1
|
Zinkow A, Grodzicki W, Czerwińska M, Dziendzikowska K. Molecular Mechanisms Linking Omega-3 Fatty Acids and the Gut-Brain Axis. Molecules 2024; 30:71. [PMID: 39795128 PMCID: PMC11721018 DOI: 10.3390/molecules30010071] [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: 11/25/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
The gut-brain axis (GBA) is a complex communication network connecting the gastrointestinal tract (GIT) and the central nervous system (CNS) through neuronal, endocrine, metabolic, and immune pathways. Omega-3 (n-3) fatty acids, particularly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are crucial food components that may modulate the function of this axis through molecular mechanisms. Derived mainly from marine sources, these long-chain polyunsaturated fatty acids are integral to cell membrane structure, enhancing fluidity and influencing neurotransmitter function and signal transduction. Additionally, n-3 fatty acids modulate inflammation by altering eicosanoid production, reducing proinflammatory cytokines, and promoting anti-inflammatory mediators. These actions help preserve the integrity of cellular barriers like the intestinal and blood-brain barriers. In the CNS, EPA and DHA support neurogenesis, synaptic plasticity, and neurotransmission, improving cognitive functions. They also regulate the hypothalamic-pituitary-adrenal (HPA) axis by reducing excessive cortisol production, associated with stress responses and mental health disorders. Furthermore, n-3 fatty acids influence the composition and function of the gut microbiota, promoting beneficial bacterial populations abundance that contribute to gut health and improve systemic immunity. Their multifaceted roles within the GBA underscore their significance in maintaining homeostasis and supporting mental well-being.
Collapse
Affiliation(s)
| | | | | | - Katarzyna Dziendzikowska
- Department of Dietetics, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences—SGGW, Nowoursynowska 159C, 02-776 Warsaw, Poland; (A.Z.); (W.G.); (M.C.)
| |
Collapse
|
2
|
Ma B, Yang R, Chen X, Wang Q, Zhang T, Wen R, Yang M, Lei C, Wang H. Synergistic antimicrobial activity of alpha-linolenic acid in combination with tetracycline or florfenicol against multidrug-resistant Salmonella typhimurium. Microb Pathog 2024; 196:106982. [PMID: 39332543 DOI: 10.1016/j.micpath.2024.106982] [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: 06/09/2024] [Revised: 09/13/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
Salmonella is a major foodborne pathogen that can be transmitted from livestock and poultry to humans through the food chain. Due to the widespread use of antibiotics, antibiotic resistance Salmonella has become an important factor threatening food safety. Combining antibiotic and non-antibiotic agents is a promising approach to address the widespread emergence of antibiotic-resistant pathogens. In this study, we investigated the antibiotic resistance profile and molecular characterization of different serotypes of Salmonella isolated from large-scale egg farms using drug susceptibility testing and whole genome sequencing. The synergistic effect of alpha-linolenic acid (ALA) with antibiotics was evaluated using the checkerboard test and time-kill curve. The molecular mechanism of α-linolenic acid synergism was explored using biochemical assays, pull-down assays, and molecular docking. In vivo efficacy of ALA in combination with florfenicol (FFC) or tetracycline (TET) against multidrug-resistant (MDR) Salmonella enterica subsp. enterica serovar typhimurium was also investigated using a mouse model. We found that ALA reduced the minimum inhibitory concentration (MIC) of tetracycline and florfenicol in all strains tested. When ALA (512 mg/L) was combined with florfenicol (32 mg/L) or tetracycline (16 mg/L), we observed disruption of cell membrane integrity, increased outer membrane permeability, lowered cell membrane potential, and inhibition of proton-drive-dependent efflux pumps. The synergistic treatment also inhibited biofilm production and promoted oxidative damage. These changes together led to an increase in bacterial antibiotic susceptibility. The improved efficacy of ALA combination treatment with antibiotics was validated in the mouse model. Molecular docking results indicate that ALA can bind to membrane proteins via hydrogen bonding. Our findings demonstrated that combined treatment using ALA and antibiotics is effective in preventing infections involving MDR bacteria. Our results are of great significance for the scientific and effective prevention and control of antibiotic resistance Salmonella, as well as ensuring food safety.
Collapse
Affiliation(s)
- Boheng Ma
- College of Life Sciences, Sichuan University, Chengdu, PR China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, PR China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, PR China
| | - Rujie Yang
- Luzhou Pinchuang Technology Co. Ltd., Luzhou, PR China; National Engineering Research Center of Solid-State Brewing, Luzhou Laojiao Co. Ltd., Luzhou, PR China
| | - Xuan Chen
- College of Life Sciences, Sichuan University, Chengdu, PR China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, PR China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, PR China
| | - Qin Wang
- College of Life Sciences, Sichuan University, Chengdu, PR China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, PR China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, PR China
| | - Tiejun Zhang
- College of Life Sciences, Sichuan University, Chengdu, PR China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, PR China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, PR China
| | - Renqiao Wen
- College of Life Sciences, Sichuan University, Chengdu, PR China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, PR China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, PR China
| | - Ming Yang
- College of Life Sciences, Sichuan University, Chengdu, PR China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, PR China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, PR China
| | - Changwei Lei
- College of Life Sciences, Sichuan University, Chengdu, PR China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, PR China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, PR China.
| | - Hongning Wang
- College of Life Sciences, Sichuan University, Chengdu, PR China; Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Chengdu, PR China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu, PR China.
| |
Collapse
|
3
|
Deng Y, Wang R, Li X, Tan X, Zhang Y, Gooneratne R, Li J. Fish Oil Ameliorates Vibrio parahaemolyticus Infection in Mice by Restoring Colonic Microbiota, Metabolic Profiles, and Immune Homeostasis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:6920-6934. [PMID: 37126589 DOI: 10.1021/acs.jafc.2c08559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The effect of fish oil (FO) on colonic function, immunity, and microbiota was investigated in Vibrio parahaemolyticus (Vp)-infected C57BL/6J mice. Mice intragastrically presupplemented with FO (4.0 mg) significantly reduced Vp infection as evidenced by stabilizing body weight and reducing disease activity index score and immune organ ratios. FO minimized colonic pathological damage, strengthened the mucosal barrier, and sustained epithelial permeability by increasing epithelial crypt depth, goblet cell numbers, and tight junctions and inhibiting colonic collagen accumulation and fibrosis protein expression. Mechanistically, FO enhanced immunity by decreasing colonic CD3+ T cells, increasing CD4+ T cells, downregulating the TLR4 pathway, reducing interleukin-17 (IL-17) and tumor necrosis factor-α, and increasing immune cytokine IL-4 and interferon-γ levels. Additionally, FO maintained colonic microbiota eubiosis by improving microbial diversity and boosting Clostridium, Akkermansia, and Roseburia growth and their derived propionic acid and butyric acid levels. Collectively, FO alleviated Vp infection by enriching beneficial colonic microbiota and metabolites and restoring immune homeostasis.
Collapse
Affiliation(s)
- Yijia Deng
- College of Food Science, Southwest University, Chongqing 400715, China
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Rundong Wang
- College of Food Science, Southwest University, Chongqing 400715, China
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Xuepeng Li
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Xiqian Tan
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Yuhao Zhang
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Ravi Gooneratne
- Department of Wine, Food and Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln 7647, Canterbury, New Zealand
| | - Jianrong Li
- College of Food Science, Southwest University, Chongqing 400715, China
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| |
Collapse
|
4
|
Zhang Q, Zhang L, Chen C, Li P, Lu B. The gut microbiota-artery axis: A bridge between dietary lipids and atherosclerosis? Prog Lipid Res 2023; 89:101209. [PMID: 36473673 DOI: 10.1016/j.plipres.2022.101209] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/09/2022]
Abstract
Atherosclerotic cardiovascular disease is one of the major leading global causes of death. Growing evidence has demonstrated that gut microbiota (GM) and its metabolites play a pivotal role in the onset and progression of atherosclerosis (AS), now known as GM-artery axis. There are interactions between dietary lipids and GM, which ultimately affect GM and its metabolites. Given these two aspects, the GM-artery axis may play a mediating role between dietary lipids and AS. Diets rich in saturated fatty acids (SFAs), omega-6 polyunsaturated fatty acids (n-6 PUFAs), industrial trans fatty acids (TFAs), and cholesterol can increase the levels of atherogenic microbes and metabolites, whereas monounsaturated fatty acids (MUFAs), ruminant TFAs, and phytosterols (PS) can increase the levels of antiatherogenic microbes and metabolites. Actually, dietary phosphatidylcholine (PC), sphingomyelin (SM), and omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been demonstrated to affect AS via the GM-artery axis. Therefore, that GM-artery axis acts as a communication bridge between dietary lipids and AS. Herein, we will describe the molecular mechanism of GM-artery axis in AS and discuss the complex interactions between dietary lipids and GM. In particular, we will highlight the evidence and potential mechanisms of dietary lipids affecting AS via GM-artery axis.
Collapse
Affiliation(s)
- Qinjun Zhang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China; Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
| | - Liangxiao Zhang
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wubhan, China
| | - Cheng Chen
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peiwu Li
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wubhan, China
| | - Baiyi Lu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China; Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
5
|
Yuan Y, Liu Y, He Y, Zhang B, Zhao L, Tian S, Wang Q, Chen S, Li Z, Liang S, Hou G, Liu B, Li Y. Intestinal-targeted nanotubes-in-microgels composite carriers for capsaicin delivery and their effect for alleviation of Salmonella induced enteritis. Biomaterials 2022; 287:121613. [PMID: 35700621 DOI: 10.1016/j.biomaterials.2022.121613] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 04/05/2022] [Accepted: 05/30/2022] [Indexed: 02/09/2023]
Abstract
Salmonella is a word-wide food-borne pathogen, which can cause severe enteritis and intestinal microbiota imbalance. Capsaicin (Cap), a food-based bioactive ingredient, has antibacterial and anti-inflammatory properties. However, its low solubility, low bioavailability and the irritation to digestive tract greatly limit its applications. Here, an intestinal responsively "nanotubes-in-microgel" composite carrier was constructed by capturing α-lactalbumin (α-lac) nanotubes in low-methoxy pectin microgels (LMP-NT) (52 μm). Cap was loaded in such system via hydrophobic interaction with a loading capacity of 38.02 mg/g. The LMP microgels remained stable and protected NT/Cap from early releasing in the gastric condition. It showed an excellent mucoadhesive capacity, which can prolong the intestinal retention up to 12 h and control release NT/Cap in intestine. Afterward, NT/Cap could penetrate across the mucus layer deeply and enter the intestinal villi epithelial cells efficiently. LMP-NT microgels achieved a mucoadhesive-to-penetrating transition in response to intestinal pH, improving the epithelium absorption and the in vivo bioavailability of Cap. Oral administration of LMP-NT/Cap could effectively alleviate enteritis caused by Salmonella infection and maintain the homeostasis of gut microbiota. Overall, this work suggested that LMP-NT composite microgels were promising for intestine-targeted and oral delivery of hydrophobic bioactive food compounds.
Collapse
Affiliation(s)
- Yu Yuan
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Ying Liu
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Yang He
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Bingkun Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Liang Zhao
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Simin Tian
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Qimeng Wang
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Shanan Chen
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Zekun Li
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Shuang Liang
- Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Guohua Hou
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Bin Liu
- Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Yuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
6
|
Cabezudo I, Lobertti CA, Véscovi EG, Furlan RLE. Effect-Directed Synthesis of PhoP/PhoQ Inhibitors to Regulate Salmonella Virulence. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:6755-6763. [PMID: 35607919 DOI: 10.1021/acs.jafc.2c01087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Salmonella spp. are among the leading bacterial causes of foodborne infections. The PhoP/PhoQ two-component regulatory system serves as a master virulence regulator in Salmonella. Although PhoP/PhoQ represents an ideal target for disarming Salmonella virulence, it has very few inhibitors reported so far. We describe a novel platform by which an inhibitor was selected out of around 185 compounds directly from reaction media containing thiosemicarbazones and mono-, di-, and trihydrazones. To achieve this, tandem library preparation, thin-layer chromatography (TLC) bioautography, and effect-directed deconvolution were applied. We illustrate the potential of this effect-directed synthesis for the identification of new useful bioactive compounds for the food field.
Collapse
Affiliation(s)
- Ignacio Cabezudo
- Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rosario 2000 Argentina
| | - Carlos A Lobertti
- Instituto de Biología Molecular y Celular de Rosario (IBR CONICET), Rosario S2000EZP Argentina
- Biofísica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario 2000 Argentina
| | - Eleonora García Véscovi
- Instituto de Biología Molecular y Celular de Rosario (IBR CONICET), Rosario S2000EZP Argentina
| | - Ricardo L E Furlan
- Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rosario 2000 Argentina
| |
Collapse
|
7
|
Dhakal J, Aldrich CG. Temperature-Dependent Antimicrobial Activity of Menhaden Fish Oil In Vitro and on Pet Food Kibbles against Salmonella. J Food Prot 2022; 85:478-483. [PMID: 34882221 DOI: 10.4315/jfp-21-336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/08/2021] [Indexed: 11/11/2022]
Abstract
ABSTRACT Fish oil inclusion into a dry pet food provides a source of long-chain omega-3 fatty acids. Polyunsaturated fatty acids in fish oil have antibacterial activity against various foodborne pathogens, such as Salmonella and pathogenic Escherichia coli. The purpose of this study was to determine the effect of temperature applied to dry pet food kibbles on the antimicrobial activity of menhaden fish oil against Salmonella spp. Sterile menhaden oil was inoculated with ∼8 log of a Salmonella cocktail (∼3% moisture; Salmonella Enteritidis, Salmonella Heidelberg, and Salmonella Typhimurium) and incubated at 25, 37, and 45°C. Microbiological evaluation of the water phase was done after 2 h on tryptic soy agar. Sterile kibbles were coated with fish oil (7.0%, w/w). Canola oil coating was kept as a control. One hour after coating, the kibbles were inoculated with ∼9 log of Salmonella and incubated at the respective temperature. The microbiological evaluation was conducted at 0, 2, 6, 12, and 24 h. The oil phase of the fish oil system was negative for Salmonella after 2 h of incubation and confirmed by enrichment and PCR. From the water phase, 8.1 and 7.3 log were recovered at 25 and 37°C, respectively, and no Salmonella was detected at 45°C. On the kibble, menhaden oil had higher antimicrobial (P ≤ 0.05) activity after 12 h at 25°C and throughout the experiment at 37°C. At 45°C, the fish oil had a superior antimicrobial activity against the Salmonella cocktail after 2 h. When the fish oil alone was compared at different temperatures, a higher antimicrobial activity was observed at 37 and 45°C across all time points. The results indicate antimicrobial activity of menhaden oil increases with temperature. This is an important finding to the pet food industry: a higher fat holding temperature (∼45°C) and the application process may help mitigate Salmonella on extruded kibbles. HIGHLIGHTS
Collapse
Affiliation(s)
- Janak Dhakal
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska 68588
| | - Charles G Aldrich
- Department of Grain Science and Industry, Kansas State University, Manhattan, Kansas 66506, USA
| |
Collapse
|
8
|
Liu J, Wang Y, Zhang G, Liu L, Peng X. Multi-Omics Analysis Reveals Changes in the Intestinal Microbiome, Transcriptome, and Methylome in a Rat Model of Chronic Non-bacterial Prostatitis: Indications for the Existence of the Gut-Prostate Axis. Front Physiol 2022; 12:753034. [PMID: 35087414 PMCID: PMC8787367 DOI: 10.3389/fphys.2021.753034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 12/15/2021] [Indexed: 12/31/2022] Open
Abstract
Chronic non-bacterial prostatitis (CNP) is one of the most prevalent diseases in human males worldwide. In 2005, the prostate-gut axis was first proposed to indicate the close relationship between the prostate and the intestine. This study investigated CNP-induced changes of the gut microbiota, gene expression and DNA methylation in a rat model by using multi-omics analysis. Firstly, 16S rDNA sequencing presented an altered structure of the microbiota in cecum of CNP rats. Then, transcriptomic analysis revealed that the expression of 185 genes in intestinal epithelium was significantly changed by CNP. These changes can participate in the immune system, digestive system, metabolic process, etc. Finally, methylC-capture sequencing (MCC-Seq) found 73,232 differentially methylated sites (DMSs) in the DNA of intestinal epithelium between control and CNP rats. A combined analysis of methylomics and transcriptomics suggested an epigenetic mechanism for CNP-induced differential expression genes correlated with intestinal barrier function, immunity, metabolism, enteric infectious disease, etc. More importantly, the transcriptomic, methylomic and gut microbial changes were highly correlated with multiple processes including intestinal immunity, metabolism and epithelial barrier function. In this study, disrupted homeostasis in the gut microbiota, gene expression and DNA methylation were reported in CNP, which supports the existence of the gut-prostate axis.
Collapse
Affiliation(s)
- Junsheng Liu
- Department of Food Science and Engineering, Jinan University, Guangzhou, China.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yihe Wang
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Guangwen Zhang
- Department of Food Science and Engineering, Jinan University, Guangzhou, China
| | - Liu Liu
- Department of Food Science and Engineering, Jinan University, Guangzhou, China
| | - Xichun Peng
- Department of Food Science and Engineering, Jinan University, Guangzhou, China
| |
Collapse
|
9
|
Dey TK, Bose P, Paul S, Karmakar BC, Saha RN, Gope A, Koley H, Ghosh A, Dutta S, Dhar P, Mukhopadhyay AKKUMAR. Protective efficacy of fish oil nanoemulsion against non-typhoidal Salmonella mediated mucosal inflammation and loss of barrier function. Food Funct 2022; 13:10083-10095. [DOI: 10.1039/d1fo04419b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Non-typhoidal Salmonella serotypes are well-adapted to utilize the inflammation for colonization in mammalian gut mucosa and bring down the integrity of the epithelial barrier in mammalian intestine. The present study...
Collapse
|
10
|
Liu J, Liu L, Zhang G, Peng X. Poria cocos polysaccharides attenuate chronic nonbacterial prostatitis by targeting the gut microbiota: Comparative study of Poria cocos polysaccharides and finasteride in treating chronic prostatitis. Int J Biol Macromol 2021; 189:346-355. [PMID: 34428489 DOI: 10.1016/j.ijbiomac.2021.08.139] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/03/2021] [Accepted: 08/16/2021] [Indexed: 02/08/2023]
Abstract
Finasteride is an antiandrogenic drug used for the clinical treatment of chronic nonbacterial prostatitis (CNP). Recently, we reported the anti-CNP activity of Poria cocos polysaccharides (PPs) in a rat model. In this study, we compared the differences between PPs and finasteride in treating CNP, especially their effects on the gut microbiota. Results showed that both PPs and finasteride significantly reduced the prostate weight and prostate index of CNP rats, and improved the histological damages in the inflamed prostate. Moreover, PPs and finasteride inhibited the production of pro-inflammatory cytokines (TNF-α, IL-2 and IL-8) and androgens (dihydrotestosterone and testosterone). By 16S rDNA sequencing, PPs and finasteride were found to reprogram the gut microbiota into distinct profiles. Further analysis presented that PPs but not finasteride recovered CNP-induced changes in the gut microbiota, including Ruminococcaceae NK4A214 group, uncultured bacterium f Ruminococcaceae, Ruminiclostridium 9, Phascolarctobacterium, Coriobacteriaceae UCG-002 and Oribacterium. LDA effect size (LEfSe) analysis revealed that PPs recovered the gut microbiota by targeting Ruminococcaceae NK4A214 group. Our results suggested that PPs alleviated CNP via different mechanisms from finasteride, especially by regulating the gut microbiota, which offers therapeutic target for the treatment of CNP.
Collapse
Affiliation(s)
- Junsheng Liu
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong 510632, PR China
| | - Liu Liu
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong 510632, PR China
| | - Guangwen Zhang
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong 510632, PR China
| | - Xichun Peng
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong 510632, PR China.
| |
Collapse
|
11
|
Liu J, Liu J, Liu L, Zhang G, Zhou A, Peng X. The gut microbiota alteration and the key bacteria in Astragalus polysaccharides (APS)-improved osteoporosis. Food Res Int 2020; 138:109811. [DOI: 10.1016/j.foodres.2020.109811] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 10/11/2020] [Accepted: 10/13/2020] [Indexed: 12/16/2022]
|
12
|
Protection of Galacto-Oligosaccharide against E. coli O157 Colonization through Enhancing Gut Barrier Function and Modulating Gut Microbiota. Foods 2020; 9:foods9111710. [PMID: 33233359 PMCID: PMC7700679 DOI: 10.3390/foods9111710] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/12/2020] [Accepted: 11/18/2020] [Indexed: 12/14/2022] Open
Abstract
Galacto-oligosaccharide (GOS) has been added to infant formula as prebiotics and can bring many benefits to human health. This study proved the effect of GOS in prevention and alleviation against E. coli O157 invasion and colonization and the mechanism behind this was explored in a mice model. The results showed that the expression of Muc2 and Occlaudin were both significantly down-regulated (p < 0.05) by E. coli O157 infection, while GOS alleviated this phenomenon, which means that GOS can reduce the colonization of E. coli O157 by enhancing the gut barrier function. Through the determination of inflammatory cytokines, we found that GOS can relieve inflammation caused by pathogens. At the same time, GOS can promote the growth of probiotics such as Akkermansia, Ruminococcaceae and Bacteroides, thus modulating microorganism environments and improving short chain fatty acid (SCFA) levels in the intestine. This study provides an explanation for the mechanism behind the protection of GOS against pathogen infection.
Collapse
|
13
|
Liu J, Yu J, Peng X. Poria cocos Polysaccharides Alleviates Chronic Nonbacterial Prostatitis by Preventing Oxidative Stress, Regulating Hormone Production, Modifying Gut Microbiota, and Remodeling the DNA Methylome. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:12661-12670. [PMID: 33119288 DOI: 10.1021/acs.jafc.0c05943] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Chronic nonbacterial prostatitis (CNP) is a common male disease with high incidence and low cure rate. This study aims to investigate the anti-CNP potential of Poria cocos polysaccharides (PPs) in a λ-carrageenan-induced CNP rat model. Results showed that PPs exerted anti-CNP functions by reducing the prostate weight and prostate index as well as the level of C-reactive protein (CRP) and pro-inflammatory cytokines (TNF-α and IL-1β). Further analysis on sex hormones revealed that PPs could favor CNP alleviation by regulating the production of testosterone (T), dihydrotestosterone (DTH), and estradiol (E2). PPs could also alleviate CNP by regulating the level of inducible nitric oxide synthase (iNOS), malonaldehyde (MDA), and superoxide diamutase (SOD) in inflamed prostate, thereby enhancing the anti-oxidative stress activity. As most non-digestive polysaccharides are fermented by gut microbiota rather than being digested directly by the host, we further analyzed PP-induced changes in gut microbiota. Microbiomic analysis revealed that PPs significantly change the profile of gut microbiota. Moreover, the relative abundance of five genera was recovered by PPs with a dose-effect relationship, thereby being suggested to play critical roles in the alleviation of CNP. Epigenomic (methylomic) analysis showed that PPs remodeled the DNA methylome of intestinal epithelia, by which PPs might modify hormone production. In the present study, we reported the anti-CNP activity of PPs as well as the involved mechanisms.
Collapse
Affiliation(s)
- Junsheng Liu
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong 510632, P. R. China
| | - Juntong Yu
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong 510632, P. R. China
| | - Xichun Peng
- Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong 510632, P. R. China
| |
Collapse
|
14
|
Herndon JL, Peters RE, Hofer RN, Simmons TB, Symes SJ, Giles DK. Exogenous polyunsaturated fatty acids (PUFAs) promote changes in growth, phospholipid composition, membrane permeability and virulence phenotypes in Escherichia coli. BMC Microbiol 2020; 20:305. [PMID: 33046008 PMCID: PMC7552566 DOI: 10.1186/s12866-020-01988-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The utilization of exogenous fatty acids by Gram-negative bacteria has been linked to many cellular processes, including fatty acid oxidation for metabolic gain, assimilation into membrane phospholipids, and control of phenotypes associated with virulence. The expanded fatty acid handling capabilities have been demonstrated in several bacteria of medical importance; however, a survey of the polyunsaturated fatty acid responses in the model organism Escherichia coli has not been performed. The current study examined the impacts of exogenous fatty acids on E. coli. RESULTS All PUFAs elicited higher overall growth, with several fatty acids supporting growth as sole carbon sources. Most PUFAs were incorporated into membrane phospholipids as determined by Ultra performance liquid chromatography-mass spectrometry, whereas membrane permeability was variably affected as measured by two separate dye uptake assays. Biofilm formation, swimming motility and antimicrobial peptide resistance were altered in the presence of PUFAs, with arachidonic and docosahexaenoic acids eliciting strong alteration to these phenotypes. CONCLUSIONS The findings herein add E. coli to the growing list of Gram-negative bacteria with broader capabilities for utilizing and responding to exogenous fatty acids. Understanding bacterial responses to PUFAs may lead to microbial behavioral control regimens for disease prevention.
Collapse
Affiliation(s)
- Joshua L. Herndon
- Department of Biology, Geology, and Environmental Science, The University of Tennessee at Chattanooga, Chattanooga, TN USA
| | - Rachel E. Peters
- Department of Biology, Geology, and Environmental Science, The University of Tennessee at Chattanooga, Chattanooga, TN USA
| | - Rachel N. Hofer
- Department of Chemistry and Physics, The University of Tennessee at Chattanooga, Chattanooga, TN USA
| | - Timothy B. Simmons
- Department of Biology, Geology, and Environmental Science, The University of Tennessee at Chattanooga, Chattanooga, TN USA
| | - Steven J. Symes
- Department of Chemistry and Physics, The University of Tennessee at Chattanooga, Chattanooga, TN USA
| | - David K. Giles
- Department of Biology, Geology, and Environmental Science, The University of Tennessee at Chattanooga, Chattanooga, TN USA
| |
Collapse
|