1
|
Wang P, Wang R, Zhao W, Zhao Y, Wang D, Zhao S, Ge Z, Ma Y, Zhao X. Gut microbiota-derived 4-hydroxyphenylacetic acid from resveratrol supplementation prevents obesity through SIRT1 signaling activation. Gut Microbes 2025; 17:2446391. [PMID: 39725607 DOI: 10.1080/19490976.2024.2446391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
Resveratrol (RSV), a natural polyphenol, has been suggested to influence glucose and lipid metabolism. However, the underlying molecular mechanism of its action remains largely unknown due to its multiple biological targets and low bioavailability. In this study, we demonstrate that RSV supplementation ameliorates high-fat-diet (HFD)-induced gut microbiota dysbiosis, enhancing the abundance of anti-obesity bacterial strains such as Akkermansia, Bacteroides and Blautia. The critical role of gut microbiota in RSV-mediated anti-obesity effects was confirmed through antibiotic-induced microbiome depletion and fecal microbiota transplantation (FMT), which showed that RSV treatment effectively mitigates body weight, histopathological damage, glucose dysregulation and systematic inflammation associated with HFD. Metabolomics analysis revealed that RSV supplementation significantly increases the levels of the gut microbial flavonoid catabolite 4-hydroxyphenylacetic acid (4-HPA). Notably, 4-HPA was sufficient to reverse obesity and glucose intolerance in HFD-fed mice. Mechanistically,4-HPA treatment markedly regulates SIRT1 signaling pathways and induces the expression of beige fat and thermogenesis-specific markers in white adipose tissue (WAT). These beneficial effects of 4-HPA are partially abolished by EX527, a known SIRT1 inhibitor. Collectively, our findings indicate that RSV improve obesity through a gut microbiota-derived 4-HPA-SIRT1 axis, highlighting gut microbiota metabolites as a promising target for obesity prevention.
Collapse
Affiliation(s)
- Pan Wang
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Ruiqi Wang
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Wenting Zhao
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Yuanyuan Zhao
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Dan Wang
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Shuang Zhao
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Zhiwen Ge
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Yue Ma
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Xiaoyan Zhao
- Institute of Agri-Food Processing and Nutrition, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Beijing Key Laboratory of Agricultural Products of Fruits and Vegetables Preservation and Processing, Key Laboratory of Vegetable Postharvest Processing, Ministry of Agriculture and Rural Affairs, Beijing, China
| |
Collapse
|
2
|
Mu R, Fu Y, Li J, Xie Q, Ma W. Effects of different milk powders on the growth and intestinal flora in weaned rats: Comparison of special formula milk powder with ordinary milk powder. Food Sci Nutr 2024; 12:10448-10462. [PMID: 39723089 PMCID: PMC11666978 DOI: 10.1002/fsn3.4387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 12/28/2024] Open
Abstract
The objective of this investigation was to examine the effects of distinct dosages of infant formula and diverse formula constituents on the growth and development of weaned rats. Fifty specific pathogen-free (SPF) male Sprague-Dawley (SD) rats aged 3 weeks were divided into the basic diet group, 20% ordinary milk powder group, 20% special formula milk powder group, 30% ordinary milk powder group, and 30% special formula milk powder group randomly. After 28 days of feeding, compared with the basic diet group, the body mass and brain/body weight of rats in the 30% ordinary and special formula milk powder groups were decreased. At the Genus level, Bacteroides in the group supplemented with 20% special formula milk powder was significantly lower than that in the basic diet group, and Parabacteroides was significantly lower than that in the 20% ordinary milk powder group. Lactobacillus was significantly higher than those in the basic diet group and the 20% ordinary milk powder group, and Blautia was significantly higher than those in the basic diet group and the 20% and 30% ordinary milk powder groups, and UBA1819 was significantly higher than those in the other groups. The abundance of Parasutterella in the basic diet group was significantly higher than those in the groups supplemented with 20% ordinary milk powder, 20% special formula milk powder, and 30% ordinary milk powder. This study found that different doses and different formula components of infant milk powder could affect body mass and intestinal flora in Sprague-Dawley (SD) rats, and the addition of low-dose (20%) special formula infant milk powder can increase the beneficial bacteria in the intestinal flora of rats and may reduce the pathogenic bacteria.
Collapse
Affiliation(s)
- Ruiqi Mu
- School of Public Health, Beijing Key Laboratory of Environmental ToxicologyCapital Medical UniversityBeijingChina
| | - Yu Fu
- School of Public Health, Beijing Key Laboratory of Environmental ToxicologyCapital Medical UniversityBeijingChina
| | - Jufang Li
- Feihe Research InstituteHeilongjiang Feihe Dairy Co., Ltd.BeijingChina
| | - Qinggang Xie
- Feihe Research InstituteHeilongjiang Feihe Dairy Co., Ltd.BeijingChina
| | - Weiwei Ma
- School of Public Health, Beijing Key Laboratory of Environmental ToxicologyCapital Medical UniversityBeijingChina
| |
Collapse
|
3
|
Zeng Z, Chen M, Liu Y, Zhou Y, Liu H, Wang S, Ji Y. Role of Akkermansia muciniphila in insulin resistance. J Gastroenterol Hepatol 2024. [PMID: 39396929 DOI: 10.1111/jgh.16747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/15/2024] [Accepted: 09/11/2024] [Indexed: 10/15/2024]
Abstract
Insulin resistance (IR) is a pathogenic factor in numerous metabolic diseases. The gut microbiota plays a crucial role in maintaining the function of the intestinal barrier and overall human health, thereby influencing IR. Dysbiosis of the gut microbiota can contribute to the development of IR. Therefore, it is essential to maintain a balanced and diverse gut microbiota for optimal health. Akkermansia muciniphila, a widely present microorganism in the human intestine, has been shown to regulate gastrointestinal mucosal barrier integrity, reduce endotoxin penetration, decrease systemic inflammation levels, and improve insulin sensitivity. Reduced abundance of A. muciniphila is associated with an increased risk of IR and other metabolic diseases, highlighting its correlation with IR. Understanding the role and regulatory mechanism of A. muciniphila is crucial for comprehending IR pathogenesis and developing novel strategies for preventing and treating related metabolic disorders. Individual variations may exist in both the gut microbiota composition and its impact on IR among different individuals. Further investigation into individual differences between A. muciniphila and IR will facilitate advancements in personalized medicine by promoting tailored interventions based on the gut microbiota composition, which is a potential future direction that would optimize insulin sensitivity while preventing metabolic disease occurrence. In this review, we describe the physiological characteristics of A. muciniphila, emphasize its roles in underlying mechanisms contributing to IR pathology, and summarize how alterations in its abundance affect IR development, thereby providing valuable insights for further research on A. muciniphila, as well as new drug development targeting diabetes.
Collapse
Affiliation(s)
- Zhijun Zeng
- Jiangxi University of Chinese Medicine, Nanchang, China
- Research Center for Differentiation and Development of TCM Basic Theory, Jiangxi Province Key Laboratory of TCM Etiopathogenesis, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Mengjie Chen
- Jiangxi University of Chinese Medicine, Nanchang, China
- Research Center for Differentiation and Development of TCM Basic Theory, Jiangxi Province Key Laboratory of TCM Etiopathogenesis, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yimin Liu
- Jiangxi University of Chinese Medicine, Nanchang, China
- Research Center for Differentiation and Development of TCM Basic Theory, Jiangxi Province Key Laboratory of TCM Etiopathogenesis, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yun Zhou
- Jiangxi University of Chinese Medicine, Nanchang, China
- Research Center for Differentiation and Development of TCM Basic Theory, Jiangxi Province Key Laboratory of TCM Etiopathogenesis, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Hongning Liu
- Jiangxi University of Chinese Medicine, Nanchang, China
- Research Center for Differentiation and Development of TCM Basic Theory, Jiangxi Province Key Laboratory of TCM Etiopathogenesis, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Shaohua Wang
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yanhua Ji
- Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
4
|
Lin T, Daddi L, Tang Y, Zhou Y, Liu B, Moore MD, Liu Z. Antrodia camphorata Supplementation during Early Life Alters Gut Microbiota and Inhibits Young-Onset Intestinal Tumorigenesis in APC1638N Mice Later in Life. Nutrients 2024; 16:2408. [PMID: 39125288 PMCID: PMC11314251 DOI: 10.3390/nu16152408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Young-onset colorectal cancer is an increasing concern worldwide due to the growing prevalence of Westernized lifestyles in childhood and adolescence. Environmental factors during early life, particularly early-life nutrition, significantly contribute to the increasing incidence. Recently, there have been reports of beneficial effects, including anti-inflammation and anti-cancer, of a unique fungus (Antrodia camphorate, AC) native to Taiwan. The objective of this study is to investigate the impact of AC supplementation in early life on the development of young-onset intestinal tumorigenesis. APC1638N mice were fed with a high-fat diet (HF) at 4-12 weeks of age, which is equivalent to human childhood/adolescence, before switching to a normal maintenance diet for an additional 12 weeks up to 24 weeks of age, which is equivalent to young to middle adulthood in humans. Our results showed that the body weight in the HF groups significantly increased after 8 weeks of feeding (p < 0.05). Following a switch to a normal maintenance diet, the change in body weight persisted. AC supplementation significantly suppressed tumor incidence and multiplicity in females (p < 0.05) and reduced IGF-1 and Wnt/β-catenin signaling (p < 0.05). Moreover, it altered the gut microbiota, suppressed inflammatory responses, and created a microenvironment towards suppressing tumorigenesis later in life.
Collapse
Affiliation(s)
- Tingchun Lin
- Department of Nutrition, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA 01003, USA; (T.L.); (Y.T.); (B.L.)
| | - Lauren Daddi
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA; (L.D.); (Y.Z.)
| | - Ying Tang
- Department of Nutrition, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA 01003, USA; (T.L.); (Y.T.); (B.L.)
| | - Yanjiao Zhou
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA; (L.D.); (Y.Z.)
| | - Buping Liu
- Department of Nutrition, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA 01003, USA; (T.L.); (Y.T.); (B.L.)
- School of Public Health and Management, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Matthew D. Moore
- Department of Food Science, University of Massachusetts Amherst, Amherst, MA 01003, USA;
| | - Zhenhua Liu
- Department of Nutrition, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA 01003, USA; (T.L.); (Y.T.); (B.L.)
- UMass Cancer Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| |
Collapse
|
5
|
Lee H, Song J, Lee B, Cha J, Lee H. Food carbohydrates in the gut: structural diversity, microbial utilization, and analytical strategies. Food Sci Biotechnol 2024; 33:2123-2140. [PMID: 39130670 PMCID: PMC11315866 DOI: 10.1007/s10068-024-01648-3] [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: 04/17/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 08/13/2024] Open
Abstract
Carbohydrates, which are a vital dietary component, undergo digestion and gut fermentation through microbial enzymes to produce beneficial short-chain fatty acids. Certain carbohydrates selectively modulate the gut microbiota, impacting host health. Carbohydrate-active enzymes within the gut microbiota significantly contribute to carbohydrate utilization and microbial diversity. Despite their importance, the structural complexity of carbohydrates poses analytical challenges. However, recent advancements, notably, mass spectrometry, have allowed for their characterization and functional analysis. This review examines the intricate relationship between dietary carbohydrates and the gut microbiota, highlighting the crucial role of advanced analytical techniques in understanding their diversity and implications. These advancements provide valuable insights into carbohydrate bioactivity. Integrating high-throughput analysis with next-generation sequencing provides deeper insights into gut microbial interactions, potentially revealing which carbohydrate structures are beneficial for gut health.
Collapse
Affiliation(s)
- HyunJi Lee
- Department of Applied Chemistry, Food Science and Technology, Dong-eui University, Busan, 47340 Republic of Korea
| | - JaeHui Song
- Department of Applied Chemistry, Food Science and Technology, Dong-eui University, Busan, 47340 Republic of Korea
| | - Bokyung Lee
- Department of Food Science and Nutrition, Dong-A University, Busan, 49315 Republic of Korea
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, 49315 Republic of Korea
| | - Jaeho Cha
- Department of Microbiology, Pusan National University, Busan, 46241 Republic of Korea
- Microbiological Resources Research Institute, Pusan National University, Busan, 46241 Republic of Korea
| | - Hyeyoung Lee
- Department of Applied Chemistry, Food Science and Technology, Dong-eui University, Busan, 47340 Republic of Korea
| |
Collapse
|
6
|
Zhang J, Hu B, Deng X, Sun R, Zhang R, Chen K, Guo W. Multiomics analysis investigating the impact of a high-fat diet in female Sprague-Dawley rats: alterations in plasma, intestinal metabolism, and microbial composition. Front Nutr 2024; 11:1359989. [PMID: 38646105 PMCID: PMC11026666 DOI: 10.3389/fnut.2024.1359989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/20/2024] [Indexed: 04/23/2024] Open
Abstract
Introduction With improvements in living conditions, modern individuals exhibit a pronounced inclination towards a high-fat diet, largely because of its distinctive gustatory appeal. However, the association between high-fat diets and metabolic complications has largely been ignored, and metabolic diseases such as obesity and non-alcoholic fatty liver disease now constitute a major public health concern. Because high-fat diets increase the risk of metabolic diseases, a thorough investigation into the impact of high-fat diets on gut microbiota and metabolism is required. Methods We utilize 16S rRNA sequencing and untargeted metabolomics analysis to demonstrate that SD rats fed a high-fat diet exhibited marked alterations in gut microbiota and plasma, intestinal metabolism. Results Changes in gut microbiota included a decreased abundance at phylum level for Verrucomicrobiota, and a decreased abundance at genus level for Akkermansia, Ralstonia, Bacteroides, and Faecalibacterium. Additionally, significant changes were observed in both intestinal and plasma metabolite levels, including an upregulation of bile acid metabolism, an upregulation of glucose-lipid metabolism, and increased levels of metabolites such as norlithocholic acid, cholic acid, D-fructose, D-mannose, fructose lactate, and glycerophosphocholine. We also investigated the correlations between microbial communities and metabolites, revealing a significant negative correlation between Akkermansia bacteria and cholic acid. Discussion Overall, our findings shed light on the relationship between symbiotic bacteria associated with high-fat diets and metabolic biomarkers, and they provide insights for identifying novel therapeutic approaches to mitigate disease risks associated with a high-fat diet.
Collapse
Affiliation(s)
- Jiacheng Zhang
- Department of Hepatobiliary, Pancreatic and Liver Transplantation Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
- Open and Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, China
| | - Binhong Hu
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
| | - Xin Deng
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
| | - Rong Sun
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
| | - Rong Zhang
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Kuo Chen
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenzhi Guo
- Department of Hepatobiliary, Pancreatic and Liver Transplantation Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Open and Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, China
- Henan Research Centre for Organ Transplantation, Zhengzhou, China
| |
Collapse
|
7
|
Bai J, Eldridge R, Houser M, Martin M, Powell C, Sutton KS, Noh HI, Wu Y, Olson T, Konstantinidis KT, Bruner DW. Multi-omics analysis of the gut microbiome and metabolites associated with the psychoneurological symptom cluster in children with cancer receiving chemotherapy. J Transl Med 2024; 22:256. [PMID: 38461265 PMCID: PMC10924342 DOI: 10.1186/s12967-024-05066-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/05/2024] [Indexed: 03/11/2024] Open
Abstract
BACKGROUND Children with cancer receiving chemotherapy commonly report a cluster of psychoneurological symptoms (PNS), including pain, fatigue, anxiety, depression, and cognitive dysfunction. The role of the gut microbiome and its functional metabolites in PNS is rarely studied among children with cancer. This study investigated the associations between the gut microbiome-metabolome pathways and PNS in children with cancer across chemotherapy as compared to healthy children. METHODS A case-control study was conducted. Cancer cases were recruited from Children's Healthcare of Atlanta and healthy controls were recruited via flyers. Participants reported PNS using the Pediatric Patient-Reported Outcomes Measurement Information System. Data for cases were collected pre-cycle two chemotherapy (T0) and post-chemotherapy (T1), whereas data for healthy controls were collected once. Gut microbiome and its metabolites were measured using fecal specimens. Gut microbiome profiling was performed using 16S rRNA V4 sequencing, and metabolome was performed using an untargeted liquid chromatography-mass spectrometry approach. A multi-omics network integration program analyzed microbiome-metabolome pathways of PNS. RESULTS Cases (n = 21) and controls (n = 14) had mean ages of 13.2 and 13.1 years. For cases at T0, PNS were significantly associated with microbial genera (e.g., Ruminococcus, Megasphaera, and Prevotella), which were linked with carnitine shuttle (p = 0.0003), fatty acid metabolism (p = 0.001) and activation (p = 0.001), and tryptophan metabolism (p = 0.008). Megasphaera, clustered with aspartate and asparagine metabolism (p = 0.034), carnitine shuttle (p = 0.002), and tryptophan (p = 0.019), was associated with PNS for cases at T1. Gut bacteria with potential probiotic functions, along with fatty acid metabolism, tryptophan, and carnitine shuttle, were more clustered in cancer cases than the control network and this linkage with PNS needs further studies. CONCLUSIONS Using multi-omics approaches, this study indicated specific microbiome-metabolome pathways linked with PNS in children with cancer across chemotherapy. Due to limitations such as antibiotic use in cancer cases, these findings need to be further confirmed in a larger cohort.
Collapse
Affiliation(s)
- Jinbing Bai
- Nell Hodgson Woodruff School of Nursing, Emory University, 1520 Clifton Road NE, Atlanta, GA, 30322, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| | - Ronald Eldridge
- Nell Hodgson Woodruff School of Nursing, Emory University, 1520 Clifton Road NE, Atlanta, GA, 30322, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Madelyn Houser
- Nell Hodgson Woodruff School of Nursing, Emory University, 1520 Clifton Road NE, Atlanta, GA, 30322, USA
| | - Melissa Martin
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Christie Powell
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Kathryn S Sutton
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- School of Medicine, Emory University, Atlanta, GA, USA
| | - Hye In Noh
- Nell Hodgson Woodruff School of Nursing, Emory University, 1520 Clifton Road NE, Atlanta, GA, 30322, USA
| | - Yuhua Wu
- Nell Hodgson Woodruff School of Nursing, Emory University, 1520 Clifton Road NE, Atlanta, GA, 30322, USA
| | - Thomas Olson
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- School of Medicine, Emory University, Atlanta, GA, USA
| | | | - Deborah W Bruner
- Nell Hodgson Woodruff School of Nursing, Emory University, 1520 Clifton Road NE, Atlanta, GA, 30322, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
8
|
Wang Y, Hu Y, Niu Z, Zhang X, Fan D, Ji X, Lv H, Wang S, Zhao Y. Immunomodulation of nutritional formula containing epigallocatechin-3-gallate, ginseng extract, and polydextrose on inflammation and macrophage polarization. Front Nutr 2024; 11:1370608. [PMID: 38445210 PMCID: PMC10912162 DOI: 10.3389/fnut.2024.1370608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 02/05/2024] [Indexed: 03/07/2024] Open
Abstract
Single nutrient likes polyphenol or dietary fiber have been exhaustively investigated to validate their positive intervention in health or disease. Meanwhile, the common interaction of inner systems with the nutrient complex has not been well elucidated, which raises the scientific issue of the modulatory effect of the nutrient complex on immunity. The representative prebiotics of epigallocatechin-3-gallate (EGCG), ginseng extract, and polydextrose (PDX) were selected on behalf of the classification of polyphenol, flavone or polysaccharides, and dietary fiber to generally cover the daily food intake in this study to explore their intervention in inflammation and macrophage polarization. The intervention of selected nutrients on inflammation and macrophage polarization has been evaluated against macrophages to unveil their comprehensive effects. The synergistic effect of selected nutrients was demonstrated by inhibiting M1 macrophage polarization and the promotion of M2 macrophage polarization. Then, the nutrient formula was set up to verify the intervention effect, and the results revealed the significant inhibition of cell inflammation and the effect on cell proliferation through promoting the cell cycle in the G2 phase. The nutrient complex could inhibit M1 macrophage polarization to inhibit M1-mediated inflammation and promote M2 macrophages for anti-inflammatory effect and enhance cell phagocytosis. Moreover, the varied intervention effects of the nutrient complex with different formulas could be summarized. In general, the formula containing EGCG, ginseng extract, and PDX was demonstrated to possess an enhanced immunomodulatory effect on cell inflammation and macrophage polarization, which could potentially inspire the investigation of complex nutrients in health and diseases.
Collapse
Affiliation(s)
- Yi Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin, China
| | - Yaozhong Hu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin, China
| | - Zhenhua Niu
- Shanghai M-Action Health Technology Co., Ltd., Shanghai, China
| | - Xuejiao Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin, China
| | - Dancai Fan
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin, China
| | - Xuemeng Ji
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin, China
| | - Huan Lv
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin, China
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin, China
| | - Yanrong Zhao
- Shanghai M-Action Health Technology Co., Ltd., Shanghai, China
| |
Collapse
|
9
|
Xu C, Jiang H, Feng LJ, Jiang MZ, Wang YL, Liu SJ. Christensenella minuta interacts with multiple gut bacteria. Front Microbiol 2024; 15:1301073. [PMID: 38440147 PMCID: PMC10910051 DOI: 10.3389/fmicb.2024.1301073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 01/30/2024] [Indexed: 03/06/2024] Open
Abstract
Introduction Gut microbes form complex networks that significantly influence host health and disease treatment. Interventions with the probiotic bacteria on the gut microbiota have been demonstrated to improve host well-being. As a representative of next-generation probiotics, Christensenella minuta (C. minuta) plays a critical role in regulating energy balance and metabolic homeostasis in human bodies, showing potential in treating metabolic disorders and reducing inflammation. However, interactions of C. minuta with the members of the networked gut microbiota have rarely been explored. Methods In this study, we investigated the impact of C. minuta on fecal microbiota via metagenomic sequencing, focusing on retrieving bacterial strains and coculture assays of C. minuta with associated microbial partners. Results Our results showed that C. minuta intervention significantly reduced the diversity of fecal microorganisms, but specifically enhanced some groups of bacteria, such as Lactobacillaceae. C. minuta selectively enriched bacterial pathways that compensated for its metabolic defects on vitamin B1, B12, serine, and glutamate synthesis. Meanwhile, C. minuta cross-feeds Faecalibacterium prausnitzii and other bacteria via the production of arginine, branched-chain amino acids, fumaric acids and short-chain fatty acids (SCFAs), such as acetic. Both metagenomic data analysis and culture experiments revealed that C. minuta negatively correlated with Klebsiella pneumoniae and 14 other bacterial taxa, while positively correlated with F. prausnitzii. Our results advance our comprehension of C. minuta's in modulating the gut microbial network. Conclusions C. minuta disrupts the composition of the fecal microbiota. This disturbance is manifested through cross-feeding, nutritional competition, and supplementation of its own metabolic deficiencies, resulting in the specific enrichment or inhibition of the growth of certain bacteria. This study will shed light on the application of C. minuta as a probiotic for effective interventions on gut microbiomes and improvement of host health.
Collapse
Affiliation(s)
- Chang Xu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - He Jiang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Li-Juan Feng
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Min-Zhi Jiang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Yu-Lin Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Shuang-Jiang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
10
|
Langhi C, Vallier M, Bron A, Otero YF, Maura M, Le Joubioux F, Blomberg N, Giera M, Guigas B, Maugard T, Chassaing B, Peltier S, Blanquet-Diot S, Bard JM, Sirvent P. A polyphenol-rich plant extract prevents hypercholesterolemia and modulates gut microbiota in western diet-fed mice. Front Cardiovasc Med 2024; 11:1342388. [PMID: 38317864 PMCID: PMC10839041 DOI: 10.3389/fcvm.2024.1342388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/03/2024] [Indexed: 02/07/2024] Open
Abstract
Introduction Totum-070 is a combination of five plant extracts enriched in polyphenols to target hypercholesterolemia, one of the main risk factors for cardiovascular diseases. The aim of this study was to investigate the effects of Totum-070 on cholesterol levels in an animal model of diet-induced hypercholesterolemia. Methods C57BL/6JOlaHsd male mice were fed a Western diet and received Totum-070, or not, by daily gavage (1g/kg and 3g/kg body weight) for 6 weeks. Results The Western diet induced obesity, fat accumulation, hepatic steatosis and increased plasma cholesterol compared with the control group. All these metabolic perturbations were alleviated by Totum-070 supplementation in a dose-dependent manner. Lipid excretion in feces was higher in mice supplemented with Totum-070, suggesting inhibition of intestinal lipid absorption. Totum-070 also increased the fecal concentration of short chain fatty acids, demonstrating a direct effect on intestinal microbiota. Discussion The characterization of fecal microbiota by 16S amplicon sequencing showed that Totum-070 supplementation modulated the dysbiosis associated with metabolic disorders. Specifically, Totum-070 increased the relative abundance of Muribaculum (a beneficial bacterium) and reduced that of Lactococcus (a genus positively correlated with increased plasma cholesterol level). Together, these findings indicate that the cholesterol-lowering effect of Totum-070 bioactive molecules could be mediated through multiple actions on the intestine and gut microbiota.
Collapse
Affiliation(s)
| | | | - Auriane Bron
- UMR 454 Microbiologie Environnement DIgestif et Santé (MEDIS), Université Clermont Auvergne, Clermont-Ferrand, France
| | | | | | | | - Niek Blomberg
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Thierry Maugard
- Equipe BCBS (Biotechnologies et Chimie des Bioressources Pour la Santé), UMR CNRS 7266 LIENSs, La Rochelle Université, La Rochelle, France
| | - Benoit Chassaing
- Team “Mucosal Microbiota in Chronic Inflammatory Diseases”, Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Cité, Paris, France
| | | | - Stéphanie Blanquet-Diot
- UMR 454 Microbiologie Environnement DIgestif et Santé (MEDIS), Université Clermont Auvergne, Clermont-Ferrand, France
| | - Jean-Marie Bard
- Laboratoire de Biochimie Générale et Appliquée, UFR de Pharmacie, ISOMer-UE 2160, IUML-Institut Universitaire Mer et Littoral-FR3473 CNRS, Université de Nantes, Nantes, France
| | | |
Collapse
|
11
|
Lerma-Aguilera AM, Pérez-Burillo S, Navajas-Porras B, León ED, Ruíz-Pérez S, Pastoriza S, Jiménez-Hernández N, Cämmerer BM, Rufián-Henares JÁ, Gosalbes MJ, Francino MP. Effects of different foods and cooking methods on the gut microbiota: an in vitro approach. Front Microbiol 2024; 14:1334623. [PMID: 38260868 PMCID: PMC10800916 DOI: 10.3389/fmicb.2023.1334623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024] Open
Abstract
To support personalized diets targeting the gut microbiota, we employed an in vitro digestion-fermentation model and 16S rRNA gene sequencing to analyze the microbiota growing on representative foods of the Mediterranean and Western diets, as well as the influence of cooking methods. Plant- and animal-derived foods had significantly different impacts on the abundances of bacterial taxa. Animal and vegetable fats, fish and dairy products led to increases in many taxa, mainly within the Lachnospiraceae. In particular, fats favored increases in the beneficial bacteria Faecalibacterium, Blautia, and Roseburia. However, butter, as well as gouda cheese and fish, also resulted in the increase of Lachnoclostridium, associated to several diseases. Frying and boiling produced the most distinct effects on the microbiota, with members of the Lachnospiraceae and Ruminococcaceae responding the most to the cooking method employed. Nevertheless, cooking effects were highly individualized and food-dependent, challenging the investigation of their role in personalized diets.
Collapse
Affiliation(s)
- Alberto M. Lerma-Aguilera
- Área de Genómica y Salud, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana-Salud Pública, Valencia, Spain
| | - Sergio Pérez-Burillo
- Departamento de Nutrición y Bromatología, Centro de Investigación Biomédica, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Granada, Granada, Spain
| | - Beatriz Navajas-Porras
- Departamento de Nutrición y Bromatología, Centro de Investigación Biomédica, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Granada, Granada, Spain
| | - E. Daniel León
- Área de Genómica y Salud, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana-Salud Pública, Valencia, Spain
| | - Sonia Ruíz-Pérez
- Área de Genómica y Salud, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana-Salud Pública, Valencia, Spain
| | - Silvia Pastoriza
- Departamento de Nutrición y Bromatología, Centro de Investigación Biomédica, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Universidad de Granada, Granada, Spain
| | - Nuria Jiménez-Hernández
- Área de Genómica y Salud, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana-Salud Pública, Valencia, Spain
- CIBER Epidemiología y Salud Pública, Instituto de Salud Carlos III, Madrid, Spain
| | - Bettina-Maria Cämmerer
- Department of Food Chemistry and Analytics, Technische Universität Berlin, Berlin, Germany
| | - José Ángel Rufián-Henares
- Departamento de Nutrición y Bromatología, Centro de Investigación Biomédica, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria (ibs.GRANADA), Universidad de Granada, Granada, Spain
| | - María José Gosalbes
- Área de Genómica y Salud, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana-Salud Pública, Valencia, Spain
- CIBER Epidemiología y Salud Pública, Instituto de Salud Carlos III, Madrid, Spain
| | - M. Pilar Francino
- Área de Genómica y Salud, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana-Salud Pública, Valencia, Spain
- CIBER Epidemiología y Salud Pública, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
12
|
Su X, Chen S, Liu J, Feng Y, Han E, Hao X, Liao M, Cai J, Zhang S, Niu J, He S, Huang S, Lo K, Zeng F. Composition of gut microbiota and non-alcoholic fatty liver disease: A systematic review and meta-analysis. Obes Rev 2024; 25:e13646. [PMID: 37813400 DOI: 10.1111/obr.13646] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 08/03/2023] [Accepted: 09/09/2023] [Indexed: 10/11/2023]
Abstract
The present systematic review and meta-analysis aimed to summarize the associations between gut microbiota composition and non-alcoholic fatty liver disease. To compare the differences between individuals with or without NAFLD, the standardized mean difference and 95% confidence interval were computed for each α-diversity index and relative abundance of gut microbes. The β-diversity indices were summarized in a qualitative manner. A total of 54 studies with 8894 participants were included. Overall, patients with NAFLD had moderate reduction in α-diversity indices including Shannon (SMD = -0.36, 95% CI = [-0.53, -0.19], p < 0.001) and Chao 1 (SMD = -0.42, 95% CI = [-0.68, -0.17], p = 0.001), but no significant differences were found for Simpson, observed species, phylogenetic diversity, richness, abundance-based coverage estimator, and evenness (p ranged from 0.081 to 0.953). Over 75% of the included studies reported significant differences in β-diversity. Although there was substantial interstudy heterogeneity, especially for analyses at the phylum, class, and family levels, the majority of the included studies showed alterations in the depletion of anti-inflammatory microbes (i.e., Ruminococcaceae and Coprococcus) and the enrichment of proinflammatory microbes (i.e., Fusobacterium and Escherichia) in patients with NAFLD. Perturbations in gut microbiota were associated with NAFLD, commonly reflected by a reduction in beneficial species and an increase in the pathogenic species.
Collapse
Affiliation(s)
- Xin Su
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Shiyun Chen
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Jiazi Liu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Yonghui Feng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Eerdun Han
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xiaolei Hao
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Minqi Liao
- Helmholtz Zentrum München-German Research Center for Environmental Health, Institute of Epidemiology, Neuherberg, PR, Germany
| | - Jun Cai
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Shiwen Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Jianxiang Niu
- General Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Shihua He
- Department of Infectious Disease, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Shaofen Huang
- Shenzhen Qianhai Shekou Free Zone Hospital, Shenzhen, China
| | - Kenneth Lo
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Research Institute for Future Food, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Fangfang Zeng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Kong G, Zhang W, Zhang S, Chen J, He K, Zhang C, Yuan X, Xie B. The gut microbiota and metabolite profiles are altered in patients with spinal cord injury. Mol Brain 2023; 16:26. [PMID: 36803646 PMCID: PMC9940348 DOI: 10.1186/s13041-023-01014-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 02/05/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Metabolites secreted by the gut microbiota may play an essential role in microbiota-gut-central nervous system crosstalk. In this study, we explored the changes occurring in the gut microbiota and their metabolites in patients with spinal cord injury (SCI) and analyzed the correlations among them. METHODS The structure and composition of the gut microbiota derived from fecal samples collected from patients with SCI (n = 11) and matched control individuals (n = 10) were evaluated using 16S rRNA gene sequencing. Additionally, an untargeted metabolomics approach was used to compare the serum metabolite profiles of both groups. Meanwhile, the association among serum metabolites, the gut microbiota, and clinical parameters (including injury duration and neurological grade) was also analyzed. Finally, metabolites with the potential for use in the treatment of SCI were identified based on the differential metabolite abundance analysis. RESULTS The composition of the gut microbiota was different between patients with SCI and healthy controls. At the genus level, compared with the control group, the abundance of UBA1819, Anaerostignum, Eggerthella, and Enterococcus was significantly increased in the SCI group, whereas that of Faecalibacterium, Blautia, Escherichia-Shigella, Agathobacter, Collinsella, Dorea, Ruminococcus, Fusicatenibacter, and Eubacterium was decreased. Forty-one named metabolites displayed significant differential abundance between SCI patients and healthy controls, including 18 that were upregulated and 23 that were downregulated. Correlation analysis further indicated that the variation in gut microbiota abundance was associated with changes in serum metabolite levels, suggesting that gut dysbiosis is an important cause of metabolic disorders in SCI. Finally, gut dysbiosis and serum metabolite dysregulation was found to be associated with injury duration and severity of motor dysfunction after SCI. CONCLUSIONS We present a comprehensive landscape of the gut microbiota and metabolite profiles in patients with SCI and provide evidence that their interaction plays a role in the pathogenesis of SCI. Furthermore, our findings suggested that uridine, hypoxanthine, PC(18:2/0:0), and kojic acid may be important therapeutic targets for the treatment of this condition.
Collapse
Affiliation(s)
- Ganggang Kong
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wenwu Zhang
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Siyun Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jiewen Chen
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Kejun He
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, Zhong Shan Er Lu, Guangzhou, 510080, Guangdong, China
| | - Changming Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, Zhong Shan Er Lu, Guangzhou, 510080, Guangdong, China
| | - Xi Yuan
- Department of Anesthesiology, Bazhong Central Hospital, Bazhong, China
| | - Baoshu Xie
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, Zhong Shan Er Lu, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
14
|
Cao F, Ding Q, Zhuge H, Lai S, Chang K, Le C, Yang G, Valencak TG, Li S, Ren D. Lactobacillus plantarum ZJUIDS14 alleviates non-alcoholic fatty liver disease in mice in association with modulation in the gut microbiota. Front Nutr 2023; 9:1071284. [PMID: 36698477 PMCID: PMC9868733 DOI: 10.3389/fnut.2022.1071284] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/23/2022] [Indexed: 01/11/2023] Open
Abstract
This present study was designed to explore the protective role of Lactobacillus plantarum ZJUIDS14 against Non-alcoholic Fatty Liver Disease (NAFLD) in a high-fat-diet (HFD)-induced C57BL/6 mice model. The probiotic (109 CFU/every other day) was administered by oral gavage for 12 weeks. We found that L. plantarum ZJUIDS14 intervention significantly alleviated HFD related hepatic steatosis, liver damage, insulin resistance, and increased hepatic expression of peroxisome proliferator activated receptor α (PPAR-α) while stimulating the activation of AMP-activated protein kinase (AMPK). Furthermore, L. plantarum ZJUIDS14 improved mitochondrial function as reflected by an increase in dynamin related protein 1 (DRP1) and a decrease of proteins associated with oxidative phosphorylation (OXPHOS) after the treatment. Additionally, mice from the L. plantarum ZJUIDS14 group had a restored intestinal flora and homeostasis involving Coprostanoligenes group, Ruminococcaceae UCG-014, Allobaculum, Ruminiclostridium 1, and Roseburia. Meanwhile, these five genera exhibited a significant (negative or positive) association with ileum inflammation mRNA levels and SCFA contents, by Spearman's correlation analysis. In general, our data demonstrated that L. plantarum ZJUIDS14 mitigates hepatic steatosis and liver damage induced by HFD. Specifically, they strengthened the integrity of the intestinal barrier, regulated gut microbiota, and improved mitochondrial function. Our data provide an experimental basis for L. plantarum ZJUIDS14 as a promising candidate to prevent NAFLD.
Collapse
Affiliation(s)
- Feiwei Cao
- College of Animal Sciences, Institute of Dairy Science, Zhejiang University, Hangzhou, China,School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qinchao Ding
- College of Animal Sciences, Institute of Dairy Science, Zhejiang University, Hangzhou, China,School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hui Zhuge
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shanglei Lai
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Kaixin Chang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chunyan Le
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Guorong Yang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Teresa G. Valencak
- College of Animal Sciences, Institute of Dairy Science, Zhejiang University, Hangzhou, China
| | - Songtao Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China,*Correspondence: Songtao Li,
| | - Daxi Ren
- College of Animal Sciences, Institute of Dairy Science, Zhejiang University, Hangzhou, China,Daxi Ren,
| |
Collapse
|
15
|
Nagasawa Y, Katagiri S, Nakagawa K, Hirota T, Yoshimi K, Uchida A, Hatasa M, Komatsu K, Shiba T, Ohsugi Y, Uesaka N, Iwata T, Tohara H. Xanthan gum-based fluid thickener decreases postprandial blood glucose associated with increase of Glp1 and Glp1r expression in ileum and alteration of gut microbiome. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
16
|
The Entero-Mammary Pathway and Perinatal Transmission of Gut Microbiota and SARS-CoV-2. Int J Mol Sci 2022; 23:ijms231810306. [PMID: 36142219 PMCID: PMC9499685 DOI: 10.3390/ijms231810306] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/02/2022] [Accepted: 09/04/2022] [Indexed: 02/08/2023] Open
Abstract
COVID-19 is a severe respiratory disease threatening pregnant women, which increases the possibility of adverse pregnancy outcomes. Several recent studies have demonstrated the ability of SARS-CoV-2 to infect the mother enterocytes, disturbing the gut microbiota diversity. The aim of this study was to characterize the entero-mammary microbiota of women in the presence of the virus during delivery. Fifty mother−neonate pairs were included in a transversal descriptive work. The presence of SARS-CoV-2 RNA was detected in nasopharyngeal, mother rectal swabs (MRS) and neonate rectal swabs (NRS) collected from the pairs, and human colostrum (HC) samples collected from mothers. The microbiota diversity was characterized by high-throughput DNA sequencing of V3-16S rRNA gene libraries prepared from HC, MRS, and NRS. Data were analyzed with QIIME2 and R. Our results indicate that several bacterial taxa are highly abundant in MRS positive for SARS-CoV-2 RNA. These bacteria mostly belong to the Firmicutes phylum; for instance, the families Bifidobacteriaceae, Oscillospiraceae, and Microbacteriaceae have been previously associated with anti-inflammatory effects, which could explain the capability of women to overcome the infection. All samples, both positive and negative for SARS-CoV-2, featured a high abundance of the Firmicutes phylum. Further data analysis showed that nearly 20% of the bacterial diversity found in HC was also identified in MRS. Spearman correlation analysis highlighted that some genera of the Proteobacteria and Actinobacteria phyla were negatively correlated with MRS and NRS (p < 0.005). This study provides new insights into the gut microbiota of pregnant women and their potential association with a better outcome during SARS-CoV-2 infection.
Collapse
|
17
|
Zhou C, Zhang W, Lin H, Zhang L, Wu F, Wang Y, Yu S, Peng X, Cheng W, Li M, Pan X, Huang Z, Zhang W. Effect of theaflavin-3,3′-digallate on leptin-deficient induced nonalcoholic fatty liver disease might be related to lipid metabolism regulated by the Fads1/PPARδ/Fabp4 axis and gut microbiota. Front Pharmacol 2022; 13:925264. [PMID: 36105184 PMCID: PMC9464872 DOI: 10.3389/fphar.2022.925264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), one of the risk factors for hepatitis, cirrhosis, and even hepatic carcinoma, has been a global public health problem. The polyphenol compound theaflavin-3,3′-digallate (TF3), mainly extracted from black tea, has been reported to produce an effect on hypoglycemic and antilipid deposition in vitro. In our study, we further investigated the function and novel mechanisms of TF3 in protecting NAFLD in vivo. By using leptin-deficient obese (ob/ob) mice with NAFLD symptoms, TF3 treatment prevented body weight and waistline gain, reduced lipid accumulation, and alleviated liver function injury, as well as decreased serum lipid levels and TG levels in livers in ob/ob mice, observing no side effects. Furthermore, the transcriptome sequencing of liver tissue showed that TF3 treatment corrected the expression profiles of livers in ob/ob mice compared with that of the model group. It is interesting to note that TF3 might regulate lipid metabolism via the Fads1/PPARδ/Fabp4 axis. In addition, 16S rRNA sequencing demonstrated that TF3 increased the abundance of Prevotellaceae_UCG-001, norank_f_Ruminococcaceae, and GCA-900066575 and significantly decreased that of Parvibacter. Taken together, the effect of TF3 on NAFLD might be related to lipid metabolism regulated by the Fads1/PPARδ/Fabp4 axis and gut microbiota. TF3 might be a promising candidate for NAFLD therapy.
Collapse
Affiliation(s)
- Cheng Zhou
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Wenji Zhang
- Guangdong Provincial Engineering & Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Hui Lin
- Department of Radiation Oncology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Luyun Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Fan Wu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Yan Wang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Susu Yu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Xinyue Peng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Wenli Cheng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Min Li
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Xiaoying Pan
- Guangdong Provincial Engineering & Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Zhenrui Huang
- Guangdong Provincial Engineering & Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- *Correspondence: Zhenrui Huang, ; Wenjuan Zhang,
| | - Wenjuan Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
- *Correspondence: Zhenrui Huang, ; Wenjuan Zhang,
| |
Collapse
|
18
|
Milton-Laskibar I, Cuevas-Sierra A, Portillo MP, Martínez JA. Effects of Resveratrol Administration in Liver Injury Prevention as Induced by an Obesogenic Diet: Role of Ruminococcaceae. Biomedicines 2022; 10:biomedicines10081797. [PMID: 35892696 PMCID: PMC9330856 DOI: 10.3390/biomedicines10081797] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 12/14/2022] Open
Abstract
Gut microbiota dysbiosis has been described in several metabolic disruptions, such as non-alcoholic fatty liver disease (NAFLD). Administration of resveratrol has been claimed to elicit benefits against NAFLD along with modulating gut microbiota composition. This investigation aims to study the putative mediating role of gut microbiota in the potential hepato-protective effects of resveratrol in a diet-induced NAFLD rat model. The involvement of bacteria from the Ruminococcaceae family in such effects was also addressed. Resveratrol administration resulted in lowered liver weight and serum total and non-HDL cholesterol concentrations, as well as in increased serum HDL cholesterol levels. The administration of this polyphenol also prevented obesogenic diet-induced serum transaminase increases. In addition, histopathological analysis revealed that resveratrol administration ameliorated the dietary-induced liver steatosis and hepatic inflammation. Gut microbiota sequencing showed an inverse relationship between some bacteria from the Ruminococcaceae family and the screened hepatic markers, whereas in other cases the opposite relationship was also found. Interestingly, an interaction was found between UBA-1819 abundance and resveratrol induced liver weight decrease, suggesting that for this marker resveratrol induced effects were greater when the abundance of this bacteria was high, while no actions were found when UBA-1819 abundance was low.
Collapse
Affiliation(s)
- Iñaki Milton-Laskibar
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, Spanish National Research Council, 28049 Madrid, Spain; (I.M.-L.); (A.C.-S.); (J.A.M.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28222 Madrid, Spain
- BIOARABA Institute of Health, 01006 Vitoria-Gasteiz, Spain
| | - Amanda Cuevas-Sierra
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, Spanish National Research Council, 28049 Madrid, Spain; (I.M.-L.); (A.C.-S.); (J.A.M.)
| | - María P. Portillo
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28222 Madrid, Spain
- BIOARABA Institute of Health, 01006 Vitoria-Gasteiz, Spain
- Nutrition and Obesity Group, Department of Pharmacy and Food Sciences, Faculty of Pharmacy and LuciLascaray Research Center, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
- Correspondence: ; Tel.: +34-945-01-30-67
| | - J. Alfredo Martínez
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, Spanish National Research Council, 28049 Madrid, Spain; (I.M.-L.); (A.C.-S.); (J.A.M.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28222 Madrid, Spain
| |
Collapse
|
19
|
Chen D, Bai R, Yong H, Zong S, Jin C, Liu J. Improving the digestive stability and prebiotic effect of carboxymethyl chitosan by grafting with gallic acid: In vitro gastrointestinal digestion and colonic fermentation evaluation. Int J Biol Macromol 2022; 214:685-696. [PMID: 35779653 DOI: 10.1016/j.ijbiomac.2022.06.170] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/31/2022] [Accepted: 06/26/2022] [Indexed: 11/05/2022]
Abstract
Carboxymethyl chitosan (CMCS) is a useful polysaccharide with potential applications in food, cosmetic and biomedical industries. Nonetheless, CMCS is unfavorable for maintaining intestinal flora balance. In this study, gallic acid (GA) was grafted with CMCS through ascorbic acid/hydrogen peroxide initiated graft copolymerization reaction, producing GA grafted CMCS (GA-g-CMCS). The digestive and fermentative behavior of CMCS and GA-g-CMCS were investigated by using in vitro simulated gastrointestinal digestion and colonic fermentation models. Results showed that the average molecular weight (Mw) of CMCS gradually decreased during saliva-gastro-intestinal digestion, changing from original sheet-like morphology to porous and rod-like fragments. However, the Mw and morphology of GA-g-CMCS were almost unchanged under saliva-gastro-intestinal digestion. Meanwhile, the grafted GA moiety was not released from GA-g-CMCS during saliva-gastro-intestinal digestion. As compared with CMCS fermentation, GA-g-CMCS fermentation significantly suppressed the relative abundance of Escherichia-Shigella, Paeniclostridium, Parabacteroides, Lachnoclostridium, Clostridium_sensu_stricto_1, UBA1819 and Butyricimonas, while facilitated the relative abundance of Enterobacter, Enterococcus, Fusobacterium and Lachnospira. In addition, GA-g-CMCS fermentation significantly enhanced the production of short-chain fatty acids. These findings suggested that the digestive stability and prebiotic effect of CMCS were improved by grafting with GA.
Collapse
Affiliation(s)
- Dan Chen
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Ruyu Bai
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Huimin Yong
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Shuai Zong
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Changhai Jin
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China
| | - Jun Liu
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, Jiangsu, China.
| |
Collapse
|