1
|
Gopalsamy RG, Antony PJ, Athesh K, Hillary VE, Montalvão MM, Hariharan G, Santana LADM, Borges LP, Gurgel RQ. Dietary essential oil components: A systematic review of preclinical studies on the management of gastrointestinal diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156630. [PMID: 40085990 DOI: 10.1016/j.phymed.2025.156630] [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: 10/28/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND The gut is responsible for the digestion and absorption of nutrients, immune regulation, and barrier function. However, factors like poor diet, stress, and infection, can disrupt the balance of the gut microbiota and lead to intestinal inflammation and dysfunction. PURPOSE This systematic review aims to evaluate the effects of dietary plants-derived essential oil components on gut health and intestinal functions in animal models. METHODS The literature was gathered from the Scopus, Web of Science, PubMed, and Embase databases by using related search terms, such as "dietary plants", "dietary sources", "essential oils", "gut health", "intestine", "anti-inflammatory", "antioxidant", and "gut microbiota". RESULTS The results indicate that plant-derived dietary essential oil components, such as butyrolactone-I, carvacrol, cinnamaldehyde, citral, D-limonene, eugenol, farnesol, geraniol, indole, nerolidol, oleic acid, thymol, trans-anethole, vanillin, α-bisabolol, α-linolenic acid, α-pinene, α-terpineol, β-carotene, β-caryophyllene, and β-myrcene have been found to regulate gut health by influencing vital signalling pathways associated with inflammation. Dietary essential oil components modulate the expression of tumor necrosis factor alpha, interleukin 1 beta (IL-1β), interleukin (IL)-6, IL-10, inducible nitric oxide synthase, cyclooxygenase-2, toll-like receptor-4, matrix metalloproteinase, and interferon gamma in mitigating gut inflammation. The primary signalling molecules controlled by these molecules were AMP-activated protein kinase (AMPK), protein kinase B, extracellular signal-regulated kinase, c-Jun N-terminal kinase, mitogen-activated protein kinase, myeloid differentiation primary response 88, nuclear factor erythroid-2-related factor-2, and phosphoinositide 3-kinase (PI3K). Moreover, these phytochemicals have been shown to improve glucose homeostasis by regulating glucose transporter 4, glucagon-like peptide-1, peroxisome proliferator-activated receptor gamma, nuclear factor kappa B, AMPK, PI3K, and uncoupling protein-1. They can also reduce thiobarbituric acid reactive substance, malondialdehyde, and oxidative stress and enhance superoxide dismutase, catalase, and glutathione peroxidase levels. CONCLUSION In conclusion, dietary plants-derived essential oil components have the potential to mitigate inflammation and oxidative stress in the gut. However, additional clinical investigations are necessary to confirm their complete potential in improving human gut health functions.
Collapse
Affiliation(s)
- Rajiv Gandhi Gopalsamy
- Division of Phytochemistry and Drug-Design, Department of Biosciences, Rajagiri College of Social Sciences (Autonomous), Kochi, Kerala, India; Postgraduate Program of Health Sciences (PPGCS), Federal University of Sergipe, Campus Prof. João Cardoso Nascimento, Aracaju, Sergipe, Brazil
| | - Poovathumkal James Antony
- Department of Microbiology, North Bengal University, St. Joseph's College, Darjeeling, West Bengal, India
| | - Kumaraswamy Athesh
- School of Sciences, Bharata Mata College (Autonomous), Kochi, Kerala, India
| | - Varghese Edwin Hillary
- Division of Phytochemistry and Drug-Design, Department of Biosciences, Rajagiri College of Social Sciences (Autonomous), Kochi, Kerala, India
| | | | | | | | - Lysandro Pinto Borges
- Department of Pharmacy, Federal University of Sergipe, São Cristovão, Sergipe, Brazil
| | - Ricardo Queiroz Gurgel
- Postgraduate Program of Health Sciences (PPGCS), Federal University of Sergipe, Campus Prof. João Cardoso Nascimento, Aracaju, Sergipe, Brazil.
| |
Collapse
|
2
|
Lu Y, Chen L, Lin Y, Zhang Y, Wang Y, Yu W, Ren F, Guo H. Fatty acid metabolism: The crossroads in intestinal homeostasis and tumor. Metabolism 2025; 169:156273. [PMID: 40280478 DOI: 10.1016/j.metabol.2025.156273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/09/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
Fatty acids (FAs) have various functions on cell regulation considering their abundant types and metabolic pathways. In addition, the relation between FA and other nutritional metabolism makes their functions more complex. As the first place for diet-derived FA metabolism, intestine is significantly influenced despite lack of clear conclusions due to the inconsistent findings. In this review, we discuss the regulation of fatty acid metabolism on the fate of intestinal stem cells in homeostasis and disorders, and also focus on the intestinal tumor development and treatment from the aspect of gut microbiota-epithelium-immune interaction. We summarize that the balances between FA oxidation and glycolysis, between oxidative phosphorylation and ketogenesis, between catabolism and anabolism, and the specific roles of individual FA types determine the diverse effects of intestinal FA metabolism in different cases. We hope this will inspire further dissection and suggest precise dietary/metabolic intervention for different demands related to intestinal health.
Collapse
Affiliation(s)
- Yao Lu
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Lining Chen
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yingying Lin
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yafei Zhang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yuqi Wang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Weiru Yu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Fazheng Ren
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| | - Huiyuan Guo
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
3
|
Samrit T, Osotprasit S, Chaiwichien A, Suksomboon P, Chansap S, Suthisintong T, Changklungmoa N, Kueakhai P. Microbial effects of cold-pressed Sacha inchi oil supplementation in rats. PLoS One 2025; 20:e0319066. [PMID: 39977445 PMCID: PMC11841868 DOI: 10.1371/journal.pone.0319066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 01/26/2025] [Indexed: 02/22/2025] Open
Abstract
Oil supplements have various benefits for metabolism, particularly Sacha inchi oil (SI), which is rich in polyunsaturated fatty acids (PUFAs) such as ω-3 and fat-soluble vitamins. However, the impacts of oil supplements on gut health remain unclear. The aim of this study was to compare the effects of an SI supplement with those of lard oil (LO), known for its high saturated fatty acid content, and a normal diet on gut health in male Sprague Dawley rats for 12 consecutive weeks. Fecal DNA was used to assess gut microbiota diversity and species abundance, diversity, and function prediction. Colon tissue from each rat was examined for colon crypt depth and histology. Rats administered the LO supplement exhibited higher dysbiosis than those administered the SI supplement, with the LO supplement influencing the relative abundance of various bacteria at the genus level. A KEGG analysis was conducted to examine the effects on metabolic pathways, revealing that the SI supplement promoted carbohydrate metabolism while reducing immune system activity. In contrast, the LO supplement increased replication, repair, and translation activities. A histological analysis of the colon tissues showed no significant alterations in crypt depth or lesions in all groups, indicating that neither supplement induced adverse structural changes in the gut. The results of this study suggest that SI supplementation modulates the gut microbiota, thereby enhancing gut health and metabolic function.
Collapse
Affiliation(s)
- Tepparit Samrit
- Food Bioactive Compounds Research Unit and Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | - Supawadee Osotprasit
- Food Bioactive Compounds Research Unit and Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | - Athit Chaiwichien
- Food Bioactive Compounds Research Unit and Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | - Phawiya Suksomboon
- Food Bioactive Compounds Research Unit and Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | - Supanan Chansap
- Food Bioactive Compounds Research Unit and Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | - Thitikul Suthisintong
- Food Bioactive Compounds Research Unit and Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | - Narin Changklungmoa
- Food Bioactive Compounds Research Unit and Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | - Pornanan Kueakhai
- Food Bioactive Compounds Research Unit and Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| |
Collapse
|
4
|
Zhang X, Lau HCH, Ha S, Liu C, Liang C, Lee HW, Ng QWY, Zhao Y, Ji F, Zhou Y, Pan Y, Song Y, Zhang Y, Lo JCY, Cheung AHK, Wu J, Li X, Xu H, Wong CC, Wong VWS, Yu J. Intestinal TM6SF2 protects against metabolic dysfunction-associated steatohepatitis through the gut-liver axis. Nat Metab 2025; 7:102-119. [PMID: 39779889 PMCID: PMC11774752 DOI: 10.1038/s42255-024-01177-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/14/2024] [Indexed: 01/11/2025]
Abstract
Transmembrane-6 superfamily member 2 (TM6SF2) regulates hepatic fat metabolism and is associated with metabolic dysfunction-associated steatohepatitis (MASH). TM6SF2 genetic variants are associated with steatotic liver disease. The pathogenesis of MASH involves genetic factors and gut microbiota alteration, yet the role of host-microbe interactions in MASH development remains unclear. Here, we discover that mice with intestinal epithelial cell-specific knockout of Tm6sf2 (Tm6sf2ΔIEC) develop MASH, accompanied by impaired intestinal barrier and microbial dysbiosis. Transplanting stools from Tm6sf2ΔIEC mice induces steatohepatitis in germ-free recipient mice, whereas MASH is alleviated in Tm6sf2ΔIEC mice co-housed with wild-type mice. Mechanistically, Tm6sf2-deficient intestinal cells secrete more free fatty acids by interacting with fatty acid-binding protein 5 to induce intestinal barrier dysfunction, enrichment of pathobionts, and elevation of lysophosphatidic acid (LPA) levels. LPA is translocated from the gut to the liver, contributing to lipid accumulation and inflammation. Pharmacological inhibition of the LPA receptor suppresses MASH in both Tm6sf2ΔIEC and wild-type mice. Hence, modulating microbiota or blocking the LPA receptor is a potential therapeutic strategy in TM6SF2 deficiency-induced MASH.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Harry Cheuk-Hay Lau
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Suki Ha
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chuanfa Liu
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Cong Liang
- Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hye Won Lee
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Queena Wing-Yin Ng
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yi Zhao
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fenfen Ji
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yunfei Zhou
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yasi Pan
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yang Song
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Gastroenterology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Yating Zhang
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jennie Ching Yin Lo
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Alvin Ho Kwan Cheung
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Xiamen, China
| | - Xiaoxing Li
- Institute of Precision Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongzhi Xu
- Department of Gastroenterology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Chi Chun Wong
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jun Yu
- Department of Medicine and Therapeutics, Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
5
|
Konopka A, Gawin K, Barszcz M. Hedgehog Signalling Pathway and Its Role in Shaping the Architecture of Intestinal Epithelium. Int J Mol Sci 2024; 25:12007. [PMID: 39596072 PMCID: PMC11593361 DOI: 10.3390/ijms252212007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
The hedgehog (Hh) signalling pathway plays a key role in both embryonic and postnatal development of the intestine and is responsible for gut homeostasis. It regulates stem cell renewal, formation of the villous-crypt axis, differentiation of goblet and Paneth cells, the cell cycle, apoptosis, development of gut innervation, and lipid metabolism. Ligands of the Hh pathway, i.e., Indian hedgehog (Ihh) and Sonic hedgehog (Shh), are expressed by superficial enterocytes but act in the mesenchyme, where they are bound by a Patched receptor localised on myofibroblasts and smooth muscle cells. This activates a cascade leading to the transcription of target genes, including those encoding G1/S-specific cyclin-D2 and -E1, B-cell lymphoma 2, fibroblast growth factor 4, and bone morphogenetic protein 4. The Hh pathway is tightly connected to Wnt signalling. Ihh is the major ligand in the Hh pathway. Its activation inhibits proliferation, while its blocking induces hyperproliferation and triggers a wound-healing response. Thus, Ihh is a negative feedback regulator of cell proliferation. There are data indicating that diet composition may affect the expression of the Hh pathway genes and proteins, which in turn, induces changes in mucosal architecture. This was shown for fat, vitamin A, haem, berberine, and ovotransferrin. The Hh signalling is also affected by the intestinal microbiota, which affects the intestinal barrier integrity. This review highlights the critical importance of the Hh pathway in shaping the intestinal mucosa and summarises the results obtained so far in research on the effect of dietary constituents on the activity of this pathway.
Collapse
Affiliation(s)
- Adrianna Konopka
- Laboratory of Analysis of Gastrointestinal Tract Protective Barrier, Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland;
| | - Kamil Gawin
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland;
| | - Marcin Barszcz
- Laboratory of Analysis of Gastrointestinal Tract Protective Barrier, Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland;
| |
Collapse
|
6
|
Wang Y, Zhang Y, Wang Q, Fan Y, Li W, Liu M, Zhang X, Zhou W, Wang M, Jiang S, Shang E, Duan J. Multi-omics combined to explore the purging mechanism of Rhei Radix et Rhizoma and Magnoliae Officinalis Cortex. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1243:124218. [PMID: 38959707 DOI: 10.1016/j.jchromb.2024.124218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/04/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
Rhei Radix et Rhizoma and Magnoliae Officinalis Cortex have been used together to treat constipation in the clinical practices for more than 2000 years. Nonetheless, their compatibility mechanism is still unclear. In this study, the amelioration of Rhei Radix et Rhizoma combined with Magnoliae Officinalis Cortex on constipation was systematically and comprehensively evaluated. The results showed that their compatibility could markedly shorten gastrointestinal transport time, increase fecal water content and frequency of defecation, improve gastrointestinal hormone disorders and protect colon tissue of constipation rats compared with the single drug. Furthermore, according to 16S rRNA sequencing in conjunction with UPLC-Q-TOF/MS, the combination of two herbal medications could greatly raise the number of salutary bacteria (Lachnospiraceae, Romboutsia and Subdoligranulum) while decreasing the abundance of pathogenic bacteria (Erysipelatoclostridiaceae). And two herb drugs could markedly improve the disorder of fecal metabolic profiles. A total of 7 different metabolites associated with constipation were remarkably shifted by the compatibility of two herbs, which were mainly related to arachidonic acid metabolism, alpha-linolenic acid metabolism, unsaturated fatty acid biosynthesis and other metabolic ways. Thus, the regulation of intestinal microbiome and its metabolism could be a potential target for Rhei Radix et Rhizoma and Magnoliae Officinalis Cortex herb pair to treat constipation. Furthermore, the multi-omics approach utilized in this study, which integrated the microbiome and metabolome, had potential for investigating the mechanism of traditional Chinese medicines.
Collapse
Affiliation(s)
- Yu Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Yun Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Quyi Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Yuwen Fan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Wenwen Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Meijuan Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Xiaoxiao Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Wenwen Zhou
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Mingyang Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China.
| | - Erxin Shang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Jinao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China.
| |
Collapse
|
7
|
Das UN. Can essential fatty acids (EFAs) prevent and ameliorate post-COVID-19 long haul manifestations? Lipids Health Dis 2024; 23:112. [PMID: 38641607 PMCID: PMC11027247 DOI: 10.1186/s12944-024-02090-4] [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: 01/17/2024] [Accepted: 03/25/2024] [Indexed: 04/21/2024] Open
Abstract
It is hypothesized that COVID-19, post-COVID and post-mRNA COVID-19 (and other related) vaccine manifestations including "long haul syndrome" are due to deficiency of essential fatty acids (EFAs) and dysregulation of their metabolism. This proposal is based on the observation that EFAs and their metabolites can modulate the swift immunostimulatory response of SARS-CoV-2 and similar enveloped viruses, suppress inappropriate cytokine release, possess cytoprotective action, modulate serotonin and bradykinin production and other neurotransmitters, inhibit NF-kB activation, regulate cGAS-STING pathway, modulate gut microbiota, inhibit platelet activation, regulate macrophage and leukocyte function, enhance wound healing and facilitate tissue regeneration and restore homeostasis. This implies that administration of EFAs could be of benefit in the prevention and management of COVID-19 and its associated complications.
Collapse
Affiliation(s)
- Undurti N Das
- UND Life Sciences, 2221 NW 5th St, Battle ground, WA, 98604, USA.
- Department of Biotechnology, Indian Institute of Technology-Hyderabad, Sangareddy, Telangana, India.
- Department of Immunology and Rheumatology, Arete Hospitals, Gachibowli, Hyderabad, 4500032, India.
| |
Collapse
|
8
|
Wittwer AE, Lee SG, Ranadheera CS. Potential associations between organic dairy products, gut microbiome, and gut health: A review. Food Res Int 2023; 172:113195. [PMID: 37689944 DOI: 10.1016/j.foodres.2023.113195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 06/24/2023] [Accepted: 06/27/2023] [Indexed: 09/11/2023]
Abstract
Organic products have received longstanding, widespread attention for their nutritional and ecological benefits, as they are said to have certain positive health attributes and contain fewer harmful compounds than conventional (or non-organic) products. We reviewed the recent literature to examine potential associations between nutrient composition, gut microbiota, and gut health effects in recent comparative studies of organic and conventional dairy products. Trends of increased ratios of omega-3 to omega-6 polyunsaturated fatty acids and unsaturated to saturated fat, increased fat-soluble vitamin content, and decreased levels of certain pernicious contaminants in organic milk were observed across the studies reviewed. Studies of the metabolism of these nutrients in both in vitro and in vivo settings, and their or their metabolites' interaction with the intestinal epithelium show that nutrients enriched in organic dairy products may support host nutrient uptake and mediate gut inflammation. Research on the effects of single food products or classes of food products on gut health is rare. The extent of these benefits is highly likely to be mediated by both the magnitude of the difference in nutrient types and quantities, and by dietary intake levels of dairy products. Intervention studies directly examining the different effects of organic and conventional dairy products on gut health in humans are needed to further elucidate this relationship.
Collapse
Affiliation(s)
- Anna Elizabeth Wittwer
- School of Agriculture, Food & Ecosystem Sciences, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Simon Gardner Lee
- School of Agriculture, Food & Ecosystem Sciences, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Chaminda Senaka Ranadheera
- School of Agriculture, Food & Ecosystem Sciences, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
9
|
Fan N, Fusco JL, Rosenberg DW. Antioxidant and Anti-Inflammatory Properties of Walnut Constituents: Focus on Personalized Cancer Prevention and the Microbiome. Antioxidants (Basel) 2023; 12:982. [PMID: 37237848 PMCID: PMC10215340 DOI: 10.3390/antiox12050982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 05/28/2023] Open
Abstract
Walnuts have been lauded as a 'superfood', containing a remarkable array of natural constituents that may have additive and/or synergistic properties that contribute to reduced cancer risk. Walnuts are a rich source of polyunsaturated fatty acids (PUFAs: alpha-linolenic acid, ALA), tocopherols, antioxidant polyphenols (including ellagitannins), and prebiotics, including fiber (2 g/oz). There is a growing body of evidence that walnuts may contribute in a positive way to the gut microbiome, having a prebiotic potential that promotes the growth of beneficial bacteria. Studies supporting this microbiome-modifying potential include both preclinical cancer models as well as several promising human clinical trials. Mediated both directly and indirectly via its actions on the microbiome, many of the beneficial properties of walnuts are related to a range of anti-inflammatory properties, including powerful effects on the immune system. Among the most potent constituents of walnuts are the ellagitannins, primarily pedunculagin. After ingestion, the ellagitannins are hydrolyzed at low pH to release ellagic acid (EA), a non-flavonoid polyphenolic that is subsequently metabolized by the microbiota to the bioactive urolithins (hydroxydibenzo[b,d]pyran-6-ones). Several urolithins, including urolithin A, reportedly have potent anti-inflammatory properties. These properties of walnuts provide the rationale for including this tree nut as part of a healthy diet for reducing overall disease risk, including colorectal cancer. This review considers the latest information regarding the potential anti-cancer and antioxidant properties of walnuts and how they may be incorporated into the diet to provide additional health benefits.
Collapse
Affiliation(s)
| | | | - Daniel W. Rosenberg
- Center for Molecular Oncology, University of Connecticut Health Center, Farmington, CT 06030-3101, USA
| |
Collapse
|
10
|
Myhill LJ, Williams AR. Diet-microbiota crosstalk and immunity to helminth infection. Parasite Immunol 2023; 45:e12965. [PMID: 36571323 DOI: 10.1111/pim.12965] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/06/2022] [Accepted: 12/10/2022] [Indexed: 12/27/2022]
Abstract
Helminths are large multicellular parasites responsible for widespread chronic disease in humans and animals. Intestinal helminths live in close proximity with the host gut microbiota and mucosal immune network, resulting in reciprocal interactions that closely influence the course of infections. Diet composition may strongly regulate gut microbiota composition and intestinal immune function and therefore may play a key role in modulating anti-helminth immune responses. Characterizing the multitude of interactions that exist between different dietary components (e.g., dietary fibres), immune cells, and the microbiota, may shed new light on regulation of helminth-specific immunity. This review focuses on the current knowledge of how metabolism of dietary components shapes immune response during helminth infection, and how this information may be potentially harnessed to design new therapeutics to manage parasitic infections and associated diseases.
Collapse
Affiliation(s)
- Laura J Myhill
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Andrew R Williams
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
11
|
Marín-Tello C, Jintaridth P, Sanchez F, González C, Zelada-Castillo L, Vásquez-Arqueros A, Guevara-Vásquez A, Vieira A. Epigenetic regulation by metabolites from the gut microbiome. Benef Microbes 2022; 13:437-444. [PMID: 36377583 DOI: 10.3920/bm2022.0006] [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] [Indexed: 11/16/2022]
Abstract
The gut microbiome can metabolise food components, such as dietary fibres and various phytochemicals; and the microbiome can also synthesise some nutrients, for example B vitamins. The metabolites produced by bacteria and other micro-organisms in the colon can have implications for health and disease risk. Some of these metabolites are epigenetically active, and can contribute to changes in the chemical modification and structure of chromatin by affecting the activity and expression of epigenetically-active enzymes, for example histone deacetylases and DNA methyltransferases. The epigenetic activity of such gut microbiome metabolites is reviewed herein.
Collapse
Affiliation(s)
- C Marín-Tello
- Food, Metabolism, and Physiology Laboratory, Facultad de Farmacia y Bioquímica, Universidad Nacional de Trujillo, Trujillo 13008, Perú
| | - P Jintaridth
- Department of Tropical Nutrition and Food Science, The Faculty of Tropical Medicine, Mahidol University, 420/6 Rachavithi Road, Rachathevi, Payatai, Bangkok 10400, Thailand
| | - F Sanchez
- Instituto De Educacion Superior Tecnológico Público, 103, Lonya Grande 01556, Perú
| | - C González
- CITE Agroindustrial Chavimochic, Virú 044, Perú
| | - L Zelada-Castillo
- Food, Metabolism, and Physiology Laboratory, Facultad de Farmacia y Bioquímica, Universidad Nacional de Trujillo, Trujillo 13008, Perú
| | - A Vásquez-Arqueros
- Food, Metabolism, and Physiology Laboratory, Facultad de Farmacia y Bioquímica, Universidad Nacional de Trujillo, Trujillo 13008, Perú
| | - A Guevara-Vásquez
- Food, Metabolism, and Physiology Laboratory, Facultad de Farmacia y Bioquímica, Universidad Nacional de Trujillo, Trujillo 13008, Perú
| | - A Vieira
- Nutrition and Metabolism Research Laboratory, BPK-9625, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, V5A 1S6, Canada
| |
Collapse
|
12
|
Luo S, Zhang X, Huang S, Feng X, Zhang X, Xiang D. A monomeric polysaccharide from Polygonatum sibiricum improves cognitive functions in a model of Alzheimer's disease by reshaping the gut microbiota. Int J Biol Macromol 2022; 213:404-415. [PMID: 35661666 DOI: 10.1016/j.ijbiomac.2022.05.185] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 05/16/2022] [Accepted: 05/29/2022] [Indexed: 12/15/2022]
Abstract
Polygonatum sibiricum polysaccharides (PSPs) have the function of nourishing the nerves and beneficial intelligence, but the underlying mechanisms remain unclear. Here we initially isolated and purified a monomeric polysaccharide named PSP-1 from PSPs. UV and IR were utilized for characterizing PSP-1. The molecular weight of PSP-1 was 18.796 kDa. Utilizing 5xFAD mice as a research model, we identified that the initial time of PSP-1 oral administration was 3 months of age for mice by determining the 16S rRNA of fecal samples from wild type (WT) and 5xFAD mice at 3 months or 6 months of age. A 3-month course of PSP-1 improved the pathological behaviors related to memory and cognition, prevented synaptic loss, enhanced microglial phagocytosis of Aβ plaques, and decreased the concentrations of Aβ1-40 and Aβ1-42 in the brains of 5xFAD mice. Moreover, PSP-1 reconstructed the gut microbiota composition, including reducing the relative abundance of Helicobacter, and increasing Akkermansia muciniphila. The gut barrier integrity damage, the inflammatory responses, and the intestinal Aβ deposition were prevented by the PSP-1 treatment. The present study identified a monomeric polysaccharide purified from PSPs that significantly attenuates the cognitive deficits in 5xFAD mice, which could be partly explained by the reshaped gut microbiome.
Collapse
Affiliation(s)
- Shilin Luo
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha 410011, PR China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, PR China
| | - Xin Zhang
- School of Medical Science, Hunan University of Medicine, Huaihua 41800, PR China
| | - Si Huang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha 410011, PR China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, PR China
| | - Xueping Feng
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha 410078, PR China
| | - Xiaojie Zhang
- Department of Psychiatry, the Second Xiangya Hospital, Central South University, Changsha 410011, PR China
| | - Daxiong Xiang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha 410011, PR China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha 410011, PR China.
| |
Collapse
|
13
|
Liu H, Li X, Zhu Y, Huang Y, Zhang Q, Lin S, Fang C, Li L, Lv Y, Mei W, Peng X, Yin J, Liu L. Effect of Plant-Derived n-3 Polyunsaturated Fatty Acids on Blood Lipids and Gut Microbiota: A Double-Blind Randomized Controlled Trial. Front Nutr 2022; 9:830960. [PMID: 35223959 PMCID: PMC8873928 DOI: 10.3389/fnut.2022.830960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/21/2022] [Indexed: 12/29/2022] Open
Abstract
Background Several cardioprotective mechanisms attributed to n-3 polyunsaturated fatty acids (PUFAs) have been widely documented. Significant interest has recently focused on the role of human gut microbiota in metabolic disorders. However, the role of plant-derived n-3 PUFAs on blood lipid profiles is controversial and the effect on gut microbiota is still unclear. Objectives We aimed to perform a double-blind randomized controlled trial to test the effect of plant-derived n-3 PUFAs on the blood lipids and gut microbiota of patients with marginal hyperlipidemia. Methods According to the inclusion and exclusion criteria, 75 participants with marginal hyperlipidemia were randomly assigned to the intervention group (supplied with n-3 PUFA-enriched plant oil) or control group (supplied with corn oil), respectively, for a 3-month treatment. Participants and assessors were blinded to the allocation. The primary outcomes of the trial were the changes in serum lipid levels. Secondary outcomes were changes in gut microbiota and metabolites. For the primary outcomes, we conducted both an intent-to-treat (ITT) analysis and a per protocol (PP) analysis. For the secondary outcomes, we only conducted the PP analysis among the participants who provided fecal sample. Results Fifty-one participants completed the trial. Relative to the control group, the n-3 PUFA supplementation resulted in significant reduction in total cholesterol (TC) levels (−0.43 mmol/L, 95% CI−0.84 to−0.01 mmol/L, P < 0.05). The n-3 PUFA supplementation was also associated with significantly increased relative abundance of Bacteroidetes in phylum level (P < 0.01; false discovery rate (FDR) corrected p = 0.11), and decreased the ratio between Firmicutes and Bacteroidetes (P < 0.05; FDR corrected p = 0.16). At genus level, the intervention of plant derived n-3 PUFAs resulted in a significant decrease in relative abundance of Phascolarctobacterium (P < 0.01; FDR corrected p = 0.18) and Veillonella (P < 0.01; FDR corrected p = 0.18) after the intervention. Conclusions Our results demonstrated that plant-derived n-3 PUFAs beneficially affected the serum levels of TC and decreased the ratio between Firmicutes and Bacteroidetes during the 12-week intervention period, which might confer advantageous consequences for lipid metabolism and intestinal health.
Collapse
Affiliation(s)
- Hongjie Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqin Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yalun Zhu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Huang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Zhang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Lin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Can Fang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linyan Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanling Lv
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenhua Mei
- Zhuhai Center for Disease Control and Prevention, Zhuhai, China
| | - Xiaolin Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Shenzhen Nanshan Centre for Chronic Disease Control, Shenzhen, China
| | - Jiawei Yin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jiawei Yin
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Liegang Liu
| |
Collapse
|
14
|
Szklany K, Engen PA, Naqib A, Green SJ, Keshavarzian A, Lopez Rincon A, Siebrand CJ, Diks MAP, van de Kaa M, Garssen J, Knippels LMJ, Kraneveld AD. Dietary Supplementation throughout Life with Non-Digestible Oligosaccharides and/or n-3 Poly-Unsaturated Fatty Acids in Healthy Mice Modulates the Gut-Immune System-Brain Axis. Nutrients 2021; 14:173. [PMID: 35011046 PMCID: PMC8746884 DOI: 10.3390/nu14010173] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/20/2021] [Accepted: 12/24/2021] [Indexed: 12/11/2022] Open
Abstract
The composition and activity of the intestinal microbial community structures can be beneficially modulated by nutritional components such as non-digestible oligosaccharides and omega-3 poly-unsaturated fatty acids (n-3 PUFAs). These components affect immune function, brain development and behaviour. We investigated the additive effect of a dietary combination of scGOS:lcFOS and n-3 PUFAs on caecal content microbial community structures and development of the immune system, brain and behaviour from day of birth to early adulthood in healthy mice. Male BALB/cByJ mice received a control or enriched diet with a combination of scGOS:lcFOS (9:1) and 6% tuna oil (n-3 PUFAs) or individually scGOS:lcFOS (9:1) or 6% tuna oil (n-3 PUFAs). Behaviour, caecal content microbiota composition, short-chain fatty acid levels, brain monoamine levels, enterochromaffin cells and immune parameters in the mesenteric lymph nodes (MLN) and spleen were assessed. Caecal content microbial community structures displayed differences between the control and dietary groups, and between the dietary groups. Compared to control diet, the scGOS:lcFOS and combination diets increased caecal saccharolytic fermentation activity. The diets enhanced the number of enterochromaffin cells. The combination diet had no effects on the immune cells. Although the dietary effect on behaviour was limited, serotonin and serotonin metabolite levels in the amygdala were increased in the combination diet group. The combination and individual interventions affected caecal content microbial profiles, but had limited effects on behaviour and the immune system. No apparent additive effect was observed when scGOS:lcFOS and n-3 PUFAs were combined. The results suggest that scGOS:lcFOS and n-3 PUFAs together create a balance-the best of both in a healthy host.
Collapse
Affiliation(s)
- Kirsten Szklany
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (K.S.); (A.L.R.); (C.J.S.); (M.A.P.D.); (M.v.d.K.); (J.G.); (L.M.J.K.)
| | - Phillip A. Engen
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush Medical College, Rush University Medical Center, Chicago, IL 60602, USA; (P.A.E.); (A.N.); (A.K.)
| | - Ankur Naqib
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush Medical College, Rush University Medical Center, Chicago, IL 60602, USA; (P.A.E.); (A.N.); (A.K.)
| | - Stefan J. Green
- Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, IL 60602, USA;
| | - Ali Keshavarzian
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush Medical College, Rush University Medical Center, Chicago, IL 60602, USA; (P.A.E.); (A.N.); (A.K.)
- Department of Medicine & Physiology, Rush University Medical Center, Chicago, IL 60602, USA
| | - Alejandro Lopez Rincon
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (K.S.); (A.L.R.); (C.J.S.); (M.A.P.D.); (M.v.d.K.); (J.G.); (L.M.J.K.)
- Department of Data Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3584 EA Utrecht, The Netherlands
| | - Cynthia J. Siebrand
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (K.S.); (A.L.R.); (C.J.S.); (M.A.P.D.); (M.v.d.K.); (J.G.); (L.M.J.K.)
| | - Mara A. P. Diks
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (K.S.); (A.L.R.); (C.J.S.); (M.A.P.D.); (M.v.d.K.); (J.G.); (L.M.J.K.)
| | - Melanie van de Kaa
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (K.S.); (A.L.R.); (C.J.S.); (M.A.P.D.); (M.v.d.K.); (J.G.); (L.M.J.K.)
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (K.S.); (A.L.R.); (C.J.S.); (M.A.P.D.); (M.v.d.K.); (J.G.); (L.M.J.K.)
- Global Centre of Excellence Immunology, Nutricia Danone Research, 3584 CT Utrecht, The Netherlands
| | - Leon M. J. Knippels
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (K.S.); (A.L.R.); (C.J.S.); (M.A.P.D.); (M.v.d.K.); (J.G.); (L.M.J.K.)
- Global Centre of Excellence Immunology, Nutricia Danone Research, 3584 CT Utrecht, The Netherlands
| | - Aletta D. Kraneveld
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands; (K.S.); (A.L.R.); (C.J.S.); (M.A.P.D.); (M.v.d.K.); (J.G.); (L.M.J.K.)
| |
Collapse
|
15
|
Al Bataineh MT, Henschel A, Mousa M, Daou M, Waasia F, Kannout H, Khalili M, Kayasseh MA, Alkhajeh A, Uddin M, Alkaabi N, Tay GK, Feng SF, Yousef AF, Alsafar HS. Gut Microbiota Interplay With COVID-19 Reveals Links to Host Lipid Metabolism Among Middle Eastern Populations. Front Microbiol 2021; 12:761067. [PMID: 34803986 PMCID: PMC8603808 DOI: 10.3389/fmicb.2021.761067] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022] Open
Abstract
The interplay between the compositional changes in the gastrointestinal microbiome, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) susceptibility and severity, and host functions is complex and yet to be fully understood. This study performed 16S rRNA gene-based microbial profiling of 143 subjects. We observed structural and compositional alterations in the gut microbiota of the SARS-CoV-2-infected group in comparison to non-infected controls. The gut microbiota composition of the SARS-CoV-2-infected individuals showed an increase in anti-inflammatory bacteria such as Faecalibacterium (p-value = 1.72 × 10-6) and Bacteroides (p-value = 5.67 × 10-8). We also revealed a higher relative abundance of the highly beneficial butyrate producers such as Anaerostipes (p-value = 1.75 × 10-230), Lachnospiraceae (p-value = 7.14 × 10-65), and Blautia (p-value = 9.22 × 10-18) in the SARS-CoV-2-infected group in comparison to the control group. Moreover, phylogenetic investigation of communities by reconstructing unobserved state (PICRUSt) functional prediction analysis of the 16S rRNA gene abundance data showed substantial differences in the enrichment of metabolic pathways such as lipid, amino acid, carbohydrate, and xenobiotic metabolism, in comparison between both groups. We discovered an enrichment of linoleic acid, ether lipid, glycerolipid, and glycerophospholipid metabolism in the SARS-CoV-2-infected group, suggesting a link to SARS-CoV-2 entry and replication in host cells. We estimate the major contributing genera to the four pathways to be Parabacteroides, Streptococcus, Dorea, and Blautia, respectively. The identified differences provide a new insight to enrich our understanding of SARS-CoV-2-related changes in gut microbiota, their metabolic capabilities, and potential screening biomarkers linked to COVID-19 disease severity.
Collapse
Affiliation(s)
- Mohammad Tahseen Al Bataineh
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.,Department of Genetics and Molecular Biology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Andreas Henschel
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.,Department of Electrical Engineering and Computer Science, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Mira Mousa
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.,Nuffield Department of Women's and Reproduction Health, Oxford University, Oxford, United Kingdom
| | - Marianne Daou
- Department of Chemistry, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Fathimathuz Waasia
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Hussein Kannout
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Mariam Khalili
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Mohd Azzam Kayasseh
- Emirates Specialty Hospital, Dubai Healthcare City, Dubai, United Arab Emirates
| | - Abdulmajeed Alkhajeh
- Medical Education and Research Department, Dubai Health Authority, Dubai, United Arab Emirates
| | - Maimunah Uddin
- Department of Pediatric Infectious Disease, Sheikh Khalifa Medical City, Abu Dhabi, United Arab Emirates
| | - Nawal Alkaabi
- Department of Pediatric Infectious Disease, Sheikh Khalifa Medical City, Abu Dhabi, United Arab Emirates
| | - Guan K Tay
- Division of Psychiatry, Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Samuel F Feng
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.,Department of Mathematics, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Ahmed F Yousef
- Department of Chemistry, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Habiba S Alsafar
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.,Department of Mathematics, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.,Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
16
|
Xie J, Li LF, Dai TY, Qi X, Wang Y, Zheng TZ, Gao XY, Zhang YJ, Ai Y, Ma L, Chang SL, Luo FX, Tian Y, Sheng J. Short-Chain Fatty Acids Produced by Ruminococcaceae Mediate α-Linolenic Acid Promote Intestinal Stem Cells Proliferation. Mol Nutr Food Res 2021; 66:e2100408. [PMID: 34708542 DOI: 10.1002/mnfr.202100408] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/09/2021] [Indexed: 12/18/2022]
Abstract
SCOPE The proliferation and differentiation of intestinal stem cells (ISCs) are the basis of intestinal renewal and regeneration, and gut microbiota plays an important role in it. Dietary nutrition has the effect of regulating the activity of ISCs; however, the regulation effect of α-linolenic acid (ALA) has seldom been reported. METHODS AND RESULTS After intervening mice with different doses of ALA for 30 days, it is found that ALA (0.5 g kg-1 ) promotes small intestinal and villus growth by activating the Wnt/β-catenin signaling pathway to stimulate the proliferation of ISCs. Furthermore, ALA administration increases the abundance of the Ruminococcaceae and Prevotellaceae, and promotes the production of short-chain fatty acids (SCFAs). Subsequent fecal transplantation and antibiotic experiments demonstrate that ALA on the proliferation of ISCs are gut microbiota dependent, among them, the functional microorganism may be derived from Ruminococcaceae. Administration of isobutyrate shows a similar effect to ALA in terms of promoting ISCs proliferation. Furthermore, ALA mitigates 5-fluorouracil-induced intestinal mucosal damage by promoting ISCs proliferation. CONCLUSION These results indicate that SCFAs produced by Ruminococcaceae mediate ALA promote ISCs proliferation by activating the Wnt/β-catenin signaling pathway, and suggest the possibility of ALA as a prebiotic agent for the prevention and treatment of intestinal mucositis.
Collapse
Affiliation(s)
- Jing Xie
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, P. R. China.,Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, 650201, P. R. China
| | - Ling-Fei Li
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, P. R. China.,National Research and Development Professional Center for Moringa Processing Technology, Yunnan Agricultural University, Kunming, 650201, P. R. China
| | - Tian-Yi Dai
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, P. R. China.,Yunnan Provincial Key Laboratory of Biological Big Data, Yunnan Agricultural University, Kunming, 650201, P. R. China
| | - Xin Qi
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, P. R. China
| | - Yan Wang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, P. R. China
| | - Tiao-Zhen Zheng
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, P. R. China
| | - Xiao-Yu Gao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, P. R. China.,Yunnan Provincial Engineering Research Center for Edible and Medicinal Homologous Functional Food, Yunnan Agricultural University, Kunming, 650201, P. R. China
| | - Yun-Juan Zhang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, P. R. China
| | - Yu Ai
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, P. R. China
| | - Li Ma
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, P. R. China
| | - Song-Lin Chang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, P. R. China
| | - Feng-Xian Luo
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, P. R. China
| | - Yang Tian
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, P. R. China.,Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, 650201, P. R. China
| | - Jun Sheng
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, 650201, P. R. China
| |
Collapse
|
17
|
Yuan Q, Xie F, Huang W, Hu M, Yan Q, Chen Z, Zheng Y, Liu L. The review of alpha-linolenic acid: Sources, metabolism, and pharmacology. Phytother Res 2021; 36:164-188. [PMID: 34553434 DOI: 10.1002/ptr.7295] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/17/2021] [Accepted: 09/08/2021] [Indexed: 12/18/2022]
Abstract
α-linolenic acid (ALA, 18:3n-3) is a carboxylic acid composed of 18 carbon atoms and three cis double bonds, and is an essential fatty acid indispensable to the human body. This study aims to systematically review related studies on the dietary sources, metabolism, and pharmacological effects of ALA. Information on ALA was collected from the internet database PubMed, Elsevier, ResearchGate, Web of Science, Wiley Online Library, and Europe PMC using a combination of keywords including "pharmacology," "metabolism," "sources." The following findings are mainly contained. (a) ALA can only be ingested from food and then converted into eicosapentaenoic acid and docosahexaenoic acid in the body. (b) This conversion process is relatively limited and affected by many factors such as dose, gender, and disease. (c) Pharmacological research shows that ALA has the anti-metabolic syndrome, anticancer, antiinflammatory, anti-oxidant, anti-obesity, neuroprotection, and regulation of the intestinal flora properties. (d) There are the most studies that prove ALA has anti-metabolic syndrome effects, including experimental studies and clinical trials. (e) The therapeutic effect of ALA will be affected by the dosage. In short, ALA is expected to treat many diseases, but further high quality studies are needed to firmly establish the clinical efficacy of ALA.
Collapse
Affiliation(s)
- Qianghua Yuan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fan Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Huang
- Hanyuan Hospital of Traditional Chinese Medicine, Yaan, China
| | - Mei Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qilu Yan
- Hanyuan Hospital of Traditional Chinese Medicine, Yaan, China
| | - Zemou Chen
- Hanyuan Hospital of Traditional Chinese Medicine, Yaan, China
| | - Yan Zheng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
18
|
Perilla Seed Oil Alleviates Gut Dysbiosis, Intestinal Inflammation and Metabolic Disturbance in Obese-Insulin-Resistant Rats. Nutrients 2021; 13:nu13093141. [PMID: 34579018 PMCID: PMC8467704 DOI: 10.3390/nu13093141] [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: 08/15/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022] Open
Abstract
Background: High-fat diet (HFD) consumption induced gut dysbiosis, inflammation, obese-insulin resistance. Perilla seed oil (PSO) is a rich source of omega-3 polyunsaturated fatty acids with health promotional effects. However, the effects of PSO on gut microbiota/inflammation and metabolic disturbance in HFD-induced obesity have not been investigated. Therefore, we aimed to compare the effects of different doses of PSO and metformin on gut microbiota/inflammation, and metabolic parameters in HFD-fed rats. Methods: Thirty-six male Wistar rats were fed either a normal diet or an HFD for 24 weeks. At week 13, HFD-fed rats received either 50, 100, and 500 mg/kg/day of PSO or 300 mg/kg/day metformin for 12 weeks. After 24 weeks, the metabolic parameters, gut microbiota, gut barrier, inflammation, and oxidative stress were determined. Results: HFD-fed rats showed gut dysbiosis, gut barrier disruption with inflammation, increased oxidative stress, metabolic endotoxemia, and insulin resistance. Treatment with PSO and metformin not only effectively attenuated gut dysbiosis, but also improved gut barrier integrity and decreased gut inflammation. PSO also decreased oxidative stress, metabolic endotoxemia, and insulin resistance in HFD-fed rats. Metformin had greater benefits than PSO. Conclusion: PSO and metformin had the beneficial effect on attenuating gut inflammation and metabolic disturbance in obese-insulin resistance.
Collapse
|
19
|
The Interplay between Nutrition, Innate Immunity, and the Commensal Microbiota in Adaptive Intestinal Morphogenesis. Nutrients 2021; 13:nu13072198. [PMID: 34206809 PMCID: PMC8308283 DOI: 10.3390/nu13072198] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 12/15/2022] Open
Abstract
The gastrointestinal tract is a functionally and anatomically segmented organ that is colonized by microbial communities from birth. While the genetics of mouse gut development is increasingly understood, how nutritional factors and the commensal gut microbiota act in concert to shape tissue organization and morphology of this rapidly renewing organ remains enigmatic. Here, we provide an overview of embryonic mouse gut development, with a focus on the intestinal vasculature and the enteric nervous system. We review how nutrition and the gut microbiota affect the adaptation of cellular and morphologic properties of the intestine, and how these processes are interconnected with innate immunity. Furthermore, we discuss how nutritional and microbial factors impact the renewal and differentiation of the epithelial lineage, influence the adaptation of capillary networks organized in villus structures, and shape the enteric nervous system and the intestinal smooth muscle layers. Intriguingly, the anatomy of the gut shows remarkable flexibility to nutritional and microbial challenges in the adult organism.
Collapse
|
20
|
Ni Z, Ding J, Zhao Q, Cheng W, Yu J, Zhou L, Sun S, Yu C. Alpha-linolenic acid regulates the gut microbiota and the inflammatory environment in a mouse model of endometriosis. Am J Reprod Immunol 2021; 86:e13471. [PMID: 34022075 DOI: 10.1111/aji.13471] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/20/2021] [Accepted: 05/18/2021] [Indexed: 01/08/2023] Open
Abstract
PROBLEM This study aims to investigate the effects of alpha-linolenic acid (ALA) on the gut microbiota (GM) and the abdominal environment in mice with endometriosis (EMS). METHODS The effects of faecal microbiota transplantation (FMT) from EMS mice on mice treated with antibiotic cocktail were conducted. The 16S rRNA sequencing and PICRUSt software were used to detect the structure and function of GM respectively. The protein levels of Claudin 4 and ZO-2 in the intestinal wall were detected using the western blotting. The level of LPS in the abdominal cavity was detected using enzyme-linked immunosorbent assay (ELISA). The content of macrophages in the abdominal cavity was detected using flow cytometry. RESULTS The exogenous supplementation of ALA could restore the abundance of Firmicutes and Bacteroidota in EMS mice. After the ALA treatment, the abundance of 125 functional pathways and 50 abnormal enzymes related to GM in EMS mice was significantly improved (p < .05). The expression of the ZO-2 protein in the intestinal wall was decreased, and the level of LPS in the abdominal cavity was significantly increased after FMT from EMS mice (p < .05). ALA could increase the expression of the ZO-2 protein in the intestinal wall of EMS mice, reduce the level of LPS in the abdominal cavity (p < .05) and reduce the aggregation of peritoneal macrophages (p < .05). CONCLUSION Alpha-linolenic acid can improve the GM, intestinal wall barrier and abdominal inflammatory environment and reduce the level of LPS in mice with EMS.
Collapse
Affiliation(s)
- Zhexin Ni
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jie Ding
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Qianqian Zhao
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Wen Cheng
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jin Yu
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Ling Zhou
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Shuai Sun
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chaoqin Yu
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
21
|
Aoki K, Yanazawa K, Tokinoya K, Sugasawa T, Suzuki T, Yoshida Y, Nakano T, Omi N, Kawakami Y, Takekoshi K. Renalase is localized to the small intestine crypt and expressed upon the activation of NF-κB p65 in mice model of fasting-induced oxidative stress. Life Sci 2020; 267:118904. [PMID: 33338501 DOI: 10.1016/j.lfs.2020.118904] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 12/22/2022]
Abstract
AIMS Renalase expression is regulated by Nuclear Factor (NF)-κB and hypoxia inducible factor (HIF)-1α, and antioxidative stress function in renal cells were reported. However, dynamics of renalase and localizes in intestine were remain unknown. We evaluated the effects of oxidative stress on renalase expression and localization using model of fasting induced oxidative stress and Caco-2 cell, and examined the its physiological effects. MAIN METHODS 24 male mice were divided into three groups: Control (Con), 72 h fasting (Fast), and 24 h refeeding after fasting (Refeed). Jejunum and ileum were collected respectively. The structure of jejunum and ileum were observed by hematoxylin and eosin (HE) stain. The expression levels of carbonylated protein, renalase, NF-κB p65 and HIF-1α were measured by immunoblotting. Localization of renalase was observed by immunofluorescent. in vitro assay was performed using Caco-2 cell. Renalase was overexpressed using adenovirus. After that, Caco-2 cell was treated with 2 mM H2O2 for 30 min or 24 h. KEY FINDINGS Renalase was increased in Fast and it was localized in crypt. HIF-1α did not increase, but NF-κB p65 increased in Fast. Renalase overexpression protects the Caco-2 cells against H2O2 induced oxidative stress. SIGNIFICANCE Renalase was localized in crypt and increased in Fast. This increase suggested protect response to oxidative stress because undifferenced cells were localized in crypt and need to be protected. Actually, renalase protected Caco-2 cells against H2O2 induced oxidative stress. Small intestinal renalase expression was regulated by NF-κB p65 and was considered to be a defense mechanism against oxidative stress.
Collapse
Affiliation(s)
- Kai Aoki
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, 1-1-1 Tennodai, 305-8577, Japan
| | - Koki Yanazawa
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, 1-1-1 Tennodai, 305-8577, Japan
| | - Katsuyuki Tokinoya
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, 1-1-1 Tennodai, 305-8577, Japan
| | - Takehito Sugasawa
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Ibaraki 305-8577, Japan
| | - Takuji Suzuki
- Department of Food Science and Nutrition, Doshisha Women's College of Liberal Arts, Tera-machi Nishiiru, Imadegawa-dori, Kamigyo-ku, Kyoto 602-0893, Japan
| | - Yasuko Yoshida
- Department of Clinical Laboratory, Faculty of Health Sciences, Tsukuba International University, 6-20-1 Manabe, Tsuchiura, Ibaraki 300-0051, Japan
| | - Takuro Nakano
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, 1-1-1 Tennodai, 305-8577, Japan
| | - Naomi Omi
- Faculty of Health and Sport Sciences, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Yasushi Kawakami
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Ibaraki 305-8577, Japan
| | - Kazuhiro Takekoshi
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Ibaraki 305-8577, Japan.
| |
Collapse
|
22
|
Aguilar-Rojas A, Olivo-Marin JC, Guillen N. Human intestinal models to study interactions between intestine and microbes. Open Biol 2020; 10:200199. [PMID: 33081633 PMCID: PMC7653360 DOI: 10.1098/rsob.200199] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
Implementations of suitable in vitro cell culture systems of the human intestine have been essential tools in the study of the interaction among organs, commensal microbiota, pathogens and parasites. Due to the great complexity exhibited by the intestinal tissue, researchers have been developing in vitro/ex vivo systems to diminish the gap between conventional cell culture models and the human intestine. These models are able to reproduce different structures and functional aspects of the tissue. In the present review, information is recapitulated on the most used models, such as cell culture, intestinal organoids, scaffold-based three-dimensional models, and organ-on-a-chip and their use in studying the interaction between human intestine and microbes, and their advantages and limitations are also discussed.
Collapse
Affiliation(s)
- Arturo Aguilar-Rojas
- Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en Medicina Reproductiva, Unidad Médica de Alta Especialidad en Ginecología y Obstetricia No. 4 ‘Dr. Luis Castelazo Ayala’, Av. Río Magdalena No. 289, Col. Tizapán San Ángel, C.P. 01090 Ciudad de México, México
- Institut Pasteur, Unité d'Analyse d'Images Biologiques, 25 Rue du Dr Roux, 75015 Paris, France
| | - Jean-Christophe Olivo-Marin
- Institut Pasteur, Unité d'Analyse d'Images Biologiques, 25 Rue du Dr Roux, 75015 Paris, France
- Centre National de la Recherche Scientifique, UMR3691, 25 Rue du Dr Roux, 75015 Paris, France
| | - Nancy Guillen
- Institut Pasteur, Unité d'Analyse d'Images Biologiques, 25 Rue du Dr Roux, 75015 Paris, France
- Centre National de la Recherche Scientifique, ERL9195, 25 Rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
23
|
dos Santos Guilherme M, Zevallos VF, Pesi A, Stoye NM, Nguyen VTT, Radyushkin K, Schwiertz A, Schmitt U, Schuppan D, Endres K. Dietary Wheat Amylase Trypsin Inhibitors Impact Alzheimer's Disease Pathology in 5xFAD Model Mice. Int J Mol Sci 2020; 21:ijms21176288. [PMID: 32878020 PMCID: PMC7503408 DOI: 10.3390/ijms21176288] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 12/19/2022] Open
Abstract
Wheat amylase trypsin inhibitors (ATIs) represent a common dietary protein component of gluten-containing cereals (wheat, rye, and barley). They act as toll-like receptor 4 ligands, and are largely resistant to intestinal proteases, eliciting a mild inflammatory response within the intestine after oral ingestion. Importantly, nutritional ATIs exacerbated inflammatory bowel disease and features of fatty liver disease and the metabolic syndrome in mice. For Alzheimer’s disease (AD), both inflammation and altered insulin resistance are major contributing factors, impacting onset as well as progression of this devastating brain disorder in patients. In this study, we evaluated the impact of dietary ATIs on a well-known rodent model of AD (5xFAD). We assessed metabolic, behavioral, inflammatory, and microbial changes in mice consuming different dietary regimes with and without ATIs, consumed ad libitum for eight weeks. We demonstrate that ATIs, with or without a gluten matrix, had an impact on the metabolism and gut microbiota of 5xFAD mice, aggravating pathological hallmarks of AD. If these findings can be translated to patients, an ATI-depleted diet might offer an alternative therapeutic option for AD and warrants clinical intervention studies.
Collapse
Affiliation(s)
- Malena dos Santos Guilherme
- Department of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg-University, 55131 Mainz, Germany; (M.d.S.G.); (N.M.S.); (V.T.T.N.)
| | - Victor F. Zevallos
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, 55131 Mainz, Germany; (V.F.Z.); (A.P.)
- Nutrition and Food Research Group, Department of Applied and Health Sciences, University of Northumbria, Newcastle Upon Tyne NE1 8ST, UK
| | - Aline Pesi
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, 55131 Mainz, Germany; (V.F.Z.); (A.P.)
| | - Nicolai M. Stoye
- Department of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg-University, 55131 Mainz, Germany; (M.d.S.G.); (N.M.S.); (V.T.T.N.)
| | - Vu Thu Thuy Nguyen
- Department of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg-University, 55131 Mainz, Germany; (M.d.S.G.); (N.M.S.); (V.T.T.N.)
| | | | | | - Ulrich Schmitt
- Leibniz Institute for Resilience Research, 55122 Mainz, Germany; (K.R.); (U.S.)
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, 55131 Mainz, Germany; (V.F.Z.); (A.P.)
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Correspondence: (D.S.); (K.E.); Tel.: +49-6131-177356 (D.S.); +49-6131-172133 (K.E.)
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg-University, 55131 Mainz, Germany; (M.d.S.G.); (N.M.S.); (V.T.T.N.)
- Correspondence: (D.S.); (K.E.); Tel.: +49-6131-177356 (D.S.); +49-6131-172133 (K.E.)
| |
Collapse
|
24
|
Prevalence of an Intestinal ST40 Enterococcus faecalis over Other E. faecalis Strains in the Gut Environment of Mice Fed Different High Fat Diets. Int J Mol Sci 2020; 21:ijms21124330. [PMID: 32570702 PMCID: PMC7352901 DOI: 10.3390/ijms21124330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022] Open
Abstract
E. faecalis is a commensal bacterium with specific strains involved in opportunistic and nosocomial infections. Therefore, it is important to know how the strains of this species are selected in the gut. In this study, fifteen E. faecalis strains, isolated over twelve weeks from the faeces of mice fed standard chow or one of three high fat diets enriched with extra virgin olive oil, refined olive oil or butter were subjected to a genetic “Multilocus Sequence Typing” study that revealed the presence of mainly two genotypes, ST9 and ST40, the latter one prevailing at the end of the research. A V3–V5 sequence comparison of the predominant ST40 strain (12B3-5) in a metagenomic study showed that this sequence was the only E. faecalis present in the mouse cohort after twelve weeks. The strain was subjected to a comparative proteomic study with a ST9 strain by 2D electrophoresis and mass spectrometry. After comparing the results with a E. faecalis database, unshared entries were compared and 12B3-5 showed higher antimicrobial production as well as greater protection from environmental factors such as xenobiotics, oxidative stress and metabolite accumulation, which could be the reason for its ability to outcompete other possible rivals in an intestinal niche.
Collapse
|