita, Japan
| |
Collapse
|
8
|
Martens N, Zhan N, Voortman G, Leijten FPJ, van Rheenen C, van Leerdam S, Geng X, Huybrechts M, Liu H, Jonker JW, Kuipers F, Lütjohann D, Vanmierlo T, Mulder MT. Activation of Liver X Receptors and Peroxisome Proliferator-Activated Receptors by Lipid Extracts of Brown Seaweeds: A Potential Application in Alzheimer's Disease? Nutrients 2023; 15:3004. [PMID: 37447330 DOI: 10.3390/nu15133004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
The nuclear liver X receptors (LXRα/β) and peroxisome proliferator-activated receptors (PPARα/γ) are involved in the regulation of multiple biological processes, including lipid metabolism and inflammation. The activation of these receptors has been found to have neuroprotective effects, making them interesting therapeutic targets for neurodegenerative disorders such as Alzheimer's Disease (AD). The Asian brown seaweed Sargassum fusiforme contains both LXR-activating (oxy)phytosterols and PPAR-activating fatty acids. We have previously shown that dietary supplementation with lipid extracts of Sargassum fusiforme prevents disease progression in a mouse model of AD, without inducing adverse effects associated with synthetic pan-LXR agonists. We now determined the LXRα/β- and PPARα/γ-activating capacity of lipid extracts of six European brown seaweed species (Alaria esculenta, Ascophyllum nodosum, Fucus vesiculosus, Himanthalia elongata, Saccharina latissima, and Sargassum muticum) and the Asian seaweed Sargassum fusiforme using a dual luciferase reporter assay. We analyzed the sterol and fatty acid profiles of the extracts by GC-MS and UPLC MS/MS, respectively, and determined their effects on the expression of LXR and PPAR target genes in several cell lines using quantitative PCR. All extracts were found to activate LXRs, with the Himanthalia elongata extract showing the most pronounced efficacy, comparable to Sargassum fusiforme, for LXR activation and transcriptional regulation of LXR-target genes. Extracts of Alaria esculenta, Fucus vesiculosus, and Saccharina latissima showed the highest capacity to activate PPARα, while extracts of Alaria esculenta, Ascophyllum nodosum, Fucus vesiculosus, and Sargassum muticum showed the highest capacity to activate PPARγ, comparable to Sargassum fusiforme extract. In CCF-STTG1 astrocytoma cells, all extracts induced expression of cholesterol efflux genes (ABCG1, ABCA1, and APOE) and suppressed expression of cholesterol and fatty acid synthesis genes (DHCR7, DHCR24, HMGCR and SREBF2, and SREBF1, ACACA, SCD1 and FASN, respectively). Our data show that lipophilic fractions of European brown seaweeds activate LXRs and PPARs and thereby modulate lipid metabolism. These results support the potential of brown seaweeds in the prevention and/or treatment of neurodegenerative diseases and possibly cardiometabolic and inflammatory diseases via concurrent activation of LXRs and PPARs.
Collapse
Affiliation(s)
- Nikita Martens
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, B-3590 Hasselt, Belgium
| | - Na Zhan
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Gardi Voortman
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Frank P J Leijten
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Connor van Rheenen
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Suzanne van Leerdam
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
| | - Xicheng Geng
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Michiel Huybrechts
- Department of Environmental Biology, Center for Environmental Sciences, Hasselt University, B-3590 Diepenbeek, Belgium
| | - Hongbing Liu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Johan W Jonker
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Folkert Kuipers
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, D-53127 Bonn, Germany
| | - Tim Vanmierlo
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, B-3590 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Monique T Mulder
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
| |
Collapse
|
9
|
Zou H, Gong Y, Ye H, Yuan C, Li T, Zhang J, Ren L. Dietary regulation of peroxisome proliferator-activated receptors in metabolic syndrome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154904. [PMID: 37267691 DOI: 10.1016/j.phymed.2023.154904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 05/15/2023] [Accepted: 05/28/2023] [Indexed: 06/04/2023]
Abstract
BACKGROUND Peroxisome proliferator-activated receptors (PPARs) are a class of ligand-activated nuclear transcription factors, members of the type nuclear receptor superfamily, with three subtypes, namely PPARα, PPARβ/δ, and PPARγ, which play a key role in the metabolic syndrome. In the past decades, a large number of studies have shown that natural products can act by regulating metabolic pathways mediated by PPARs. PURPOSE This work summarizes the physiological importance and clinical significance of PPARs and reviews the experimental evidence that natural products mediate metabolic syndrome via PPARs. METHODS This study reviews relevant literature on clinical trials, epidemiology, animals, and cell cultures published in NCBI PubMed, Scopus, Web of Science, Google Scholar, and other databases from 2001 to October 2022. Search keywords were "natural product" OR "botanical" OR "phytochemical" AND "PPAR" as well as free text words. RESULTS The modulatory involvement of PPARs in the metabolic syndrome has been supported by prior research. It has been observed that many natural products can treat metabolic syndrome by altering PPARs. The majority of currently described natural compounds are mild PPAR-selective agonists with therapeutic effects that are equivalent to synthetic medicines but less harmful adverse effects. CONCLUSION PPAR agonists can be combined with natural products to treat and prevent metabolic syndrome. Further human investigations are required because it is unknown how natural products cause harm and how they might have negative impacts.
Collapse
Affiliation(s)
- Haoyang Zou
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Yiyao Gong
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Haiqing Ye
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Cuiping Yuan
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences, Changchun 130033, China
| | - Tiezhu Li
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences, Changchun 130033, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Li Ren
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| |
Collapse
|
10
|
Yang S, Wang C, Huang X, Jansen CA, Savelkoul HFJ, Liu G. Linoleic acid stimulation results in TGF-β1 production and inhibition of PEDV infection in vitro. Virology 2023; 581:89-96. [PMID: 36934552 DOI: 10.1016/j.virol.2023.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/26/2023] [Accepted: 03/10/2023] [Indexed: 03/15/2023]
Abstract
Linoleic acid (LA) is recommended to improve pork quality. However, whether it affects the intestinal immune response in pigs is still unclear. Our ex vivo experiments demonstrated that LA stimulation resulted in increased frequencies of Tregs in PBMCs but not in Peyer's Patches (PPs). The results of RT-qPCR, flow cytometry, and ELISA indicated that LA increased the TGF-β1 expression level in DCs isolated from PEDV infected pigs. Furthermore, RT-qPCR and flow cytometry results demonstrated that TGF-β1 was associated with higher frequencies of Tregs both in PBMCs and PPs. Additional investigations showed that TGF-β1 inhibited PEDV infection in vitro. Besides, knocking-out TGF-β1 in IPEC-J2 cells resulted in higher viral load. Taken together, our results demonstrated that LA stimulation resulted in enhanced production of TGF-β1 by DC, which resulted in higher frequencies of Tregs production and inhibition of PEDV infection.
Collapse
Affiliation(s)
- Shanshan Yang
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, China; Cell Biology and Immunology Group, Wageningen University and Research, Wageningen, the Netherlands
| | - Caiying Wang
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, China; Cell Biology and Immunology Group, Wageningen University and Research, Wageningen, the Netherlands
| | - Xin Huang
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, China
| | - Christine A Jansen
- Cell Biology and Immunology Group, Wageningen University and Research, Wageningen, the Netherlands
| | - Huub F J Savelkoul
- Cell Biology and Immunology Group, Wageningen University and Research, Wageningen, the Netherlands
| | - Guangliang Liu
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, China.
| |
Collapse
|
11
|
Panezai J, van Dyke T. Polyunsaturated Fatty Acids and Their Immunomodulatory Actions in Periodontal Disease. Nutrients 2023; 15:nu15040821. [PMID: 36839179 PMCID: PMC9965392 DOI: 10.3390/nu15040821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/20/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Polyunsaturated fatty acids (PUFAs) are a diverse set of molecules with remarkable contributions to human physiology. They not only serve as sources of fuel but also cellular structural components as well as substrates that provide bioactive metabolites. A growing body of evidence demonstrates their role in inflammation. Inflammation in the presence of a polymicrobial biofilm contributes to the pathology of periodontitis. The role PUFAs in modulating immuno-inflammatory reactions in periodontitis is only beginning to be uncovered as research continues to unravel their far-reaching immunologic implications.
Collapse
Affiliation(s)
- Jeneen Panezai
- Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA 02142, USA
- Department of Microbiology, Faculty of Life Sciences and Informatics, Balochistan University of Information Technology, Engineering and Management Sciences, Quetta 87300, Pakistan
| | - Thomas van Dyke
- Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA 02142, USA
- Centre for Clinical and Translational Research, The Forsyth Institute, Cambridge, MA 02142, USA
- Department of Oral Medicine, Infection and Immunity, Harvard Faculty of Medicine, Boston, MA 02115, USA
- Correspondence:
| |
Collapse
|
12
|
Belanche A, Diago S, Fondevila M. Inclusion of a fish oil processing fraction as additive in diets for weaning piglets. Anim Feed Sci Technol 2022. [DOI: 10.1016/j.anifeedsci.2022.115517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
13
|
Badia-Soteras A, de Vries J, Dykstra W, Broersen LM, Verkuyl JM, Smit AB, Verheijen MHG. High-Throughput Analysis of Astrocyte Cultures Shows Prevention of Reactive Astrogliosis by the Multi-Nutrient Combination Fortasyn Connect. Cells 2022; 11:cells11091428. [PMID: 35563732 PMCID: PMC9099974 DOI: 10.3390/cells11091428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 04/07/2022] [Accepted: 04/20/2022] [Indexed: 12/23/2022] Open
Abstract
Astrocytes are specialized glial cells that tile the central nervous system (CNS) and perform numerous essential functions. Astrocytes react to various forms of CNS insults by altering their morphology and molecular profile, through a process known as reactive astrogliosis. Accordingly, astrocyte reactivity is apparent in many neurodegenerative diseases, among which one is Alzheimer’s disease (AD). Recent clinical trials on early-stage AD have demonstrated that Fortasyn Connect (FC), a multi-nutrient combination providing specific precursors and cofactors for phospholipid synthesis, helps to maintain neuronal functional connectivity and cognitive performance of patients. Several studies have shown that FC may act through its effects on neuronal survival and synaptogenesis, leading to reduced astrocyte reactivity, but whether FC can directly counteract astrocyte reactivity remains to be elucidated. Hence, we developed an in vitro model of reactive astrogliosis using the pro-inflammatory cytokines TNF-α and IFN-γ together with an automated high-throughput assay (AstroScan) to quantify molecular and morphological changes that accompany reactive astrogliosis. Next, we showed that FC is potent in preventing cytokine-induced reactive astrogliosis, a finding that might be of high relevance to understand the beneficial effects of FC-based interventions in the context of neurodegenerative diseases.
Collapse
Affiliation(s)
- Aina Badia-Soteras
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (A.B.-S.); (J.d.V.); (W.D.); (A.B.S.)
| | - Janneke de Vries
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (A.B.-S.); (J.d.V.); (W.D.); (A.B.S.)
| | - Werner Dykstra
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (A.B.-S.); (J.d.V.); (W.D.); (A.B.S.)
| | - Laus M. Broersen
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (L.M.B.); (J.M.V.)
| | - Jan Martin Verkuyl
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (L.M.B.); (J.M.V.)
| | - August B. Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (A.B.-S.); (J.d.V.); (W.D.); (A.B.S.)
| | - Mark H. G. Verheijen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (A.B.-S.); (J.d.V.); (W.D.); (A.B.S.)
- Correspondence:
| |
Collapse
|
14
|
Sundaram TS, Giromini C, Rebucci R, Pistl J, Bhide M, Baldi A. Role of omega-3 polyunsaturated fatty acids, citrus pectin, and milk-derived exosomes on intestinal barrier integrity and immunity in animals. J Anim Sci Biotechnol 2022; 13:40. [PMID: 35399093 PMCID: PMC8996583 DOI: 10.1186/s40104-022-00690-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/07/2022] [Indexed: 11/10/2022] Open
Abstract
The gastrointestinal tract of livestock and poultry is prone to challenge by feedborne antigens, pathogens, and other stress factors in the farm environment. Excessive physiological inflammation and oxidative stress that arises firstly disrupts the intestinal epithelial barrier followed by other components of the gastrointestinal tract. In the present review, the interrelationship between intestinal barrier inflammation and oxidative stress that contributes to the pathogenesis of inflammatory bowel disease was described. Further, the role of naturally existing immunomodulatory nutrients such as the omega-3 polyunsaturated fatty acids, citrus pectin, and milk-derived exosomes in preventing intestinal barrier inflammation was discussed. Based on the existing evidence, the possible molecular mechanism of these bioactive nutrients in the intestinal barrier was outlined for application in animal diets.
Collapse
Affiliation(s)
- Tamil Selvi Sundaram
- Department of Veterinary Science for Health, Animal Production and Food Safety, University of Milan, Via Trentacoste 2, 20134, Milan, Italy.
- University of Veterinary Medicine and Pharmacy in Košice, Komenského 68/73, 04181, Košice, Slovakia.
| | - Carlotta Giromini
- Department of Veterinary Science for Health, Animal Production and Food Safety, University of Milan, Via Trentacoste 2, 20134, Milan, Italy
| | - Raffaella Rebucci
- Department of Veterinary Science for Health, Animal Production and Food Safety, University of Milan, Via Trentacoste 2, 20134, Milan, Italy
| | - Juraj Pistl
- University of Veterinary Medicine and Pharmacy in Košice, Komenského 68/73, 04181, Košice, Slovakia
| | - Mangesh Bhide
- University of Veterinary Medicine and Pharmacy in Košice, Komenského 68/73, 04181, Košice, Slovakia
| | - Antonella Baldi
- Department of Veterinary Science for Health, Animal Production and Food Safety, University of Milan, Via Trentacoste 2, 20134, Milan, Italy
| |
Collapse
|
15
|
Wu XY, Nie L, Lu XJ, Fei CJ, Chen J. Molecular characterization, expression and functional analysis of large yellow croaker (Larimichthys crocea) peroxisome proliferator-activated receptor gamma. FISH & SHELLFISH IMMUNOLOGY 2022; 123:50-60. [PMID: 35227879 DOI: 10.1016/j.fsi.2022.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 06/14/2023]
Abstract
The peroxisome proliferator-activated receptor gamma (PPARγ) are nuclear receptors with distinct roles in energy metabolism and immunity. Although extensively studied in mammals, immunomodulatory roles of this molecule in teleost fish remain to be investigated. In this study, large yellow croaker (Larimichthys crocea) PPARγ (LcPPARγ) sequence was cloned, which encodes a polypeptide of 541 amino acids that include signature domains belonging to the nuclear receptor superfamily. Phylogenetically, LcPPARγ was most closely related to PPARγ derived from European sea bass (Dicentrarchus labrax). Quantitative analysis shown a ubiquitous expression of this molecule, with highest expression level detected in the intestine. The expression of LcPPARγ was decreased in the intestine, muscle, body kidney, spleen and head kidney-derived monocytes/macrophages (MO/MФs) over the course of Vibrio alginolyticus (V. alginolyticus) infection. In contrast, an up-regulation of LcPPARγ was observed in head kidney-derived MO/MФs following docosahexaenoic acid (DHA) treatment. This increase in LcPPARγ leads to an up-regulation of LcCD11b and LcCD18 and an enhancement of complement-mediated phagocytosis. Furthermore, cytokine secretions of V. alginolyticus-stimulated MO/MФs were modulated following LcPPARγ activations that up-regulated the expression of LcIL-10, while decreased the expression of LcIL-1β, LcTNF-α and LcTGF-β1. Overall, our results indicated that LcPPARγ plays a role in regulating functions of MO/MФs and likely contribute to MO/MФs polarization.
Collapse
Affiliation(s)
- Xiang-Yu Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo City, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo City, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, School of Marine Sciences, Ningbo University, Ningbo City, China
| | - Li Nie
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo City, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, School of Marine Sciences, Ningbo University, Ningbo City, China
| | - Xin-Jiang Lu
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo City, China
| | - Chen-Jie Fei
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo City, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, School of Marine Sciences, Ningbo University, Ningbo City, China.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo City, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo City, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, School of Marine Sciences, Ningbo University, Ningbo City, China.
| |
Collapse
|
16
|
Suau R, Pardina E, Domènech E, Lorén V, Manyé J. The Complex Relationship Between Microbiota, Immune Response and Creeping Fat in Crohn's Disease. J Crohns Colitis 2022; 16:472-489. [PMID: 34528668 DOI: 10.1093/ecco-jcc/jjab159] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the last decade, there has been growing interest in the pathological involvement of hypertrophic mesenteric fat attached to the serosa of the inflamed intestinal segments involved in Crohn's disease [CD], known as creeping fat. In spite of its protective nature, creeping fat harbours an aberrant inflammatory activity which, in an already inflamed intestine, may explain why creeping fat is associated with a greater severity of CD. The transmural inflammation of CD facilitates the interaction of mesenteric fat with translocated intestinal microorganisms, contributing to activation of the immune response. This may be not the only way in which microorganisms alter the homeostasis of this fatty tissue: intestinal dysbiosis may also impair xenobiotic metabolism. All these CD-related alterations have a functional impact on nuclear receptors such as the farnesoid X receptor or the peroxisome proliferator-activated receptor γ, which are implicated in regulation of the immune response, adipogenesis and the maintenance of barrier function, as well as on creeping fat production of inflammatory-associated cells such as adipokines. The dysfunction of creeping fat worsens the inflammatory course of CD and may favour intestinal fibrosis and fistulizing complications. However, our current knowledge of the pathophysiology and pathogenic role of creeping fat is controversial and a better understanding might provide new therapeutic targets for CD. Here we aim to review and update the key cellular and molecular alterations involved in this inflammatory process that link the pathological components of CD with the development of creeping fat.
Collapse
Affiliation(s)
- Roger Suau
- IBD Research Group, 'Germans Trias i Pujol' Research Institute (IGTP), Badalona (Catalonia), Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain
| | - Eva Pardina
- Biochemistry and Molecular Biomedicine Department, University of Barcelona, Barcelona (Catalonia), Spain
| | - Eugeni Domènech
- IBD Research Group, 'Germans Trias i Pujol' Research Institute (IGTP), Badalona (Catalonia), Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain.,Gastroenterology Department, 'Germans Trias i Pujol' University Hospital, Badalona (Catalonia), Spain
| | - Violeta Lorén
- IBD Research Group, 'Germans Trias i Pujol' Research Institute (IGTP), Badalona (Catalonia), Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain
| | - Josep Manyé
- IBD Research Group, 'Germans Trias i Pujol' Research Institute (IGTP), Badalona (Catalonia), Spain.,Centro de Investigación Biomédica en Red (CIBER), Madrid, Spain
| |
Collapse
|
17
|
Mabaya L, Matarira HT, Tanyanyiwa DM, Musarurwa C, Mukwembi J, Mudluli TE, Marera T. Polyunsaturated Fatty Acid Composition in Breast Milk Plasma of HIV-infected and Uninfected Mothers in Relation to Infant Clinical Outcomes. Nutr Metab Insights 2022; 15:11786388211072768. [PMID: 35153488 PMCID: PMC8829717 DOI: 10.1177/11786388211072768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/18/2021] [Indexed: 11/28/2022] Open
Abstract
Background: The increased coverage of prevention of mother to child transmission (PMTCT) services has significantly reduced paediatric HIV infection incidence. The aim of the study was to compare breast milk omega-6 and omega-3 polyunsaturated fatty acid profiles of HIV infected and uninfected mothers and determine the association between fatty acid profiles and postnatal transmission of HIV, morbidity/mortality of HIV exposed and unexposed infants. Methods: A prospective cohort study of 57 HIV infected and 57 HIV uninfected lactating mothers was conducted in Gweru, Zimbabwe from July 2019 to March 2020. The women’s 114 babies (term and preterm) were also enrolled and stratified by HIV exposure and infection status. The mother-infant pairs were followed up at 6 weeks, 16 weeks and 6 months postpartum to determine, HIV transmission rate, breast milk polyunsaturated fatty acid profiles as well as infant clinical outcomes. Results: The mean breast milk docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) levels in HIV uninfected mothers (0.82 ± 0.92; 0.47 ± 0.75 μg/ml respectively) at 6 weeks postpartum were significantly higher compared to those of HIV infected mothers (0.33 ± 0.32; 0.08 ± 0.14 μg/ml) respectively. The same pattern was observed at 16 weeks postpartum in terms of DHA and EPA mean concentration. However, the arachidonic acid (AA) levels and AA/DHA ratio measured at 6 weeks postpartum were significantly higher in HIV infected mothers (2.31 ± 2.01; 17.18 ± 52.47 respectively) compared to HIV uninfected mothers (0.82 ± 0.54; 9.71 ± 21.80; P < .001). A higher morbidity rate was observed amongst HIV exposed infants than HIV unexposed infants (3.26 ± 0.13; 2.49 ± 0.09; P < .001) respectively. A significant positive correlation was observed between AA and infant morbidity (r = .388; P < .001). Conclusion: Deficiencies in breast milk omega-3 fatty acids were observed in HIV infected women. Maintaining a healthy balance between omega-6 and omega-3 fatty acid diets is critical for breast feeding mothers regardless of their HIV status. The adverse clinical outcomes observed amongst HIV exposed infants emphasise their vulnerability under conditions of maternal universal antiretroviral therapy.
Collapse
Affiliation(s)
- Lucy Mabaya
- Department of Chemical Pathology, Faculty of Medicine and Health Sciences, Midlands State University, Gweru, Zimbabwe
- Department of Chemical Pathology, University of Zimbabwe Medical School Harare, Harare, Zimbabwe
| | - Hilda Tendisa Matarira
- Department of Chemical Pathology, University of Zimbabwe Medical School Harare, Harare, Zimbabwe
| | - Donald Moshen Tanyanyiwa
- Division of Chemical Pathology, Department of Pathology, University of Witwatersrand/ National Health Laboratory Services, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
| | - Cuthbert Musarurwa
- Department of Chemical Pathology, University of Zimbabwe Medical School Harare, Harare, Zimbabwe
| | - Johannes Mukwembi
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health Sciences, Midlands State University, Gweru, Zimbabwe
| | - Taona Emmah Mudluli
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Midlands State University, Gweru, Zimbabwe
| | - Tatenda Marera
- Scientific and Industrial Research and Development Centre, Hatcliffe, Harare, Zimbabwe
| |
Collapse
|
18
|
Chatterjee D, Chatterjee A, Kalra D, Kapoor A, Vijay S, Jain S. Role of adjunct use of omega 3 fatty acids in periodontal therapy of periodontitis. A systematic review and meta-analysis. J Oral Biol Craniofac Res 2022; 12:55-62. [PMID: 34760614 PMCID: PMC8566999 DOI: 10.1016/j.jobcr.2021.10.005] [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] [Received: 04/24/2021] [Accepted: 10/04/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Host modulation therapy has emerged as a new concept for the treatment of periodontal disease. Recently, a lot of research is being done in product containing docosahexaenoic acid (DHA) and eicosapentanoic acid (EPA). Omega-3 PUFA have therapeutic, anti-inflammatory and protective properties. This systematic review analysed the adjunctive use of omega-3 fatty acids in periodontal therapy of periodontitis patients. METHODS PICO question (patient, intervention, comparison, and outcome) was formed. Keywords were generated and were fed in databases. The databases were Pubmed, Cochrane library and LIVIVO. Studies selected are Randomized clinical trial, clinical studies, and longitudinal studies. Meta -analysis were performed for Pocket depth (PD), Clinical attachment level (CAL), Gingival index (GI) and Plaque Index (PI). Risk of bias was also assessed. RESULTS On analysis of all the 8 studies at 3 months showed significant effect of omega -3 fatty acid on clinical attachment level (CAL), Pocket depth (PD). There was significant effect of omega-3 fatty acids in 4 studies at 6 months. CONCLUSION Within the limitation of the review, omega- 3 polyunsaturated fatty acids seems to have a positive effect on periodontal healing following periodontal therapy. Chronic periodontitis patient should be counselled to incorporate omega -3 fatty acid in their diet along with standard periodontal therapy.
Collapse
Affiliation(s)
| | | | - Dheeraj Kalra
- Department of Public Health Dentistry, YMT Dental College, Mumbai, India
| | | | | | | |
Collapse
|
19
|
Piotrowska M, Binienda A, Fichna J. The role of fatty acids in Crohn's disease pathophysiology - An overview. Mol Cell Endocrinol 2021; 538:111448. [PMID: 34480991 DOI: 10.1016/j.mce.2021.111448] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/30/2021] [Indexed: 12/14/2022]
Abstract
Crohn's disease (CD) is an inflammatory bowel disease (IBD) which is characterized by chronic and relapsing inflammation of the gastrointestinal (GI) tract. The etiology of CD is unknown, but factors such as epithelial barrier dysfunction, immune system imbalance, microbiota dysbiosis and environmental influences are thought to be involved in its pathogenesis. Recent studies have shown that short chain fatty acids (SCFAs) and long chain fatty acids (LCFAs) play a vital role in pathophysiology and development of CD by various mechanisms affecting pro- and anti-inflammatory mediators, and maintaining the intestinal homeostasis and regulation of gene expression. SCFAs and LCFAs activate signaling cascades that control immune functions through interaction with cell surface free fatty acid receptors (FFARs), i.e. FFAR1, FFAR2, FFAR3, and FFAR4. This review highlights the role of fatty acids in maintenance of intestinal and immune homeostasis and supports the supplementation of fatty acids as a promising adjunctive treatment for CD.
Collapse
Affiliation(s)
- Marta Piotrowska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, 92-215, Lodz, Poland.
| | - Agata Binienda
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, 92-215, Lodz, Poland.
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, 92-215, Lodz, Poland.
| |
Collapse
|
20
|
Taylor EN, Beckmann M, Villarreal-Ramos B, Vordermeier HM, Hewinson G, Rooke D, Mur LAJ, Koets AP. Metabolomic Changes in Naturally MAP-Infected Holstein-Friesian Heifers Indicate Immunologically Related Biochemical Reprogramming. Metabolites 2021; 11:metabo11110727. [PMID: 34822384 PMCID: PMC8625860 DOI: 10.3390/metabo11110727] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/04/2021] [Accepted: 10/21/2021] [Indexed: 11/28/2022] Open
Abstract
Johne’s disease, caused by Mycobacterium avium subsp. paratuberculosis (MAP), causes weight loss, diarrhoea, and reduced milk yields in clinically infected cattle. Asymptomatic, subclinically infected cattle shed MAP bacteria but are frequently not detected by diagnostic tests. Herein, we compare the metabolite profiles of sera from subclinically infected Holstein–Friesian heifers and antibody binding to selected MAP antigens. The study used biobanked serum samples from 10 naturally MAP-infected and 10 control heifers, sampled monthly from ~1 to 19 months of age. Sera were assessed using flow infusion electrospray–high-resolution mass spectrometry (FIE–HRMS) on a Q Exactive hybrid quadrupole–Orbitrap mass spectrometer for high-throughput, sensitive, non-targeted metabolite fingerprinting. Partial least-squares discriminant analyses (PLS-DA) and hierarchical cluster analysis (HCA) of the data discriminated between naturally MAP-infected and control heifers. In total, 33 metabolites that differentially accumulated in naturally MAP-infected heifers compared to controls were identified. Five were significantly elevated within MAP-infected heifers throughout the study, i.e., leukotriene B4, bicyclo prostaglandin E2 (bicyclo PGE2), itaconic acid, 2-hydroxyglutaric acid and N6-acetyl-L-lysine. These findings highlight the potential of metabolomics in the identification of novel MAP diagnostic markers and particular biochemical pathways, which may provide insights into the bovine immune response to MAP.
Collapse
Affiliation(s)
- Emma N. Taylor
- Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Ceredigion SY23 3DA, UK; (E.N.T.); (M.B.); (B.V.-R.); (H.-M.V.); (G.H.)
| | - Manfred Beckmann
- Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Ceredigion SY23 3DA, UK; (E.N.T.); (M.B.); (B.V.-R.); (H.-M.V.); (G.H.)
| | - Bernardo Villarreal-Ramos
- Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Ceredigion SY23 3DA, UK; (E.N.T.); (M.B.); (B.V.-R.); (H.-M.V.); (G.H.)
- Centre of Excellence for Bovine Tuberculosis, Aberystwyth University, Ceredigion SY23 3DA, UK
- Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK
| | - Hans-Martin Vordermeier
- Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Ceredigion SY23 3DA, UK; (E.N.T.); (M.B.); (B.V.-R.); (H.-M.V.); (G.H.)
- Centre of Excellence for Bovine Tuberculosis, Aberystwyth University, Ceredigion SY23 3DA, UK
- Animal and Plant Health Agency, Weybridge, Surrey KT15 3NB, UK
| | - Glyn Hewinson
- Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Ceredigion SY23 3DA, UK; (E.N.T.); (M.B.); (B.V.-R.); (H.-M.V.); (G.H.)
- Centre of Excellence for Bovine Tuberculosis, Aberystwyth University, Ceredigion SY23 3DA, UK
| | - David Rooke
- ProTEM Services Ltd., Horsham, West Sussex RH12 4BD, UK;
| | - Luis A. J. Mur
- Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Ceredigion SY23 3DA, UK; (E.N.T.); (M.B.); (B.V.-R.); (H.-M.V.); (G.H.)
- Correspondence: (L.A.J.M.); (A.P.K.)
| | - Ad P. Koets
- Wageningen Bioveterinary Research, 8221 RA Lelystad, The Netherlands
- Population Health Systems, Faculty of Veterinary Medicine, Utrecht University, 3584 CS Utrecht, The Netherlands
- Correspondence: (L.A.J.M.); (A.P.K.)
| |
Collapse
|
21
|
Marion-Letellier R, Leboutte M, Amamou A, Raman M, Savoye G, Ghosh S. Diet in Intestinal Fibrosis: A Double-Edged Sword. Nutrients 2021; 13:nu13093148. [PMID: 34579023 PMCID: PMC8470259 DOI: 10.3390/nu13093148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/07/2021] [Accepted: 09/07/2021] [Indexed: 11/30/2022] Open
Abstract
The natural history of inflammatory bowel diseases, especially Crohn’s disease, is frequently complicated by intestinal fibrosis. Because of the lack of effective treatments for intestinal fibrosis, there is an urgent need to develop new therapies. Factors promoting intestinal fibrosis are currently unclear, but diet is a potential culprit. Diet may influence predisposition to develop intestinal fibrosis or alter its natural history by modification of both the host immune response and intestinal microbial composition. Few studies have documented the effects of dietary factors in modulating IBD-induced intestinal fibrosis. As the mechanisms behind fibrogenesis in the gut are believed to be broadly similar to those from extra-intestinal organs, it may be relevant to investigate which dietary components can inhibit or promote fibrosis factors such as myofibroblasts progenitor activation in other fibrotic diseases.
Collapse
Affiliation(s)
- Rachel Marion-Letellier
- UNIROUEN, INSERM UMR 1073 Nutrition, Inflammation and Gut-Brain Axis, Normandie University, 76183 Rouen, France; (M.L.); (G.S.)
- Institute for Research and Innovation in Biomedicine (IRIB), UNIROUEN, 76183 Rouen, France
- Correspondence:
| | - Mathilde Leboutte
- UNIROUEN, INSERM UMR 1073 Nutrition, Inflammation and Gut-Brain Axis, Normandie University, 76183 Rouen, France; (M.L.); (G.S.)
- Institute for Research and Innovation in Biomedicine (IRIB), UNIROUEN, 76183 Rouen, France
| | - Asma Amamou
- APC Microbiome Ireland, Biosciences Building, University College Cork, Cork, Ireland; (A.A.); (S.G.)
| | - Maitreyi Raman
- Division of Gastroenterology, University of Calgary, Calgary, AB T2N 4N1, Canada;
- Department of Community Health Sciences, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Guillaume Savoye
- UNIROUEN, INSERM UMR 1073 Nutrition, Inflammation and Gut-Brain Axis, Normandie University, 76183 Rouen, France; (M.L.); (G.S.)
- Institute for Research and Innovation in Biomedicine (IRIB), UNIROUEN, 76183 Rouen, France
- Department of Gastroenterology, Rouen University Hospital, 76031 Rouen, France
| | - Subrata Ghosh
- APC Microbiome Ireland, Biosciences Building, University College Cork, Cork, Ireland; (A.A.); (S.G.)
| |
Collapse
|
22
|
Del Cornò M, Varì R, Scazzocchio B, Varano B, Masella R, Conti L. Dietary Fatty Acids at the Crossroad between Obesity and Colorectal Cancer: Fine Regulators of Adipose Tissue Homeostasis and Immune Response. Cells 2021; 10:cells10071738. [PMID: 34359908 PMCID: PMC8304920 DOI: 10.3390/cells10071738] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) is among the major threatening diseases worldwide, being the third most common cancer, and a leading cause of death, with a global incidence expected to increase in the coming years. Enhanced adiposity, particularly visceral fat, is a major risk factor for the development of several tumours, including CRC, and represents an important indicator of incidence, survival, prognosis, recurrence rates, and response to therapy. The obesity-associated low-grade chronic inflammation is thought to be a key determinant in CRC development, with the adipocytes and the adipose tissue (AT) playing a significant role in the integration of diet-related endocrine, metabolic, and inflammatory signals. Furthermore, AT infiltrating immune cells contribute to local and systemic inflammation by affecting immune and cancer cell functions through the release of soluble mediators. Among the factors introduced with diet and enriched in AT, fatty acids (FA) represent major players in inflammation and are able to deeply regulate AT homeostasis and immune cell function through gene expression regulation and by modulating the activity of several transcription factors (TF). This review summarizes human studies on the effects of dietary FA on AT homeostasis and immune cell functions, highlighting the molecular pathways and TF involved. The relevance of FA balance in linking diet, AT inflammation, and CRC is also discussed. Original and review articles were searched in PubMed without temporal limitation up to March 2021, by using fatty acid as a keyword in combination with diet, obesity, colorectal cancer, inflammation, adipose tissue, immune cells, and transcription factors.
Collapse
|
23
|
Bernabe-García M, Calder PC, Villegas-Silva R, Rodríguez-Cruz M, Chávez-Sánchez L, Cruz-Reynoso L, Mateos-Sánchez L, Lara-Flores G, Aguilera-Joaquín AR, Sánchez-García L. Efficacy of Docosahexaenoic Acid for the Prevention of Necrotizing Enterocolitis in Preterm Infants: A Randomized Clinical Trial. Nutrients 2021; 13:nu13020648. [PMID: 33671220 PMCID: PMC7922869 DOI: 10.3390/nu13020648] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 01/10/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is an inflammatory bowel disease and a leading cause of morbidity and mortality in preterm infants. In this study, a randomized double-blind parallel-group (1:1) trial was carried out in two neonatal intensive care units of two tertiary hospitals. Two hundred and twenty-five preterm newborns with an expected functional gastrointestinal tract were recruited and received an enteral dose of 75 mg of docosahexaenoic acid (DHA)/kg body weight or high-oleic sunflower oil daily for 14 days from the first enteral feed after birth. Confirmed NEC was evaluated with Bell’s scale from stage ≥ IIa. Two hundred and fourteen randomized infants were analyzed in terms of the intent-to-treat (DHA-group: n = 105; control-group: n = 109); data for two hundred infants were analysed per protocol. Confirmed NEC was lower in infants from the DHA-group compared with the control-group (0/100 vs. 7/100; p = 0.007), with RR = 0.93 (95% CI 0.881 to 0.981), risk difference = −7%, (95% CI −12.00 to −1.99), and number needed-to-treat = 15 (95% CI 8.3 to 50). Intent-to-treat analysis showed a lower level of treatment failure in the DHA-group compared with the control-group (6/105 (6%) vs. 16/109 (15%); p = 0.03, RR = 0.905, (95% CI 0.826 to 0.991)). The results after multivariate-regression analysis remained significant. Adverse events (apart from the incidence of NEC) were not different between groups. A daily dose of DHA for 14 days starting with the first enteral feed may prevent NEC in preterm infants.
Collapse
Affiliation(s)
- Mariela Bernabe-García
- Unidad de Investigación Médica en Nutrición, UMAE Hospital de Pediatría, CMN Siglo XXI, Instituto Mexicano del Seguro Social, México City 06720, Mexico; (M.B.-G.); (M.R.-C.)
| | - Philip C. Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton SO16 6YD, UK
- Correspondence:
| | - Raúl Villegas-Silva
- Neonatología, Hospital Infantil de México Federico Gómez, México City 06720, Mexico;
| | - Maricela Rodríguez-Cruz
- Unidad de Investigación Médica en Nutrición, UMAE Hospital de Pediatría, CMN Siglo XXI, Instituto Mexicano del Seguro Social, México City 06720, Mexico; (M.B.-G.); (M.R.-C.)
| | - Luis Chávez-Sánchez
- Unidad de Investigación Médica en Inmunología, UMAE Hospital de Pediatría, CMN Siglo XXI, Instituto Mexicano del Seguro Social, México City 06720, Mexico;
| | - Leonardo Cruz-Reynoso
- Unidad de Cuidados Intensivos Neonatales, UMAE Hospital de Gineco-Obstetricia No.3, CMN La Raza, Instituto Mexicano del Seguro Social, México City 02990, Mexico; (L.C.-R.); (A.R.A.-J.); (L.S.-G.)
| | - Leovigildo Mateos-Sánchez
- Unidad de Cuidados Intensivos Neonatales, UMAE Hospital de Gineco-Obstetricia N° 4 “Luis Castelazo Ayala”, Instituto Mexicano del Seguro Social, México City 01090, Mexico; (L.M.-S.); (G.L.-F.)
| | - Gabriel Lara-Flores
- Unidad de Cuidados Intensivos Neonatales, UMAE Hospital de Gineco-Obstetricia N° 4 “Luis Castelazo Ayala”, Instituto Mexicano del Seguro Social, México City 01090, Mexico; (L.M.-S.); (G.L.-F.)
| | - Augusto R. Aguilera-Joaquín
- Unidad de Cuidados Intensivos Neonatales, UMAE Hospital de Gineco-Obstetricia No.3, CMN La Raza, Instituto Mexicano del Seguro Social, México City 02990, Mexico; (L.C.-R.); (A.R.A.-J.); (L.S.-G.)
| | - Luisa Sánchez-García
- Unidad de Cuidados Intensivos Neonatales, UMAE Hospital de Gineco-Obstetricia No.3, CMN La Raza, Instituto Mexicano del Seguro Social, México City 02990, Mexico; (L.C.-R.); (A.R.A.-J.); (L.S.-G.)
| |
Collapse
|
24
|
Basson AR, Chen C, Sagl F, Trotter A, Bederman I, Gomez-Nguyen A, Sundrud MS, Ilic S, Cominelli F, Rodriguez-Palacios A. Regulation of Intestinal Inflammation by Dietary Fats. Front Immunol 2021; 11:604989. [PMID: 33603741 PMCID: PMC7884479 DOI: 10.3389/fimmu.2020.604989] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
With the epidemic of human obesity, dietary fats have increasingly become a focal point of biomedical research. Epidemiological studies indicate that high-fat diets (HFDs), especially those rich in long-chain saturated fatty acids (e.g., Western Diet, National Health Examination survey; NHANES 'What We Eat in America' report) have multi-organ pro-inflammatory effects. Experimental studies have confirmed some of these disease associations, and have begun to elaborate mechanisms of disease induction. However, many of the observed effects from epidemiological studies appear to be an over-simplification of the mechanistic complexity that depends on dynamic interactions between the host, the particular fatty acid, and the rather personalized genetics and variability of the gut microbiota. Of interest, experimental studies have shown that certain saturated fats (e.g., lauric and myristic fatty acid-rich coconut oil) could exert the opposite effect; that is, desirable anti-inflammatory and protective mechanisms promoting gut health by unanticipated pathways. Owing to the experimental advantages of laboratory animals for the study of mechanisms under well-controlled dietary settings, we focus this review on the current understanding of how dietary fatty acids impact intestinal biology. We center this discussion on studies from mice and rats, with validation in cell culture systems or human studies. We provide a scoping overview of the most studied diseases mechanisms associated with the induction or prevention of Inflammatory Bowel Disease in rodent models relevant to Crohn's Disease and Ulcerative Colitis after feeding either high-fat diet (HFD) or feed containing specific fatty acid or other target dietary molecule. Finally, we provide a general outlook on areas that have been largely or scarcely studied, and assess the effects of HFDs on acute and chronic forms of intestinal inflammation.
Collapse
Affiliation(s)
- Abigail R. Basson
- Division of Gastroenterology and Liver Diseases, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Cleveland Digestive Diseases Research Core, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Digestive Health Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Christy Chen
- Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Filip Sagl
- Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Ashley Trotter
- Division of Gastroenterology and Liver Diseases, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Hospital Medicine, Pritzker School of Medicine, NorthShore University Health System, Chicago, IL, United States
| | - Ilya Bederman
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Adrian Gomez-Nguyen
- Division of Gastroenterology and Liver Diseases, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Cleveland Digestive Diseases Research Core, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Mark S. Sundrud
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL, United States
| | - Sanja Ilic
- Department of Human Sciences, Human Nutrition, College of Education and Human Ecology, The Ohio State University, Columbus, OH, United States
| | - Fabio Cominelli
- Division of Gastroenterology and Liver Diseases, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Cleveland Digestive Diseases Research Core, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Digestive Health Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Alex Rodriguez-Palacios
- Division of Gastroenterology and Liver Diseases, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Cleveland Digestive Diseases Research Core, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Digestive Health Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
- University Hospitals Research and Education Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
25
|
Durkin LA, Childs CE, Calder PC. Omega-3 Polyunsaturated Fatty Acids and the Intestinal Epithelium-A Review. Foods 2021; 10:foods10010199. [PMID: 33478161 PMCID: PMC7835870 DOI: 10.3390/foods10010199] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Epithelial cells (enterocytes) form part of the intestinal barrier, the largest human interface between the internal and external environments, and responsible for maintaining regulated intestinal absorption and immunological control. Under inflammatory conditions, the intestinal barrier and its component enterocytes become inflamed, leading to changes in barrier histology, permeability, and chemical mediator production. Omega-3 (ω-3) polyunsaturated fatty acids (PUFAs) can influence the inflammatory state of a range of cell types, including endothelial cells, monocytes, and macrophages. This review aims to assess the current literature detailing the effects of ω-3 PUFAs on epithelial cells. Marine-derived ω-3 PUFAs, eicosapentaenoic acid and docosahexaenoic acid, as well as plant-derived alpha-linolenic acid, are incorporated into intestinal epithelial cell membranes, prevent changes to epithelial permeability, inhibit the production of pro-inflammatory cytokines and eicosanoids and induce the production of anti-inflammatory eicosanoids and docosanoids. Altered inflammatory markers have been attributed to changes in activity and/or expression of proteins involved in inflammatory signalling including nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), peroxisome proliferator activated receptor (PPAR) α and γ, G-protein coupled receptor (GPR) 120 and cyclooxygenase (COX)-2. Effective doses for each ω-3 PUFA are difficult to determine due to inconsistencies in dose and time of exposure between different in vitro models and between in vivo and in vitro models. Further research is needed to determine the anti-inflammatory potential of less-studied ω-3 PUFAs, including docosapentaenoic acid and stearidonic acid.
Collapse
Affiliation(s)
- Luke A. Durkin
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (C.E.C.); (P.C.C.)
- Correspondence:
| | - Caroline E. Childs
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (C.E.C.); (P.C.C.)
- Institute of Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Philip C. Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (C.E.C.); (P.C.C.)
- Institute of Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
26
|
Jaffal S, Oran S, Alsalem M. Anti-inflammatory and antipyretic potential of Arbutus andrachne L. methanolic leaf extract in rats. Asian Pac J Trop Biomed 2021. [DOI: 10.4103/2221-1691.328056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
27
|
Krishnamoorthy P, Raj AS, Roy S, Kumar NS, Kumar H. Comparative transcriptome analysis of SARS-CoV, MERS-CoV, and SARS-CoV-2 to identify potential pathways for drug repurposing. Comput Biol Med 2021; 128:104123. [PMID: 33260034 PMCID: PMC7683955 DOI: 10.1016/j.compbiomed.2020.104123] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022]
Abstract
The ongoing COVID-19 pandemic caused by the coronavirus, SARS-CoV-2, has already caused in excess of 1.25 million deaths worldwide, and the number is increasing. Knowledge of the host transcriptional response against this virus and how the pathways are activated or suppressed compared to other human coronaviruses (SARS-CoV, MERS-CoV) that caused outbreaks previously can help in the identification of potential drugs for the treatment of COVID-19. Hence, we used time point meta-analysis to investigate available SARS-CoV and MERS-CoV in-vitro transcriptome datasets in order to identify the significant genes and pathways that are dysregulated at each time point. The subsequent over-representation analysis (ORA) revealed that several pathways are significantly dysregulated at each time point after both SARS-CoV and MERS-CoV infection. We also performed gene set enrichment analyses of SARS-CoV and MERS-CoV with that of SARS-CoV-2 at the same time point and cell line, the results of which revealed that common pathways are activated and suppressed in all three coronaviruses. Furthermore, an analysis of an in-vivo transcriptomic dataset of COVID-19 patients showed that similar pathways are enriched to those identified in the earlier analyses. Based on these findings, a drug repurposing analysis was performed to identify potential drug candidates for combating COVID-19.
Collapse
Affiliation(s)
- Pandikannan Krishnamoorthy
- Department of Biological Sciences, Laboratory of Immunology and Infectious Disease Biology, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, 462066, MP, India
| | - Athira S Raj
- Department of Biological Sciences, Laboratory of Immunology and Infectious Disease Biology, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, 462066, MP, India
| | - Swagnik Roy
- Microbiology Department, Zoram Medical College, Falkawn, Mizoram, 796005, India
| | | | - Himanshu Kumar
- Department of Biological Sciences, Laboratory of Immunology and Infectious Disease Biology, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal, 462066, MP, India; Laboratory of Host Defense, WPI Immunology, Frontier Research Centre, Osaka University, Osaka, 5650871, Japan.
| |
Collapse
|
28
|
Rosa F, Matazel KS, Bowlin AK, Williams KD, Elolimy AA, Adams SH, Bode L, Yeruva L. Neonatal Diet Impacts the Large Intestine Luminal Metabolome at Weaning and Post-Weaning in Piglets Fed Formula or Human Milk. Front Immunol 2020; 11:607609. [PMID: 33365033 PMCID: PMC7750455 DOI: 10.3389/fimmu.2020.607609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
The impact of human milk (HM) or dairy milk-based formula (MF) on the large intestine’s metabolome was not investigated. Two-day old male piglets were randomly assigned to HM or MF diet (n = 26/group), from postnatal day (PND) 2 through 21 and weaned to a solid diet until PND 51. Piglets were euthanized at PND 21 and PND 51, luminal contents of the cecum, proximal (PC) and distal colons (DC), and rectum were collected and subjected to metabolomics analysis. Data analyses were performed using Metaboanalyst. In comparison to MF, the HM diet resulted in higher levels of fatty acids in the lumen of the cecum, PC, DC, and rectum at PND 21. Glutamic acid was greater in the lumen of cecum, PC, and DC relative to the MF group at PND 21. Also, spermidine was higher in the DC and rectal contents of HM relative to MF at PND 21. MF diet resulted in greater abundances of amino acids in the cecal lumen relative to HM diet at PND 21. Additionally, several sugar metabolites were higher in various regions of the distal gut of MF fed piglets relative to HM group at PND 21. In contrast, at PND 51, in various regions there were higher levels of erythritol, maltotriose, isomaltose in HM versus MF fed piglets. This suggests a post weaning shift in sugar metabolism that is impacted by neonatal diet. The data also suggest that infant diet type and host-microbiota interactions likely influence the lower gut metabolome.
Collapse
Affiliation(s)
- Fernanda Rosa
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Katelin S Matazel
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Arkansas Children's Research Institute, Little Rock, AR, United States
| | - Anne K Bowlin
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Keith D Williams
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock AR, United States
| | - Ahmed A Elolimy
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Sean H Adams
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Lars Bode
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA, United States.,Department of Pediatrics, University of California San Diego, La Jolla, CA, United States
| | - Laxmi Yeruva
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Arkansas Children's Research Institute, Little Rock, AR, United States
| |
Collapse
|
29
|
Marino N, German R, Rao X, Simpson E, Liu S, Wan J, Liu Y, Sandusky G, Jacobsen M, Stoval M, Cao S, Storniolo AMV. Upregulation of lipid metabolism genes in the breast prior to cancer diagnosis. NPJ Breast Cancer 2020; 6:50. [PMID: 33083529 PMCID: PMC7538898 DOI: 10.1038/s41523-020-00191-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
Histologically normal tissue adjacent to the tumor can provide insight of the microenvironmental alterations surrounding the cancerous lesion and affecting the progression of the disease. However, little is known about the molecular changes governing cancer initiation in cancer-free breast tissue. Here, we employed laser microdissection and whole-transcriptome profiling of the breast epithelium prior to and post tumor diagnosis to identify the earliest alterations in breast carcinogenesis. Furthermore, a comprehensive analysis of the three tissue compartments (microdissected epithelium, stroma, and adipose tissue) was performed on the breast donated by either healthy subjects or women prior to the clinical manifestation of cancer (labeled "susceptible normal tissue"). Although both susceptible and healthy breast tissues appeared histologically normal, the susceptible breast epithelium displayed a significant upregulation of genes involved in fatty acid uptake/transport (CD36 and AQP7), lipolysis (LIPE), and lipid peroxidation (AKR1C1). Upregulation of lipid metabolism- and fatty acid transport-related genes was observed also in the microdissected susceptible stromal and adipose tissue compartments, respectively, when compared with the matched healthy controls. Moreover, inter-compartmental co-expression analysis showed increased epithelium-adipose tissue crosstalk in the susceptible breasts as compared with healthy controls. Interestingly, reductions in natural killer (NK)-related gene signature and CD45+/CD20+ cell staining were also observed in the stromal compartment of susceptible breasts. Our study yields new insights into the cancer initiation process in the breast. The data suggest that in the early phase of cancer development, metabolic activation of the breast, together with increased epithelium-adipose tissue crosstalk may create a favorable environment for final cell transformation, proliferation, and survival.
Collapse
Affiliation(s)
- Natascia Marino
- Susan G. Komen Tissue Bank at the IU Simon Cancer Center, Indianapolis, IN 46202 USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Rana German
- Susan G. Komen Tissue Bank at the IU Simon Cancer Center, Indianapolis, IN 46202 USA
| | - Xi Rao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Ed Simpson
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - George Sandusky
- Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Max Jacobsen
- Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Miranda Stoval
- Susan G. Komen Tissue Bank at the IU Simon Cancer Center, Indianapolis, IN 46202 USA
| | - Sha Cao
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Anna Maria V Storniolo
- Susan G. Komen Tissue Bank at the IU Simon Cancer Center, Indianapolis, IN 46202 USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| |
Collapse
|
30
|
Augimeri G, Giordano C, Gelsomino L, Plastina P, Barone I, Catalano S, Andò S, Bonofiglio D. The Role of PPARγ Ligands in Breast Cancer: From Basic Research to Clinical Studies. Cancers (Basel) 2020; 12:cancers12092623. [PMID: 32937951 PMCID: PMC7564201 DOI: 10.3390/cancers12092623] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/11/2020] [Accepted: 09/12/2020] [Indexed: 02/06/2023] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ), belonging to the nuclear receptor superfamily, is a ligand-dependent transcription factor involved in a variety of pathophysiological conditions such as inflammation, metabolic disorders, cardiovascular disease, and cancers. In this latter context, PPARγ is expressed in many tumors including breast cancer, and its function upon binding of ligands has been linked to the tumor development, progression, and metastasis. Over the last decade, much research has focused on the potential of natural agonists for PPARγ including fatty acids and prostanoids that act as weak ligands compared to the strong and synthetic PPARγ agonists such as thiazolidinedione drugs. Both natural and synthetic compounds have been implicated in the negative regulation of breast cancer growth and progression. The aim of the present review is to summarize the role of PPARγ activation in breast cancer focusing on the underlying cellular and molecular mechanisms involved in the regulation of cell proliferation, cell cycle, and cell death, in the modulation of motility and invasion as well as in the cross-talk with other different signaling pathways. Besides, we also provide an overview of the in vivo breast cancer models and clinical studies. The therapeutic effects of natural and synthetic PPARγ ligands, as antineoplastic agents, represent a fascinating and clinically a potential translatable area of research with regards to the battle against cancer.
Collapse
Affiliation(s)
- Giuseppina Augimeri
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (C.G.); (L.G.); (P.P.); (I.B.); (S.C.); (S.A.)
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (C.G.); (L.G.); (P.P.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (C.G.); (L.G.); (P.P.); (I.B.); (S.C.); (S.A.)
| | - Pierluigi Plastina
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (C.G.); (L.G.); (P.P.); (I.B.); (S.C.); (S.A.)
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (C.G.); (L.G.); (P.P.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (C.G.); (L.G.); (P.P.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (C.G.); (L.G.); (P.P.); (I.B.); (S.C.); (S.A.)
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (C.G.); (L.G.); (P.P.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
- Correspondence: ; Tel.: +39-0984-496208
| |
Collapse
|
31
|
Radzikowska U, Rinaldi AO, Çelebi Sözener Z, Karaguzel D, Wojcik M, Cypryk K, Akdis M, Akdis CA, Sokolowska M. The Influence of Dietary Fatty Acids on Immune Responses. Nutrients 2019; 11:E2990. [PMID: 31817726 PMCID: PMC6950146 DOI: 10.3390/nu11122990] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/25/2019] [Accepted: 12/02/2019] [Indexed: 12/16/2022] Open
Abstract
Diet-derived fatty acids (FAs) are essential sources of energy and fundamental structural components of cells. They also play important roles in the modulation of immune responses in health and disease. Saturated and unsaturated FAs influence the effector and regulatory functions of innate and adaptive immune cells by changing membrane composition and fluidity and by acting through specific receptors. Impaired balance of saturated/unsaturated FAs, as well as n-6/n-3 polyunsaturated FAs has significant consequences on immune system homeostasis, contributing to the development of many allergic, autoimmune, and metabolic diseases. In this paper, we discuss up-to-date knowledge and the clinical relevance of the influence of dietary FAs on the biology, homeostasis, and functions of epithelial cells, macrophages, dendritic cells, neutrophils, innate lymphoid cells, T cells and B cells. Additionally, we review the effects of dietary FAs on the pathogenesis of many diseases, including asthma, allergic rhinitis, food allergy, atopic dermatitis, rheumatoid arthritis, multiple sclerosis as well as type 1 and 2 diabetes.
Collapse
Affiliation(s)
- Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos Wolfgang, Switzerland
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos Wolfgang, Switzerland
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Arturo O Rinaldi
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos Wolfgang, Switzerland
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos Wolfgang, Switzerland
| | - Zeynep Çelebi Sözener
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos Wolfgang, Switzerland
- Department of Chest Disease, Division of Allergy and Clinical Immunology, Ankara University School of Medicine, 06100 Ankara, Turkey
| | - Dilara Karaguzel
- Department of Biology, Faculty of Science, Hacettepe University, 06800 Ankara, Turkey
| | - Marzena Wojcik
- Department of Structural Biology, Medical University of Lodz, 90-752 Lodz, Poland
| | - Katarzyna Cypryk
- Department of Internal Medicine and Diabetology, Medical University of Lodz, 90-549 Lodz, Poland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos Wolfgang, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos Wolfgang, Switzerland
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos Wolfgang, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos Wolfgang, Switzerland
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos Wolfgang, Switzerland
| |
Collapse
|
32
|
Ma C, Vasu R, Zhang H. The Role of Long-Chain Fatty Acids in Inflammatory Bowel Disease. Mediators Inflamm 2019; 2019:8495913. [PMID: 31780872 PMCID: PMC6874876 DOI: 10.1155/2019/8495913] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/03/2019] [Indexed: 02/05/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a complicated disease involving multiple pathogenic factors. The complex relationships between long-chain fatty acids (LCFAs) and the morbidity of IBD drive numerous studies to unravel the underlying mechanisms. A better understanding of the role of LCFAs in IBD will substitute or boost the current IBD therapies, thereby obtaining mucosal healing. In this review, we focused on the roles of LCFAs on the important links of inflammatory regulation in IBD, including in the pathogen recognition phase and in the inflammatory resolving phase, and the effects of LCFAs on immune cells in IBD.
Collapse
Affiliation(s)
- Chunxiang Ma
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Reshma Vasu
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Hu Zhang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
33
|
Morshedzadeh N, Shahrokh S, Aghdaei HA, Amin Pourhoseingholi M, Chaleshi V, Hekmatdoost A, Karimi S, Zali MR, Mirmiran P. Effects of flaxseed and flaxseed oil supplement on serum levels of inflammatory markers, metabolic parameters and severity of disease in patients with ulcerative colitis. Complement Ther Med 2019; 46:36-43. [PMID: 31519285 DOI: 10.1016/j.ctim.2019.07.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/15/2019] [Accepted: 07/18/2019] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE The present study aimed to evaluate the possible effect of grounded flaxseed and flaxseed oil on serum levels of inflammatory markers, metabolic parameters, and the severity of disease in patients with UC. METHODS In this open-labeled randomized controlled trial, 90 UC patients were randomly assigned to one of the 3 groups for 12 weeks: grounded flaxseed (GF; 30 g/day), flaxseed oil (FO; 10 g/day) and control group. The weight, waist circumference, systolic and diastolic blood pressure, serum inflammatory markers (interleukin-6 (IL-6), interferon gamma (INF-γ), transforming growth factor beta (TGF-β), and Erythrocyte Sedimentation Rate (ESR)), and fecal calprotectin were measured at the baseline and end of the study. RESULTS Totally, 75 patients (43 men and 32 women) with a mean age of 31.54 ± 9.84 years participated in the present study. Comparing the change of the variables indicated a significant decrease in fecal calprotectin (P < 0.001), Mayo score (P < 0.001), ESR (P < 0.001), INF-γ (P < 0.001), IL-6 (P < 0.001), waist circumference (P = 0.02), Diastolic Blood Pressure (DBP) (P < 0.001), and Systolic Blood Pressure (SBP) (P < 0.001) and a significant increase in TGF-β (P < 0.001) and Inflammatory Bowel Disease Questionnaire-Short form (IBDQ-9) score (P < 0.001) in the GF and FO groups compared to the control. No difference was obvious between the FO and GF groups except for TGF-β. CONCLUSION The present study showed that both flaxseed and flaxseed oil, attenuate inflammatory markers, disease severity, blood pressure, and WC. However, the effect of flaxseed on weight and BMI was not evident.
Collapse
Affiliation(s)
- Nava Morshedzadeh
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shabnam Shahrokh
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohamad Amin Pourhoseingholi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Chaleshi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azita Hekmatdoost
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soheila Karimi
- Nutrition and Endocrine Research Center, Research Institute For Endocrine Sciences, Shahid Beheshti University Of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Parvin Mirmiran
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Nutrition and Endocrine Research Center, Research Institute For Endocrine Sciences, Shahid Beheshti University Of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Seiri P, Abi A, Soukhtanloo M. PPAR-γ: Its ligand and its regulation by microRNAs. J Cell Biochem 2019; 120:10893-10908. [PMID: 30770587 DOI: 10.1002/jcb.28419] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 12/04/2018] [Indexed: 01/24/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear receptor superfamily. PPARs are categorized into three subtypes, PPARα, β/δ, and γ, encoded by different genes, expressed in diverse tissues and participate in various biological functions and can be activated by their metabolic derivatives in the body or dietary fatty acids. The PPAR-γ also takes parts in the regulation of energy balance, lipoprotein metabolism, insulin sensitivity, oxidative stress, and inflammatory signaling. It has been implicated in the pathology of numerous diseases including obesity, diabetes, atherosclerosis, and cancers. Among various cellular and molecular targets that are able to regulate PPAR-γ and its underlying pathways, microRNAs (miRNAs) appeared as important regulators. Given that the deregulation of these molecules via targeting PPAR-γ could affect initiation and progression of various diseases, identification of miRNAs that affects PPAR-γ could contribute to the better understanding of roles of PPAR-γ in various biological and pathological conditions. Here, we have summarized the function and various ligands of PPAR-γ and have highlighted various miRNAs involved in the regulation of PPAR-γ.
Collapse
Affiliation(s)
- Parvaneh Seiri
- Department of Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Abi
- Department of Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Soukhtanloo
- Department of Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
35
|
Xu F, Zhu L, Qian C, Zhou J, Geng D, Li P, Xuan W, Wu F, Zhao K, Kong W, Qin Y, Liang L, Liu L, Liu X. Impairment of Intestinal Monocarboxylate Transporter 6 Function and Expression in Diabetic Rats Induced by Combination of High-Fat Diet and Low Dose of Streptozocin: Involvement of Butyrate-Peroxisome Proliferator-Activated Receptor- γ Activation. Drug Metab Dispos 2019; 47:556-566. [PMID: 30923035 DOI: 10.1124/dmd.118.085803] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 03/26/2019] [Indexed: 12/13/2022] Open
Abstract
Generally, diabetes remarkably alters the expression and function of intestinal drug transporters. Nateglinide and bumetanide are substrates of monocarboxylate transporter 6 (MCT6). We investigated whether diabetes down-regulated the function and expression of intestinal MCT6 and the possible mechanism in diabetic rats induced by a combination of high-fat diet and low-dose streptozocin. Our results indicated that diabetes significantly decreased the oral plasma exposure of nateglinide. The plasma peak concentration and area under curve in diabetic rats were 16.9% and 28.2% of control rats, respectively. Diabetes significantly decreased the protein and mRNA expressions of intestinal MCT6 and oligopeptide transporter 1 (PEPT1) but up-regulated peroxisome proliferator-activated receptor γ (PPARγ) protein level. Single-pass intestinal perfusion demonstrated that diabetes prominently decreased the absorption of nateglinide and bumetanide. The MCT6 inhibitor bumetanide, but not PEPT1 inhibitor glycylsarcosine, significantly inhibited intestinal absorption of nateglinide in rats. Coadministration with bumetanide remarkably decreased the oral plasma exposure of nateglinide in rats. High concentrations of butyrate were detected in the intestine of diabetic rats. In Caco-2 cells (a human colorectal adenocarcinoma cell line), bumetanide and MCT6 knockdown remarkably inhibited the uptake of nateglinide. Butyrate down-regulated the function and expression of MCT6 in a concentration-dependent manner but increased PPARγ expression. The decreased expressions of MCT6 by PPARγ agonist troglitazone or butyrate were reversed by both PPARγ knockdown and PPARγ antagonist 2-chloro-5-nitro-N-phenylbenzamide (GW9662). Four weeks of butyrate treatment significantly decreased the oral plasma concentrations of nateglinide in rats, accompanied by significantly higher intestinal PPARγ and lower MCT6 protein levels. In conclusion, diabetes impaired the expression and function of intestinal MCT6 partly via butyrate-mediated PPARγ activation, decreasing the oral plasma exposure of nateglinide.
Collapse
Affiliation(s)
- Feng Xu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Liang Zhu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Chaoqun Qian
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Junjie Zhou
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Donghao Geng
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Ping Li
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Wenjing Xuan
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Fangge Wu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Kaijing Zhao
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Weimin Kong
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Yuanyuan Qin
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Limin Liang
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Li Liu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xiaodong Liu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
36
|
Uddin MS, Kabir MT, Jakaria M, Mamun AA, Niaz K, Amran MS, Barreto GE, Ashraf GM. Endothelial PPARγ Is Crucial for Averting Age-Related Vascular Dysfunction by Stalling Oxidative Stress and ROCK. Neurotox Res 2019; 36:583-601. [PMID: 31055770 DOI: 10.1007/s12640-019-00047-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/01/2019] [Accepted: 04/11/2019] [Indexed: 02/07/2023]
Abstract
Aging plays a significant role in the progression of vascular diseases and vascular dysfunction. Activation of the ADP-ribosylation factor 6 and small GTPases by inflammatory signals may cause vascular permeability and endothelial leakage. Pro-inflammatory molecules have a significant effect on smooth muscle cells (SMC). The migration and proliferation of SMC can be promoted by tumor necrosis factor alpha (TNF-α). TNF-α can also increase oxidative stress in SMCs, which has been identified to persuade DNA damage resulting in apoptosis and cellular senescence. Peroxisome proliferator-activated receptor (PPAR) acts as a ligand-dependent transcription factor and a member of the nuclear receptor superfamily. They play key roles in a wide range of biological processes, including cell differentiation and proliferation, bone formation, cell metabolism, tissue remodeling, insulin sensitivity, and eicosanoid signaling. The PPARγ activation regulates inflammatory responses, which can exert protective effects in the vasculature. In addition, loss of function of PPARγ enhances cardiovascular events and atherosclerosis in the vascular endothelium. This appraisal, therefore, discusses the critical linkage of PPARγ in the inflammatory process and highlights a crucial defensive role for endothelial PPARγ in vascular dysfunction and disease, as well as therapy for vascular aging.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.
| | | | - Md Jakaria
- Department of Applied Life Sciences, Graduate School, Konkuk University, Chungju, South Korea
| | | | - Kamal Niaz
- Department of Pharmacology and Toxicology, Faculty of Bio-Sciences, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Md Shah Amran
- Department of Pharmaceutical Chemistry, University of Dhaka, Dhaka, Bangladesh
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia. .,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
37
|
Arizmendi N, Hou C, Guo F, Li Y, Kulka M. Bicyclic eremophilane-type petasite sesquiterpenes potentiate peroxisome proliferator-activated receptor γ activator-mediated inhibition of dendritic cells. Int J Immunopathol Pharmacol 2018; 32:2058738418787739. [PMID: 30014756 PMCID: PMC6050815 DOI: 10.1177/2058738418787739] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Dendritic cell (DC) activation induces expression of co-stimulatory surface
molecules, as well as migration into secondary lymphoid organs, where they
activate naïve T-cells. A family of plant derivatives, eremophilane-type
petasite sesquiterpenes, can regulate the immune system through DC targeting due
to their anti-inflammatory effects. Peroxisome proliferator–activated receptor
gamma (PPARγ) is involved in inhibition of inflammatory responses and induction
of DCs to acquire a mucosal phenotype. Since mucosal DCs are central in innate
immune responses, we hypothesized that eremophilane-type petasite sesquiterpenes
exerted their anti-inflammatory effects by inhibiting DC maturation and
activation through PPARγ. This study assessed the bicyclic eremophilane-type
petasite sesquiterpene compounds Fukinone and
10βH-8α,12-Epidioxyeremophil-7(11)-en-8β-ol (ZYFDC21 and ZYFDC22) in the
maturation and activation of mouse DC. We measured surface expression of
co-stimulatory molecules by flow cytometry and cell-free supernatant cytokine
production upon lipopolysaccharide stimulation by enzyme-linked immunosorbent
assays (ELISAs) in the presence or absence of PPARγ agonists. DCs were generated
from C57BL/6 mice bone marrow cells and harvested. Cells were exposed to
bicyclic eremophilane-type petasite sesquiterpenes ZYFDC21 or ZYFDC22 in the
presence or absence of synthetic PPARγ agonists (GW1929 and TGZ) or the natural
PPARγ ligand 15d-PGJ2, followed by overnight activation with LPS. We
observed differences in the upregulation of surface expression of CD86, along
with TNF, IL-6, and IL-12p70 released by DCs stimulated with LPS, when using
combinations of bicyclic eremophilane-type petasite sesquiterpenes ZYFDC21 or
ZYFDC22, and PPARγ agonists, in particular the PPARγ ligand 15d-PGJ2.
Our results indicate that bicyclic eremophilane-type petasite sesquiterpenes
ZYFDC21 or ZYFDC22 inhibit maturation and activation of DC, and this activity is
augmented upon PPARγ activation.
Collapse
Affiliation(s)
- Narcy Arizmendi
- 1 Nanotechnology Research Center, National Research Council Canada, Edmonton, AB, Canada
| | - Chenjie Hou
- 2 School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fujiang Guo
- 2 School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Li
- 2 School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Marianna Kulka
- 1 Nanotechnology Research Center, National Research Council Canada, Edmonton, AB, Canada.,3 Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
38
|
Yan Y, Wang Z, Wang D, Lawrence P, Wang X, Kothapalli KSD, Greenwald J, Liu R, Park HG, Brenna JT. BCFA-enriched vernix-monoacylglycerol reduces LPS-induced inflammatory markers in human enterocytes in vitro. Pediatr Res 2018; 83:874-879. [PMID: 29166379 DOI: 10.1038/pr.2017.297] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/08/2017] [Indexed: 01/28/2023]
Abstract
BackgroundExcess vernix caseosa produced by the fetal skin appears as particles suspended in the amniotic fluid in late gestation, is swallowed by the fetus, and is found throughout the newborn gastrointestinal tract as the first organisms are arriving to colonize the gut. Lipid-rich vernix contains an unusually high 29% branched chain fatty acids (BCFA). BCFAs reduce the incidence of necrotizing enterocolitis in an animal model, and were recently found predominantly in the sn-2 position of human milk triacylglycerols. Nothing is known about the influence of vernix BCFA on proinflammatory markers in human enterocytes.MethodsWe investigated the effect of vernix-monoacylglycerides (MAGs) (enriched with 30% BCFA) on interleukin (IL)-8 and NF-κB production in a human intestinal epithelial cell line (Caco-2). Caco-2 cells were pretreated with vernix-MAG or vernix-free fatty acid (FFA) prior to lipopolysaccharide (LPS) activation.ResultsBoth vernix-MAG and vernix-FFA increased cell BCFA and eliminated an LPS-induced 20% reduction in cell viability. In stimulated Caco-2 cells, vernix-MAG was more effective than vernix-FFA in suppressing IL-8 and NF-κB. Activated vernix-MAG-treated cells expressed less of the cell-surface Toll-like receptor4 (TLR-4) compared with controls.ConclusionThis is the first study to show the reduction of proinflammatory markers in human cells mediated by BCFA-MAG.
Collapse
Affiliation(s)
- Yuanyuan Yan
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, National Engineering Research Center for Functional Food, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Zhen Wang
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Donghao Wang
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Peter Lawrence
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Xingguo Wang
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, National Engineering Research Center for Functional Food, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Kumar S D Kothapalli
- Department of Pediatrics, Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin, Austin, Texas
| | - Jacelyn Greenwald
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
| | - Ruijie Liu
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, National Engineering Research Center for Functional Food, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hui Gyu Park
- Department of Pediatrics, Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin, Austin, Texas
| | - J Thomas Brenna
- Department of Pediatrics, Dell Pediatric Research Institute, Dell Medical School, University of Texas at Austin, Austin, Texas
| |
Collapse
|
39
|
Baynes HW, Mideksa S, Ambachew S. The role of polyunsaturated fatty acids (n-3 PUFAs) on the pancreatic β-cells and insulin action. Adipocyte 2018. [PMID: 29537934 DOI: 10.1080/21623945.2018.1443662] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Polyunsaturated Fatty acids have multiple effects in peripheral tissues and pancreatic beta cell function. The n-3 Polyunsaturated Fatty acids prevent and reverse high-fat-diet induced adipose tissue inflammation and insulin resistance. Insulin secretion is stimulated by glucose, amino acids, and glucagon- like peptide-1 in tissue containing high levels of n-3 Polyunsaturated Fatty acids than lower level of n-3 Polyunsaturated Fatty acids. Also, n-3 Polyunsaturated Fatty acids led to decreased production of prostaglandin, which in turn contributed to the elevation of insulin secretion. N-3 polyunsaturated fatty acids prevent cytokine-induced cell death in pancreatic islets. Supplementation of n-3 Polyunsaturated Fatty acids for human subjects prevent beta cell destruction and insulin resistance. It also enhances insulin secretion, reduction in lipid profiles and glucose concentration particularly in type II diabetes patients. Therefore there should be a focus on the treatment mechanism of insulin related obesity and diabetes by n-3 polyunsaturated fatty acids.
Collapse
Affiliation(s)
- Habtamu Wondifraw Baynes
- Department of Clinical Chemistry, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Northwest Ethiopia
| | - Seifu Mideksa
- Clinical Chemistry laboratory, Ayder Comprehensive Specialized Hospital, Mekelle, Northern, Ethiopia
| | - Sintayehu Ambachew
- Department of Clinical Chemistry, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Northwest Ethiopia
| |
Collapse
|
40
|
Dietary n-3 PUFA May Attenuate Experimental Colitis. Mediators Inflamm 2018; 2018:8430614. [PMID: 29670469 PMCID: PMC5833476 DOI: 10.1155/2018/8430614] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/13/2017] [Accepted: 10/31/2017] [Indexed: 02/06/2023] Open
Abstract
Background Inflammatory bowel diseases (IBD) occurred in genetically predisposed people exposed to environmental triggers. Diet has long been suspected to contribute to the development of IBD. Supplementation with n-3 polyunsaturated fatty acids (PUFA) protects against intestinal inflammation in rodent models while clinical trials showed no benefits. We hypothesized that intervention timing is crucial and dietary fatty acid pattern may influence intestinal environment to modify inflammation genesis. The aim of this study was to evaluate the dietary effect of PUFA composition on intestinal inflammation. Methods Animals received diet varying in their PUFA composition for four weeks before TNBS-induced colitis. Colon inflammatory markers and gut barrier function parameters were assessed. Inflammatory pathway PCR arrays were determined. Results n-3 diet significantly decreased colon iNOS, COX-2 expression, IL-6 production, and LTB4 production but tended to decrease colon TNFα production (P = 0.0617) compared to control diet. Tight junction protein (claudin-1, occludin) expressions and MUC2 and TFF3 mRNA levels were not different among groups. n-9 diet also decreased colon IL-6 production (P < 0.05). Conclusions Dietary n-3 PUFA influence colitis development by attenuating inflammatory markers. Further research is required to better define dietary advice with a scientific rationale.
Collapse
|
41
|
Lombardo L, Grasso F, Lanciano F, Loria S, Monetti E. Broad-Spectrum Health Protection of Extra Virgin Olive Oil Compounds. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/b978-0-444-64057-4.00002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
|
42
|
Byndloss MX, Olsan EE, Rivera-Chávez F, Tiffany CR, Cevallos SA, Lokken KL, Torres TP, Byndloss AJ, Faber F, Gao Y, Litvak Y, Lopez CA, Xu G, Napoli E, Giulivi C, Tsolis RM, Revzin A, Lebrilla CB, Bäumler AJ. Microbiota-activated PPAR-γ signaling inhibits dysbiotic Enterobacteriaceae expansion. Science 2017; 357:570-575. [PMID: 28798125 DOI: 10.1126/science.aam9949] [Citation(s) in RCA: 787] [Impact Index Per Article: 98.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 06/22/2017] [Indexed: 12/12/2022]
Abstract
Perturbation of the gut-associated microbial community may underlie many human illnesses, but the mechanisms that maintain homeostasis are poorly understood. We found that the depletion of butyrate-producing microbes by antibiotic treatment reduced epithelial signaling through the intracellular butyrate sensor peroxisome proliferator-activated receptor γ (PPAR-γ). Nitrate levels increased in the colonic lumen because epithelial expression of Nos2, the gene encoding inducible nitric oxide synthase, was elevated in the absence of PPAR-γ signaling. Microbiota-induced PPAR-γ signaling also limits the luminal bioavailability of oxygen by driving the energy metabolism of colonic epithelial cells (colonocytes) toward β-oxidation. Therefore, microbiota-activated PPAR-γ signaling is a homeostatic pathway that prevents a dysbiotic expansion of potentially pathogenic Escherichia and Salmonella by reducing the bioavailability of respiratory electron acceptors to Enterobacteriaceae in the lumen of the colon.
Collapse
Affiliation(s)
- Mariana X Byndloss
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Erin E Olsan
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Fabian Rivera-Chávez
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Connor R Tiffany
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Stephanie A Cevallos
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Kristen L Lokken
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Teresa P Torres
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Austin J Byndloss
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Franziska Faber
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Yandong Gao
- Department of Biomedical Engineering, College of Engineering, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Yael Litvak
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Christopher A Lopez
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Gege Xu
- Department of Chemistry, College of Letters and Sciences, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Eleonora Napoli
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Renée M Tsolis
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Alexander Revzin
- Department of Biomedical Engineering, College of Engineering, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Carlito B Lebrilla
- Department of Chemistry, College of Letters and Sciences, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Avenue, Davis, CA 95616, USA.
| |
Collapse
|
43
|
Abstract
There are many mechanisms to explain how food may drive and ameliorate inflammation. Although there are no consistent macronutrient associations inflammatory bowel disease (IBD) development, many exclusion diets have been described: IgG-4 guided exclusion diet; semivegetarian diet; low-fat, fiber-limited exclusion diet; Paleolithic diet; Maker's diet; vegan diet; Life without Bread diet; exclusive enteral nutrition (EEN), the Specific Carbohydrate Diet (SCD) and the low FODMAP diet. The literature on diet and IBD is reviewed with a particular focus on EEN, SCD, and low FODMAP diets. Lessons learned from the existing observations and strengths and shortcomings of existing data are presented.
Collapse
|
44
|
Omega-3 and Omega-6 Fatty Acids Act as Inhibitors of the Matrix Metalloproteinase-2 and Matrix Metalloproteinase-9 Activity. Protein J 2017. [PMID: 28646265 DOI: 10.1007/s10930-017-9727-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Polyunsaturated fatty acids have been reported to play a protective role in a wide range of diseases characterized by an increased metalloproteinases (MMPs) activity. The recent finding that omega-3 and omega-6 fatty acids exert an anti-inflammatory effect in periodontal diseases has stimulated the present study, designed to determine whether such properties derive from a direct inhibitory action of these compounds on the activity of MMPs. To this issue, we investigated the effect exerted by omega-3 and omega-6 fatty acids on the activity of MMP-2 and MMP-9, two enzymes that actively participate to the destruction of the organic matrix of dentin following demineralization operated by bacteria acids. Data obtained (both in vitro and on ex-vivo teeth) reveal that omega-3 and omega-6 fatty acids inhibit the proteolytic activity of MMP-2 and MMP-9, two enzymes present in dentin. This observation is of interest since it assigns to these compounds a key role as MMPs inhibitors, and stimulates further study to better define their therapeutic potentialities in carious decay.
Collapse
|
45
|
He X, Liu W, Shi M, Yang Z, Zhang X, Gong P. Docosahexaenoic acid attenuates LPS-stimulated inflammatory response by regulating the PPARγ/NF-κB pathways in primary bovine mammary epithelial cells. Res Vet Sci 2017; 112:7-12. [PMID: 28095338 DOI: 10.1016/j.rvsc.2016.12.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 12/15/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND Docosahexaenoic acid (DHA) is a major dietary n-3 polyunsaturated fatty acid (n-3 PUFA) in fish oil, and has been reported to possess a number of biological properties, such as anti-inflammatory, antitumor and immune-regulatory properties. However, whether DHA exert anti-inflammatory effect on lipopolysaccharide (LPS)-induced mastitis remains unclear. In this study, we investigate the effect and underlying mechanisms of the effects of DHA on LPS-stimulated primary bovine mammary epithelial cells (bMEC). METHODS The experiment was divided into six groups as followed: control group, GW9662+LPS+DHA (100μM) group, LPS and LPS+DHA (25, 50 and 100μM) groups. bMEC were treated with DHA for 3h before LPS (200μg/ml) stimulation, and incubated with the PPARγ inhibitor GW9662 for 12h before DHA treatment. The mRNA levels of TNF-α, IL-6 and IL-1β were measured by quantitative real-time PCR (qRT-PCR). Western blot was employed for measuring the transcriptional activity of NF-κB and PPARγ. RESULTS Our results showed that DHA pretreatment significantly decreased the mRNA expression of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and interleukin-1β (IL-1β) in bMEC stimulated with LPS. Besides, DHA suppressed the phosphorylation of nuclear transcription factor-kappaB (NF-κB) p65 and degradation inhibitor of NF-κBα (IκBα) in NF-κB signal pathway, and activated proliferator activated receptor gamma (PPARγ). But, all those effects were obviously abolished by addition of GW9662, a specific inhibitor of PPARγ. CONCLUSION In conclusion, these results indicated that DHA may attenuate LPS-stimulated inflammatory response in bMEC by suppressing NF-κB activation through a mechanism partly dependent on PPARγ activation.
Collapse
Affiliation(s)
- Xuexiu He
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Weijian Liu
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Mingyu Shi
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Zhengtao Yang
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Xichen Zhang
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Pengtao Gong
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China.
| |
Collapse
|
46
|
Yan Y, Wang Z, Greenwald J, Kothapalli KSD, Park HG, Liu R, Mendralla E, Lawrence P, Wang X, Brenna JT. BCFA suppresses LPS induced IL-8 mRNA expression in human intestinal epithelial cells. Prostaglandins Leukot Essent Fatty Acids 2017; 116:27-31. [PMID: 28088291 DOI: 10.1016/j.plefa.2016.12.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 11/29/2016] [Accepted: 12/01/2016] [Indexed: 12/28/2022]
Abstract
Branched chain fatty acids (BCFA) are components of common food fats and are major constituents of the normal term human newborn GI tract. Polyunsaturated fatty acids (PUFA) have been suggested to reduce the risk and development of inflammatory bowel diseases (IBD); however, little is known about the influence of BCFA on inflammation. We investigated the effect of BCFA on interleukin (IL)-8 and NF-κB production in a human intestinal epithelial cell line (Caco-2). Cells were pre-treated with specific BCFA, or DHA, or EPA, and then activated with lipopolysaccharide (LPS). Both anteiso- and iso- BCFA reduce IL-8. Anteiso-BCFA more effectively suppressed IL-8 than iso-BCFA in LPS stimulated Caco-2 cells. However BCFA in general were less effective than DHA or EPA. Activated BCFA-treated cells expressed less of the cell surface Toll-like receptor 4 (TLR-4) compared to controls. These are the first data to show the reduction of pro-inflammatory markers in human cells mediated by BCFA.
Collapse
Affiliation(s)
- Y Yan
- State Key Laboratory of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Z Wang
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - J Greenwald
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - K S D Kothapalli
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - H G Park
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - R Liu
- State Key Laboratory of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - E Mendralla
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - P Lawrence
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - X Wang
- State Key Laboratory of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| | - J T Brenna
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
47
|
Schübel R, Jaudszus A, Krüger R, Roth A, Klempt M, Barth SW. Dietary essential α-linolenic acid and linoleic acid differentially modulate TNFα-induced NFκB activity in FADS2-deficient HEK-293 cells. Int J Food Sci Nutr 2016; 68:553-559. [DOI: 10.1080/09637486.2016.1265918] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Ruth Schübel
- Department of Physiology and Biochemistry of Nutrition, Karlsruhe, Germany
| | - Anke Jaudszus
- Department of Physiology and Biochemistry of Nutrition, Karlsruhe, Germany
| | - Ralf Krüger
- Department of Physiology and Biochemistry of Nutrition, Karlsruhe, Germany
| | - Alexander Roth
- Department of Physiology and Biochemistry of Nutrition, Karlsruhe, Germany
| | - Martin Klempt
- Department of Quality and Safety of Milk and Fish, Federal Research Institute of Nutrition and Food, Max Rubner-Institut, Kiel, Germany
| | | |
Collapse
|
48
|
Marion-Letellier R, Savoye G, Ghosh S. IBD: In Food We Trust. J Crohns Colitis 2016; 10:1351-1361. [PMID: 27194533 DOI: 10.1093/ecco-jcc/jjw106] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/10/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Both science and patients associate diet with inflammatory bowel disease [IBD]. There is no doubt that links between IBD and diet are numerous, based on both epidemiological studies and experimental studies. However, scientific evidence to support dietary advice is currently lacking, and dietary counselling for IBD patients is often limited in clinical practice to the improvement of nutrient intake. This review aimed to focus on both patient's beliefs about and molecular mechanisms for crosstalk between nutrients and inflammation. METHODS A literature search using PubMed was performed to identify relevant studies on diet and/or nutrients and their role in IBD. Pubmed [from inception to January 20, 2016] was searched using the terms: 'Crohn', 'colitis',' intestinal epithelial cells', and a list of terms relating to diet or numerous specific nutrients. Terms associated with nutrients were individually tested in the context of IBD. Reference lists from studies selected were manually searched to identify further relevant reports. Manuscripts about diet in the context of IBD from basic science, epidemiological studies, or clinical trials were selected and reviewed. Only articles published in English were included. RESULTS Epidemiological studies highlight the key role of diet in IBD development, and many IBD patients report diet as a triggering factor in relapse of disease. In addition, we present research on the impact of nutrients on innate immunity. CONCLUSION Diet may offer an alternative approach to restoring deficient innate immunity in IBD, and this may be the scientific rationale for providing dietary counselling for IBD patients.
Collapse
Affiliation(s)
| | - Guillaume Savoye
- INSERM Unit UMR1073, Rouen University and Rouen University Hospital, Rouen cedex, France.,Department of Gastroenterology, Rouen University Hospital, Rouen cedex, France
| | - Subrata Ghosh
- Division of Gastroenterology, University of Calgary, Alberta, Canada
| |
Collapse
|
49
|
Hintze KJ, Tawzer J, Ward RE. Concentration and ratio of essential fatty acids influences the inflammatory response in lipopolysaccharide challenged mice. Prostaglandins Leukot Essent Fatty Acids 2016; 111:37-44. [PMID: 27021356 DOI: 10.1016/j.plefa.2016.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 03/01/2016] [Indexed: 12/31/2022]
Abstract
The goal of this study was to evaluate the role of both the % of dietary, 18-carbon PUFA (2.5%, 5% and 10%) and the n-6:n-3 ratio (1:1, 10:1 and 20:1) on the acute inflammatory response. Mice were fed diets for 8 weeks and injected intraperitoneally with LPS to induce acute inflammation. After 24h mice were sacrificed and plasma cytokines measured. Diets significantly affected the erythrocyte PUFA composition and the effect of PUFA ratio was more prominent than of PUFA concentration. The % dietary PUFA affected feed efficiency (p<0.05) and there was a PUFA×ratio interaction with body fat (p<0.01). In mice fed high %kcal from PUFA, those given a low n-6:n-3 ratio had more body fat than those fed a high ratio. Of the twelve cytokines measured, eleven were significantly affected by the % PUFA (p<0.05), whereas five were affected by the ratio (p<0.05). For seven cytokines, there was a significant PUFA×ratio interaction according to a two way ANOVA (p<0.05). These data indicate that dietary polyunsaturated fatty acids can affect LPS induced-inflammation.
Collapse
Affiliation(s)
- K J Hintze
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, UMC 8700, Logan, UT 84322, USA; Applied Nutrition Research, Utah Science Technology and Research Initiative (USTAR), Logan, UT 84322, USA
| | - J Tawzer
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, UMC 8700, Logan, UT 84322, USA
| | - R E Ward
- Department of Nutrition, Dietetics and Food Sciences, Utah State University, UMC 8700, Logan, UT 84322, USA; Applied Nutrition Research, Utah Science Technology and Research Initiative (USTAR), Logan, UT 84322, USA.
| |
Collapse
|
50
|
Hashimoto M, Hossain S, Al Mamun A, Matsuzaki K, Arai H. Docosahexaenoic acid: one molecule diverse functions. Crit Rev Biotechnol 2016; 37:579-597. [DOI: 10.1080/07388551.2016.1207153] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Michio Hashimoto
- Department of Environmental Physiology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Shahdat Hossain
- Department of Environmental Physiology, Shimane University Faculty of Medicine, Izumo, Japan
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Dhaka, Bangladesh
| | - Abdullah Al Mamun
- Department of Environmental Physiology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Kentaro Matsuzaki
- Department of Environmental Physiology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Hiroyuki Arai
- Department of Geriatrics and Gerontology, Division of Brain Sciences, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| |
Collapse
|