1
|
Wu Y, Zhai S, Fang M, Zhang H, Chen Y. Evaluation of the growth performance, meat quality, and gut microbiota of broilers fed diets containing walnut green husk extract. Poult Sci 2024; 103:104176. [PMID: 39180783 PMCID: PMC11387355 DOI: 10.1016/j.psj.2024.104176] [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: 05/29/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/26/2024] Open
Abstract
This study was conducted to evaluate the effects of walnut green husk extract (WGHe) on the growth performance, meat quality, antioxidative status, gut morphology, and microbiota diversity of broilers. A total of 216 one-day-old broilers were divided into 4 groups, each consisting of 9 replicates (6 birds per replicate) as follows: 1) control group, basal diet; 2) antibiotic group, basal diet supplemented with enduracidin and colistin sulfate; 3) low-dose group, basal diet supplemented with 5.0 g/kg WGHe; and 4) high-dose group, basal diet supplemented with 10.0 g/kg WGHe. The results revealed that the percentage of abdominal fat decreased, and the ratio of the duodenal villus length to crypt depth (V/C), as well as the α-diversity of the ileal microbiota, increased with 10.0 g/kg WGHe supplementation (P < 0.05). The shear force of the breast muscle and plasma malondialdehyde (MDA) concentration decreased, whereas the plasma peroxidase (POD) activity, Trolox equivalent antioxidant capacity (TEAC), and jejunal villus length increased in response to WGHe supplementation (P < 0.05). Compared with the antibiotic diet, the addition of 5.0 g/kg WGHe resulted in a significant increase in the relative abundances of Candidatus Arthromitus, Eubacterium coprostanoligenes, and Ruminococcaceae UCG-014 (P < 0.01). Furthermore, the addition of 10.0 g/kg WGHe increased the relative abundances of Candidatus Arthromitus and Lachnoclostridium, whereas the relative abundance of unidentified Chloroplast decreased (P < 0.05). In conclusion, dietary supplementation with 10.0 g/kg WGHe is advantageous for intestinal health, meat quality, and antioxidant status in broilers, suggesting its potential as a functional additive in poultry production.
Collapse
Affiliation(s)
- Ying Wu
- College of Animal Science, Xinjiang Agricultural University, Urumqi, Xinjiang, 830052, China
| | - Shaohua Zhai
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang, 830052, China
| | - Meiyan Fang
- College of Animal Science, Xinjiang Agricultural University, Urumqi, Xinjiang, 830052, China
| | - Huiling Zhang
- College of Animal Science, Xinjiang Agricultural University, Urumqi, Xinjiang, 830052, China
| | - Yong Chen
- College of Animal Science, Xinjiang Agricultural University, Urumqi, Xinjiang, 830052, China.
| |
Collapse
|
2
|
Cui Y, Wang H, Wang Y. Plasma metabolites as mediators in the relationship between inflammation-related proteins and benign prostatic hyperplasia: insights from mendelian randomization. Sci Rep 2024; 14:26152. [PMID: 39478098 PMCID: PMC11525667 DOI: 10.1038/s41598-024-77515-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024] Open
Abstract
Benign prostatic hyperplasia (BPH) is a condition commonly observed in aging males. Inflammatory and metabolic factors are pivotal in the development and progression of BPH. The degree to which the effects of 91 inflammation-related proteins on BPH are mediated by 1400 plasma metabolites remains ambiguous. Our research analyzed the impact of these traits utilizing genetic evidence.Two-sample Mendelian randomization (MR) and multivariable MR (MVMR) were utilized in our study to infer the genetic causal effect of inflammation-related proteins on BPH, with metabolites serving as mediators. Increased levels of IL-2 were linked to a heightened incidence of BPH (β = 0.071, OR:1.074, 95% CI [1.002-1.152], p = 0.045), whereas lower concentrations of N6,N6-dimethyllysine were associated with decreased risk (β1=-0.127, p = 0.02; β2=-0.039, p = 0.008). The mediation effect was 0.005 (95% CI [0.0004, 0.012], OR: 1.005, 95% CI [1.000, 1.012]), accounting for 7.04% of the total effect. subsequently, we examined the phenotypic co-localization of the two pairings independently, revealing that the posterior probability of rs145516501 associated with IL-2 and BPH was 80.7%, whereas the posterior likelihood of rs4917820 linked to N6,N6-dimethyllysine levels and BPH was 95.9%. The research indicated that N6,N6-dimethyllysine levels seem to influence the causative relationship between IL-2 and BPH. These results elucidate the complex interplay between inflammation-related proteins and metabolism in the context of BPH, offering novel diagnostic and therapeutic avenues and enhancing our comprehension of the disease's etiology for prospective research.
Collapse
Affiliation(s)
- Yan Cui
- Department of Ultrasound, First Affiliated Hospital of Dalian Medical University, Dalian, 116100, Liaoning, China
| | - Hui Wang
- Department of Ultrasound, First Affiliated Hospital of Dalian Medical University, Dalian, 116100, Liaoning, China.
| | - Yuting Wang
- Harbin Medical University, Harbin, 150000, Heilongjiang, China
| |
Collapse
|
3
|
Tyagi SC. Lactobacillus Eats Amyloid Plaque and Post-Biotically Attenuates Senescence Due to Repeat Expansion Disorder and Alzheimer's Disease. Antioxidants (Basel) 2024; 13:1225. [PMID: 39456478 PMCID: PMC11506100 DOI: 10.3390/antiox13101225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Patients with Alzheimer's disease and related dementia (ADRD) are faced with a formidable challenge of focal amyloid deposits and cerebral amyloid angiopathy (CAA). The treatment of amyloid deposits in ADRD by targeting only oxidative stress, inflammation and hyperlipidemia has not yielded significant positive clinical outcomes. The chronic high-fat diet (HFD), or gut dysbiosis, is one of the major contributors of ADRD in part by disrupted transport, epigenetic DNMT1 and the folate 1-carbon metabolism (FOCM) cycle, i.e., rhythmic methylation/de-methylation on DNA, an active part of epigenetic memory during genes turning off and on by the gene writer (DNMT1) and eraser (TET2/FTO) and the transsulfuration pathway by mitochondrial 3-mercaptopyruvate sulfur transferase (3MST)-producing H2S. The repeat CAG expansion and m6A disorder causes senescence and AD. We aim to target the paradigm-shift pathway of the gut-brain microbiome axis that selectively inhibits amyloid deposits and increases mitochondrial transsulfuration and H2S. We have observed an increase in DNMT1 and decreased FTO levels in the cortex of the brain of AD mice. Interestingly, we also observed that probiotic lactobacillus-producing post-biotic folate and lactone/ketone effectively prevented FOCM-associated gut dysbiosis and amyloid deposits. The s-adenosine-methionine (SAM) transporter (SLC25A) was increased by hyperhomocysteinemia (HHcy). Thus, we hypothesize that chronic gut dysbiosis induces SLC25A, the gene writer, and HHcy, and decreases the gene eraser, leading to a decrease in SLC7A and mitochondrial transsulfuration H2S production and bioenergetics. Lactobacillus engulfs lipids/cholesterol and a tri-directional post-biotic, folic acid (an antioxidant and inhibitor of beta amyloid deposits; reduces Hcy levels), and the lactate ketone body (fuel for mitochondria) producer increases SLC7A and H2S (an antioxidant, potent vasodilator and neurotransmitter gas) production and inhibits amyloid deposits. Therefore, it is important to discuss whether lactobacillus downregulates SLC25A and DNMT1 and upregulates TET2/FTO, inhibiting β-amyloid deposits by lowering homocysteine. It is also important to discuss whether lactobacillus upregulates SLC7A and inhibits β-amyloid deposits by increasing the mitochondrial transsulfuration of H2S production.
Collapse
Affiliation(s)
- Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
4
|
Van Hul M, Cani PD, Petitfils C, De Vos WM, Tilg H, El-Omar EM. What defines a healthy gut microbiome? Gut 2024; 73:1893-1908. [PMID: 39322314 PMCID: PMC11503168 DOI: 10.1136/gutjnl-2024-333378] [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: 07/16/2024] [Accepted: 08/16/2024] [Indexed: 09/27/2024]
Abstract
The understanding that changes in microbiome composition can influence chronic human diseases and the efficiency of therapies has driven efforts to develop microbiota-centred therapies such as first and next generation probiotics, prebiotics and postbiotics, microbiota editing and faecal microbiota transplantation. Central to microbiome research is understanding how disease impacts microbiome composition and vice versa, yet there is a problematic issue with the term 'dysbiosis', which broadly links microbial imbalances to various chronic illnesses without precision or definition. Another significant issue in microbiome discussions is defining 'healthy individuals' to ascertain what characterises a healthy microbiome. This involves questioning who represents the healthiest segment of our population-whether it is those free from illnesses, athletes at peak performance, individuals living healthily through regular exercise and good nutrition or even elderly adults or centenarians who have been tested by time and achieved remarkable healthy longevity.This review advocates for delineating 'what defines a healthy microbiome?' by considering a broader range of factors related to human health and environmental influences on the microbiota. A healthy microbiome is undoubtedly linked to gut health. Nevertheless, it is very difficult to pinpoint a universally accepted definition of 'gut health' due to the complexities of measuring gut functionality besides the microbiota composition. We must take into account individual variabilities, the influence of diet, lifestyle, host and environmental factors. Moreover, the challenge in distinguishing causation from correlation between gut microbiome and overall health is presented.The review also highlights the resource-heavy nature of comprehensive gut health assessments, which hinders their practicality and broad application. Finally, we call for continued research and a nuanced approach to better understand the intricate and evolving concept of gut health, emphasising the need for more precise and inclusive definitions and methodologies in studying the microbiome.
Collapse
Affiliation(s)
- Matthias Van Hul
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute, Wavre, Belgium
| | - Patrice D Cani
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute, Wavre, Belgium
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Camille Petitfils
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute, Wavre, Belgium
| | - Willem M De Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medizinische Universitat Innsbruck, Innsbruck, Austria
| | - Emad M El-Omar
- Microbiome Research Centre, St George and Sutherland Clinical Campuses, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
5
|
Wu L, Li Y, Chen S, Yang Y, Tang B, Weng M, Shen H, Chen J, Lai P. Widely Targeted Lipidomics and Microbiomics Perspectives Reveal the Mechanism of Auricularia auricula Polysaccharide's Effect of Regulating Glucolipid Metabolism in High-Fat-Diet Mice. Foods 2024; 13:2743. [PMID: 39272508 PMCID: PMC11395039 DOI: 10.3390/foods13172743] [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: 07/19/2024] [Revised: 08/19/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
The role of Auricularia auricula polysaccharide (AP) in the regulation of glycolipid metabolism was investigated using a high-fat-diet-induced hyperlipidemic mouse model. In a further step, its potential mechanism of action was investigated using microbiome analysis and widely targeted lipidomics. Compared to high-fat mice, dietary AP supplementation reduced body weight by 13.44%, liver index by 21.30%, epididymal fat index by 50.68%, fasting blood glucose (FBG) by 14.27%, serum total cholesterol (TC) by 20.30%, serum total triglycerides (TGs) by 23.81%, liver non-esterified fatty acid (NEFA) by 20.83%, liver TGs by 20.00%, and liver malondialdehyde (MDA) by 21.05%, and increased liver glutathione oxidase (GSH-PX) activity by 52.24%, total fecal bile acid (TBA) by 46.21%, and fecal TG by 27.16%, which significantly regulated glucose and lipid metabolism. Microbiome analysis showed that AP significantly downregulated the abundance of the Desulfobacterota phylum, as well as the genii Desulfovibrio, Bilophila, and Oscillbacter in the cecum of hyperlipidemic mice, which are positively correlated with high lipid indexes, while it upregulated the abundance of the families Eubacterium_coprostanoligenes_group and Ruminococcaceae, as well as the genii Eubacterum_xylanophilum_group, Lachnospiraceae_NK4A136_group, Eubacterium_siraeum_group, and Parasutterella, which were negatively correlated with high lipid indexes. In addition, AP promoted the formation of SCFAs by 119.38%. Widely targeted lipidomics analysis showed that AP intervention regulated 44 biomarkers in metabolic pathways such as sphingolipid metabolism and the AGE-RAGE signaling pathway in the hyperlipidemic mice (of which 15 metabolites such as unsaturated fatty acids, phosphatidylserine, and phosphatidylethanolamine were upregulated, and 29 metabolites such as phosphatidylcholine, ceramide, carnitine, and phosphatidylinositol were downregulated), thereby correcting glucose and lipid metabolism disorders.
Collapse
Affiliation(s)
- Li Wu
- Institute of Food Science and Technology, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China
- National R & D Center of Edible Fungi Processing, Fuzhou 350003, China
- Key Laboratory of Subtropical Characteristic Fruits, Vegetables and Edible Fungi Processing (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Fuzhou 350003, China
- Fujian Province Key Laboratory of Agricultural Products (Food) Processing Technology, Fuzhou 350003, China
- Fujian Characteristic Agricultural Products Processing Technology and Economic Integration Service Platform, Fuzhou 350003, China
| | - Yibin Li
- Institute of Food Science and Technology, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China
- National R & D Center of Edible Fungi Processing, Fuzhou 350003, China
- Key Laboratory of Subtropical Characteristic Fruits, Vegetables and Edible Fungi Processing (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Fuzhou 350003, China
- Fujian Province Key Laboratory of Agricultural Products (Food) Processing Technology, Fuzhou 350003, China
- Fujian Characteristic Agricultural Products Processing Technology and Economic Integration Service Platform, Fuzhou 350003, China
| | - Shouhui Chen
- Institute of Food Science and Technology, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China
- National R & D Center of Edible Fungi Processing, Fuzhou 350003, China
| | - Yanrong Yang
- Institute of Food Science and Technology, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China
- National R & D Center of Edible Fungi Processing, Fuzhou 350003, China
| | - Baosha Tang
- Institute of Food Science and Technology, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China
- National R & D Center of Edible Fungi Processing, Fuzhou 350003, China
- Key Laboratory of Subtropical Characteristic Fruits, Vegetables and Edible Fungi Processing (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Fuzhou 350003, China
- Fujian Province Key Laboratory of Agricultural Products (Food) Processing Technology, Fuzhou 350003, China
- Fujian Characteristic Agricultural Products Processing Technology and Economic Integration Service Platform, Fuzhou 350003, China
| | - Minjie Weng
- Institute of Food Science and Technology, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China
- National R & D Center of Edible Fungi Processing, Fuzhou 350003, China
- Key Laboratory of Subtropical Characteristic Fruits, Vegetables and Edible Fungi Processing (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Fuzhou 350003, China
- Fujian Province Key Laboratory of Agricultural Products (Food) Processing Technology, Fuzhou 350003, China
- Fujian Characteristic Agricultural Products Processing Technology and Economic Integration Service Platform, Fuzhou 350003, China
| | - Hengsheng Shen
- Institute of Food Science and Technology, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China
- National R & D Center of Edible Fungi Processing, Fuzhou 350003, China
| | - Junchen Chen
- Institute of Food Science and Technology, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China
- National R & D Center of Edible Fungi Processing, Fuzhou 350003, China
- Key Laboratory of Subtropical Characteristic Fruits, Vegetables and Edible Fungi Processing (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Fuzhou 350003, China
- Fujian Province Key Laboratory of Agricultural Products (Food) Processing Technology, Fuzhou 350003, China
- Fujian Characteristic Agricultural Products Processing Technology and Economic Integration Service Platform, Fuzhou 350003, China
| | - Pufu Lai
- Institute of Food Science and Technology, Fujian Academy of Agricultural Sciences, Fuzhou 350003, China
- National R & D Center of Edible Fungi Processing, Fuzhou 350003, China
- Key Laboratory of Subtropical Characteristic Fruits, Vegetables and Edible Fungi Processing (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Fuzhou 350003, China
- Fujian Province Key Laboratory of Agricultural Products (Food) Processing Technology, Fuzhou 350003, China
- Fujian Characteristic Agricultural Products Processing Technology and Economic Integration Service Platform, Fuzhou 350003, China
| |
Collapse
|
6
|
Duan Z, Yang M, Yang J, Wu Z, Zhu Y, Jia Q, Ma X, Yin Y, Zheng J, Yang J, Jiang S, Hu L, Zhang J, Liu D, Huo Y, Yao L, Sun Y. AGFG1 increases cholesterol biosynthesis by disrupting intracellular cholesterol homeostasis to promote PDAC progression. Cancer Lett 2024; 598:217130. [PMID: 39089666 DOI: 10.1016/j.canlet.2024.217130] [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: 05/14/2024] [Revised: 07/01/2024] [Accepted: 07/17/2024] [Indexed: 08/04/2024]
Abstract
PURPOSE Cholesterol metabolism reprograming has been acknowledged as a novel feature of cancers. Pancreatic ductal adenocarcinoma (PDAC) is a cancer with a high demand of cholesterol for rapid growth. The underlying mechanism of how cholesterol metabolism homestasis are disturbed in PDAC is explored. EXPERIMENTAL DESIGN The relevance between PDAC and cholesterol was confirmed in TCGA database. The expression and clinical association were discovered in TCGA and GEO datasets. Knockdown and overexpression of AGFG1 was adopted to perform function studies. RNA sequencing, cholesterol detection, transmission electron microscope, co-immunoprecipitation, and immunofluorescence et al. were utilized to reveal the underlying mechanism. RESULTS AGFG1 was identified as one gene positively correlated with cholesterol metabolism in PDAC as revealed by bioinformatics analysis. AGFG1 expression was then found associated with poor prognosis in PDAC. AGFG1 knockdown led to decreased proliferation of tumor cells both in vitro and in vivo. By RNA sequencing, we found AGFG1 upregulated expression leads to enhanced intracellular cholesterol biosynthesis. AGFG1 knockdown suppressed cholesterol biosynthesis and an accumulation of cholesterol in the ER. Mechanistically, we confirmed that AGFG1 interacted with CAV1 to relocate cholesterol for the proceeding of cholesterol biosynthesis, therefore causing disorders in intracellular cholesterol metabolism. CONCLUSIONS Our study demonstrates the tumor-promoting role of AGFG1 by disturbing cholesterol metabolism homestasis in PDAC. Our study has present a new perspective on cancer therapeutic approach based on cholerstrol metabolism in PDAC.
Collapse
Affiliation(s)
- Zonghao Duan
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Minwei Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Jian Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, PR China; Department of General Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, PR China; Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100029, PR China
| | - Zheng Wu
- Department of Radiation Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Yuheng Zhu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Qinyuan Jia
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Xueshiyu Ma
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Yifan Yin
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Jiahao Zheng
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China; State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Jianyu Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Shuheng Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Lipeng Hu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Junfeng Zhang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China; Department of General Surgery, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, PR China
| | - Dejun Liu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China.
| | - Yanmiao Huo
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China.
| | - Linli Yao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, PR China.
| | - Yongwei Sun
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, PR China.
| |
Collapse
|
7
|
Bellach L, Kautzky-Willer A, Heneis K, Leutner M, Kautzky A. The Effects of Caloric Restriction and Clinical Psychological Intervention on the Interplay of Gut Microbial Composition and Stress in Women. Nutrients 2024; 16:2584. [PMID: 39203721 PMCID: PMC11357322 DOI: 10.3390/nu16162584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/23/2024] [Accepted: 08/03/2024] [Indexed: 09/03/2024] Open
Abstract
Both mental and metabolic disorders are steadily becoming more prevalent, increasing interest in non-pharmacological lifestyle interventions targeting both types of disorders. However, the combined effect of diet and psychological interventions on the gut microbiome and mental health outcomes remains underexplored. Thus, in this study, we randomized 41 women into two caloric restriction (CR) dietary groups, namely very-low-calorie diet (VLCD) and F.X. Mayr diet (FXM). The patients were then further randomized to either receive clinical psychological intervention (CPI) or no CPI. Blood and fecal samples were collected before and after two weeks of CR. Psychometric outcomes were assessed using the Perceived Stress Scale (PSS), Brief Symptom Index (BSI), and Burnout Dimension Inventory (BODI). Stool samples underwent 16S-rRNA sequencing. Upon two weeks of CR, α-diversity decreased overall and longitudinal PERMANOVA models revealed significant shifts in β-diversity according to diet, CPI, age, and body-mass-index. Furthermore, Agathobacter, Fusicatenibacter, and Subdoligranulum decreased in abundance. However, the Oscillibacter genus was enriched solely in FXM. CPI had a negligible effect on the microbiome. Dimension reduction models revealed clusters of taxa which distinctly associated with psychometric outcomes. Members of the Oscillospiraceae family were linked to favorable psychometric outcomes after two weeks of CR. Despite α-diversity reductions after CR, enrichment of Oscillospiraceae spp., solely seen in FXM, correlated with improved psychometric outcomes. This study suggests a promising direction for future interventions targeting mental health through gut microbial modulation.
Collapse
Affiliation(s)
- Luise Bellach
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, 1090 Vienna, Austria
| | - Alexandra Kautzky-Willer
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, 1090 Vienna, Austria
| | - Kathrin Heneis
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Leutner
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, 1090 Vienna, Austria
| | - Alexander Kautzky
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
8
|
Adolph TE, Tilg H. Western diets and chronic diseases. Nat Med 2024; 30:2133-2147. [PMID: 39085420 DOI: 10.1038/s41591-024-03165-6] [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: 03/01/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024]
Abstract
'Westernization', which incorporates industrial, cultural and dietary trends, has paralleled the rise of noncommunicable diseases across the globe. Today, the Western-style diet emerges as a key stimulus for gut microbial vulnerability, chronic inflammation and chronic diseases, affecting mainly the cardiovascular system, systemic metabolism and the gut. Here we review the diet of modern times and evaluate the threat it poses for human health by summarizing recent epidemiological, translational and clinical studies. We discuss the links between diet and disease in the context of obesity and type 2 diabetes, cardiovascular diseases, gut and liver diseases and solid malignancies. We collectively interpret the evidence and its limitations and discuss future challenges and strategies to overcome these. We argue that healthcare professionals and societies must react today to the detrimental effects of the Western diet to bring about sustainable change and improved outcomes in the future.
Collapse
Affiliation(s)
- Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
9
|
Liedike B, Khatib M, Tabarsi B, Harris M, Wilson SL, Ortega-Santos CP, Mohr AE, Vega-López S, Whisner CM. Evaluating the Effects of Corn Flour Product Consumption on Cardiometabolic Outcomes and the Gut Microbiota in Adults with Elevated Cholesterol: A Randomized Crossover. J Nutr 2024; 154:2437-2447. [PMID: 38880174 DOI: 10.1016/j.tjnut.2024.06.003] [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: 03/12/2024] [Revised: 05/24/2024] [Accepted: 06/12/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Consumption of whole grains is associated with a reduction in chronic diseases and offers benefits for cardiovascular health and metabolic regulation. The relationship between whole-grain corn and corn bran with the gut microbiota (GM) remains an area of growing interest, particularly regarding their influence on cardiometabolic health. OBJECTIVES To investigate the effects of different corn flours on cardiometabolic outcomes and GM changes in adults with elevated low-density lipoprotein cholesterol (LDL cholesterol) concentrations. METHODS In this crossover study, 36 adults with LDL cholesterol above 110 mg/dL consumed 48 g/d of 3 corn flour types for 4 wk: whole-grain corn meal, refined corn meal (RCM), and a blend of RCM and corn bran (RCM + B). We assessed the impact on cardiometabolic markers [LDL cholesterol, high-density lipoprotein cholesterol (HDL cholesterol), total cholesterol, and triglycerides)] and GM composition and estimated function. Statistical analyses included mixed-effects modeling and responder (>5% decrease in LDL cholesterol) analysis to evaluate changes in GM related to lipid profile improvements. RESULTS Of the 3 corn flour types, only RCM + B significantly decreased LDL cholesterol over time (-10.4 ± 3.6 mg/dL, P = 0.005) and marginally decreased total cholesterol (-9.2 ± 3.9 mg/dL, P = 0.072) over time. There were no significant effects on HDL cholesterol or triglyceride concentrations. No significant changes were observed in GM alpha diversity, whereas beta diversity metrics indicated individual variability. Two genera, unclassified Lachnospiraceae and Agathobaculum (Padj ≤ 0.096), differed significantly by treatment, but only Agathobaculum remained significantly elevated in the whole-grain corn meal, compared to RCM and RCM + B, after adjustment for multiple comparisons. CONCLUSIONS The type of corn flour, particularly RCM + B, notably influenced LDL cholesterol concentrations in adults with elevated LDL cholesterol. This study suggests that incorporating milled fractions (e.g., bran) of whole-grain corn with refined corn flour may be a viable alternative to supplementing manufactured grain products with isolated or synthetic fibers for improved metabolic health. This trial was registered at clinicaltrials.gov as NCT03967990.
Collapse
Affiliation(s)
- Bethany Liedike
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States
| | - Maissa Khatib
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States
| | - Baharak Tabarsi
- Community Health Center, Valleywise Health, Phoenix, AZ, United States
| | - Michelle Harris
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States
| | - Shannon L Wilson
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States
| | - Carmen P Ortega-Santos
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States; Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Alex E Mohr
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States; Center for Health Through Microbiomes, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Sonia Vega-López
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States; Southwest Interdisciplinary Research Center, Arizona State University, Phoenix, AZ, United States.
| | - Corrie M Whisner
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States; Center for Health Through Microbiomes, The Biodesign Institute, Arizona State University, Tempe, AZ, United States.
| |
Collapse
|
10
|
Fei SF, Hou C, Jia F. Effects of salidroside on atherosclerosis: potential contribution of gut microbiota. Front Pharmacol 2024; 15:1400981. [PMID: 39092226 PMCID: PMC11292615 DOI: 10.3389/fphar.2024.1400981] [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: 05/08/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024] Open
Abstract
Much research describes gut microbiota in atherosclerotic cardiovascular diseases (ASCVD) for that the composition of the intestinal microbiome or its metabolites can directly participate in the development of endothelial dysfunction, atherosclerosis and its adverse complications. Salidroside, a natural phenylpropane glycoside, exhibits promising biological activity against the progression of ASCVD. Recent studies suggested that the gut microbiota played a crucial role in mediating the diverse beneficial effects of salidroside on health. Here, we describe the protective effects of salidroside against the progression of atherosclerosis. Salidroside regulates the abundance of gut microbiotas and gut microbe-dependent metabolites. Moreover, salidroside improves intestinal barrier function and maintains intestinal epithelial barrier function integrity. In addition, salidroside attenuates the inflammatory responses exacerbated by gut microbiota disturbance. This review delves into how salidroside functions to ameliorate atherosclerosis by focusing on its interaction with gut microbiota, uncovering the potential roles of gut microbiota in the diverse biological impacts of salidroside.
Collapse
Affiliation(s)
| | | | - Fang Jia
- Department of Cardiovascular Medicine, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
11
|
Nkeh YB, Elgin RG, Saher G. From the unknown to spotlight: newly identified hormone adjusts hepatic cholesterol synthesis to dietary uptake. Signal Transduct Target Ther 2024; 9:165. [PMID: 38909021 PMCID: PMC11193724 DOI: 10.1038/s41392-024-01882-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 06/24/2024] Open
Affiliation(s)
- Yakum Bertrand Nkeh
- Project Group Biology of Lipids, Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Resul Gökberk Elgin
- Project Group Biology of Lipids, Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Gesine Saher
- Project Group Biology of Lipids, Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
Collapse
|
12
|
Shi L, Liu X, Li E, Zhang S, Zhou A. Association of lipid-lowering drugs with gut microbiota: A Mendelian randomization study. J Clin Lipidol 2024:S1933-2874(24)00187-9. [PMID: 38971663 DOI: 10.1016/j.jacl.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/07/2024] [Accepted: 05/21/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND The gut microbiota can be influenced by lipid metabolism. We aimed to evaluate the impact of lipid-lowering medications, such as proproteinconvertase subtilisin/kexin type 9 (PCSK9) inhibitors, Niemann-Pick C1-like protein (NPC1L1) inhibitors, and 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) inhibitors, on gut microbiota through drug target Mendelian randomization (MR) investigation. METHODS We used genetic variants that were associated with low-density lipoprotein cholesterol (LDL-C) in genome-wide association studies and located within or near drug target genes as proxies for lipid-lowering drug exposure. In addition, expression trait loci in drug target genes were used as complementary genetic tools. We used effect estimates calculated using inverse variance weighted MR (IVW-MR) and summary data-based MR (SMR). Multiple sensitivity analyses were performed. RESULTS Genetic proxies for lipid-lowering drugs broadly affected the abundance of gut microbiota. High expression of NPC1L1 was significantly associated with an increase in the genus Eggerthella (β = 1.357, SE = 0.337, P = 5.615 × 10-5). An HMGCR-mediated increase in LDL-C was significantly associated with the order Pasteurellales (β = 0.489, SE = 0.123, P = 6.955 × 10-5) and the genus Haemophilus (β = 0.491, SE = 0.125, P = 8.379 × 10-5), whereas a PCSK9-mediated increase in LDL-C was associated with the genus Terrisporobacter (β = 0.666, SE = 0.127, P = 1.649 × 10-5). No pleiotropy was detected. CONCLUSIONS This drug target MR highlighted the potential interventional effects of lipid-lowering drugs on the gut microbiota and separately revealed the possible effects of different types of lipid-lowering drugs on specific gut microbiota.
Collapse
Affiliation(s)
- Lubo Shi
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing, PR China (Drs Shi, Zhang, Zhou)
| | - Xiaoduo Liu
- Department of Neurology & Innovation Center for Neurological Disorders , Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, PR China (Dr Liu)
| | - Enze Li
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, National Clinical Research Center for Cardiovascular Diseases, Beijing, PR China (Dr Li)
| | - Shutian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing, PR China (Drs Shi, Zhang, Zhou).
| | - Anni Zhou
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Diseases, Beijing Digestive Disease Center, Beijing, PR China (Drs Shi, Zhang, Zhou).
| |
Collapse
|
13
|
Sodders MJ, Avila-Pacheco J, Okorie EC, Shen M, Kumari N, Marathi A, Lakhani M, Bullock K, Pierce K, Dennis C, Jeanfavre S, Sarkar S, Scherzer CR, Clish C, Olsen AL. Genetic screening and metabolomics identify glial adenosine metabolism as a therapeutic target in Parkinson's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594309. [PMID: 38798570 PMCID: PMC11118494 DOI: 10.1101/2024.05.15.594309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder and lacks disease-modifying therapies. We developed a Drosophila model for identifying novel glial-based therapeutic targets for PD. Human alpha-synuclein is expressed in neurons and individual genes are independently knocked down in glia. We performed a forward genetic screen, knocking down the entire Drosophila kinome in glia in alpha-synuclein expressing flies. Among the top hits were five genes (Ak1, Ak6, Adk1, Adk2, and awd) involved in adenosine metabolism. Knockdown of each gene improved locomotor dysfunction, rescued neurodegeneration, and increased brain adenosine levels. We determined that the mechanism of neuroprotection involves adenosine itself, as opposed to a downstream metabolite. We dove deeper into the mechanism for one gene, Ak1, finding rescue of dopaminergic neuron loss, alpha-synuclein aggregation, and bioenergetic dysfunction after glial Ak1 knockdown. We performed metabolomics in Drosophila and in human PD patients, allowing us to comprehensively characterize changes in purine metabolism and identify potential biomarkers of dysfunctional adenosine metabolism in people. These experiments support glial adenosine as a novel therapeutic target in PD.
Collapse
|
14
|
Hindson J. Multi-omic links between gut microbiome and cardiovascular disease. Nat Rev Gastroenterol Hepatol 2024; 21:376. [PMID: 38744959 DOI: 10.1038/s41575-024-00941-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
|
15
|
Fernández-Ruiz I. Gut bacteria can break down cholesterol. Nat Rev Cardiol 2024; 21:357. [PMID: 38627565 DOI: 10.1038/s41569-024-01026-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
|
16
|
Kandalgaonkar MR, Kumar V, Vijay‐Kumar M. Digestive dynamics: Unveiling interplay between the gut microbiota and the liver in macronutrient metabolism and hepatic metabolic health. Physiol Rep 2024; 12:e16114. [PMID: 38886098 PMCID: PMC11182692 DOI: 10.14814/phy2.16114] [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: 04/11/2024] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
Although the liver is the largest metabolic organ in the body, it is not alone in functionality and is assisted by "an organ inside an organ," the gut microbiota. This review attempts to shed light on the partnership between the liver and the gut microbiota in the metabolism of macronutrients (i.e., proteins, carbohydrates, and lipids). All nutrients absorbed by the small intestines are delivered to the liver for further metabolism. Undigested food that enters the colon is metabolized further by the gut microbiota that produces secondary metabolites, which are absorbed into portal circulation and reach the liver. These microbiota-derived metabolites and co-metabolites include ammonia, hydrogen sulfide, short-chain fatty acids, secondary bile acids, and trimethylamine N-oxide. Further, the liver produces several compounds, such as bile acids that can alter the gut microbial composition, which can in turn influence liver health. This review focuses on the metabolism of these microbiota metabolites and their influence on host physiology. Furthermore, the review briefly delineates the effect of the portosystemic shunt on the gut microbiota-liver axis, and current understanding of the treatments to target the gut microbiota-liver axis.
Collapse
Affiliation(s)
- Mrunmayee R. Kandalgaonkar
- Department of Physiology and PharmacologyUniversity of Toledo College of Medicine and Life SciencesToledoOhioUSA
| | - Virender Kumar
- College of Pharmacy and Pharmaceutical SciencesUniversity of ToledoToledoOhioUSA
| | - Matam Vijay‐Kumar
- Department of Physiology and PharmacologyUniversity of Toledo College of Medicine and Life SciencesToledoOhioUSA
| |
Collapse
|
17
|
Nowogrodzki J. Gut bacteria break down cholesterol - hinting at probiotic treatments. Nature 2024:10.1038/d41586-024-00955-3. [PMID: 38565909 DOI: 10.1038/d41586-024-00955-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
|
18
|
Kraimi N, Ross T, Pujo J, De Palma G. The gut microbiome in disorders of gut-brain interaction. Gut Microbes 2024; 16:2360233. [PMID: 38949979 PMCID: PMC11218806 DOI: 10.1080/19490976.2024.2360233] [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: 01/30/2024] [Accepted: 05/21/2024] [Indexed: 07/03/2024] Open
Abstract
Functional gastrointestinal disorders (FGIDs), chronic disorders characterized by either abdominal pain, altered intestinal motility, or their combination, have a worldwide prevalence of more than 40% and impose a high socioeconomic burden with a significant decline in quality of life. Recently, FGIDs have been reclassified as disorders of gut-brain interaction (DGBI), reflecting the key role of the gut-brain bidirectional communication in these disorders and their impact on psychological comorbidities. Although, during the past decades, the field of DGBIs has advanced significantly, the molecular mechanisms underlying DGBIs pathogenesis and pathophysiology, and the role of the gut microbiome in these processes are not fully understood. This review aims to discuss the latest body of literature on the complex microbiota-gut-brain interactions and their implications in the pathogenesis of DGBIs. A better understanding of the existing communication pathways between the gut microbiome and the brain holds promise in developing effective therapeutic interventions for DGBIs.
Collapse
Affiliation(s)
- Narjis Kraimi
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Taylor Ross
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Julien Pujo
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Giada De Palma
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| |
Collapse
|
19
|
Roy M, Dumay A, Adiba S, Rozes S, Kobayashi S, Paradis V, Postic C, Rainteau D, Ogier-Denis E, Le Gall M, Meinzer U, Viennois E, Casado-Bedmar M, Mosca A, Hugot JP. Entamoeba muris mitigates metabolic consequences of high-fat diet in mice. Gut Microbes 2024; 16:2409210. [PMID: 39396247 PMCID: PMC11485694 DOI: 10.1080/19490976.2024.2409210] [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: 03/20/2024] [Revised: 09/12/2024] [Accepted: 09/22/2024] [Indexed: 10/15/2024] Open
Abstract
Metabolic syndrome (MetS) is a cluster of several human conditions including abdominal obesity, hypertension, dyslipidemia, and hyperglycemia, all of which are risk factors of type 2 diabetes, cardiovascular disease, and metabolic dysfunction-associated steatotic liver disease (MASLD). Dietary pattern is a well-recognized MetS risk factor, but additional changes related to the modern Western life-style may also contribute to MetS. Here we hypothesize that the disappearance of amoebas in the gut plays a role in the emergence of MetS in association with dietary changes. Four groups of C57B/6J mice fed with a high-fat diet (HFD) or a normal diet (ND) were colonized or not with Entamoeba muris, a commensal amoeba. Seventy days after inoculation, cecal microbiota, and bile acid compositions were analyzed by high-throughput sequencing of 16S rDNA and mass spectrometry, respectively. Cytokine concentrations were measured in the gut, liver, and mesenteric fat looking for low-grade inflammation. The impact of HFD on liver metabolic dysfunction was explored by Oil Red O staining, triglycerides, cholesterol concentrations, and the expression of genes involved in β-oxidation and lipogenesis. Colonization with E. muris had a beneficial impact, with a reduction in dysbiosis, lower levels of fecal secondary bile acids, and an improvement in hepatic steatosis, arguing for a protective role of commensal amoebas in MetS and more specifically HFD-associated MASLD.
Collapse
Affiliation(s)
- Maryline Roy
- Inflammation Research Centre, UMR 1149, INSERM, Université Paris Cité, Paris, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Anne Dumay
- Inflammation Research Centre, UMR 1149, INSERM, Université Paris Cité, Paris, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Sandrine Adiba
- Département de biologie, institut de Biologie de l’ENS, Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Sylvana Rozes
- Inflammation Research Centre, UMR 1149, INSERM, Université Paris Cité, Paris, France
- Department of pediatric gastroenterology and nutrition, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Seiki Kobayashi
- Department of Parasitology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Valérie Paradis
- Inflammation Research Centre, UMR 1149, INSERM, Université Paris Cité, Paris, France
- Department of pathology, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, Clichy, France
| | - Catherine Postic
- Institut Cochin, CNRS, INSERM, Université Paris Cité, Paris, France
| | - Dominique Rainteau
- INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Sorbonne Université, Paris, France
| | - Eric Ogier-Denis
- Inflammation Research Centre, UMR 1149, INSERM, Université Paris Cité, Paris, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Maud Le Gall
- Inflammation Research Centre, UMR 1149, INSERM, Université Paris Cité, Paris, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Ulrich Meinzer
- Inflammation Research Centre, UMR 1149, INSERM, Université Paris Cité, Paris, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
- Department of pediatric gastroenterology and nutrition, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Emilie Viennois
- Inflammation Research Centre, UMR 1149, INSERM, Université Paris Cité, Paris, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Maite Casado-Bedmar
- Inflammation Research Centre, UMR 1149, INSERM, Université Paris Cité, Paris, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Alexis Mosca
- Inflammation Research Centre, UMR 1149, INSERM, Université Paris Cité, Paris, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
- Department of pediatric gastroenterology and nutrition, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Pierre Hugot
- Inflammation Research Centre, UMR 1149, INSERM, Université Paris Cité, Paris, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
- Department of pediatric gastroenterology and nutrition, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
20
|
Zhang H, Xie Y, Cao F, Song X. Gut microbiota-derived fatty acid and sterol metabolites: biotransformation and immunomodulatory functions. Gut Microbes 2024; 16:2382336. [PMID: 39046079 PMCID: PMC11271093 DOI: 10.1080/19490976.2024.2382336] [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: 12/28/2023] [Revised: 05/26/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
Commensal microorganisms in the human gut produce numerous metabolites by using small molecules derived from the host or diet as precursors. Host or dietary lipid molecules are involved in energy metabolism and maintaining the structural integrity of cell membranes. Notably, gut microbes can convert these lipids into bioactive signaling molecules through their biotransformation and synthesis pathways. These microbiota-derived lipid metabolites can affect host physiology by influencing the body's immune and metabolic processes. This review aims to summarize recent advances in the microbial transformation and host immunomodulatory functions of these lipid metabolites, with a special focus on fatty acids and steroids produced by our gut microbiota.
Collapse
Affiliation(s)
- Haohao Zhang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yadong Xie
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Fei Cao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xinyang Song
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|