1
|
Brigagão Pacheco da Silva C, Nascimento-Silva EA, Zaramela LS, da Costa BRB, Rodrigues VF, De Martinis BS, Carlos D, Tostes RC. Drinking pattern and sex modulate the impact of ethanol consumption on the mouse gut microbiome. Physiol Genomics 2025; 57:179-194. [PMID: 39918827 DOI: 10.1152/physiolgenomics.00031.2024] [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: 03/25/2024] [Revised: 04/17/2024] [Accepted: 01/28/2025] [Indexed: 03/04/2025] Open
Abstract
Gut microbiota impacts host homeostasis and diseases. Chronic plus binge ethanol consumption has been linked to increased injuries than chronic or binge ethanol intake alone. We hypothesized that distinct shapes in gut microbiota composition are induced by chronic, binge, and the association of these treatments, thereby affecting host functions and contributing to sex-based differences in alcohol use disorders. Male and female C57BL/6J mice were submitted to chronic, binge, or chronic plus binge ethanol feeding. DNA was extracted from fecal microbiota, followed by analysis of the V3-V4 region of the 16S rRNA gene and sequencing on an Illumina platform. Gut microbiome analysis was performed using QIIME v2022.2.0. Functional profiling of the gut microbiome was performed using PICRUSt2. Ethanol differentially affected the gut microbiota of female and male mice. Decreased α diversity was observed in male and female mice from the chronic plus binge and chronic groups, respectively. The genera Faecalibaculum, Lachnospiraceae, and Alistipes were identified as major potential biomarkers for gut dysbiosis induced by ethanol consumption. In addition, ethanol-induced gut dysbiosis altered several metabolic pathways. Ethanol consumption modifies the mouse gut microbiome in a drinking pattern- and sex-dependent manner, potentially leading to different susceptibility to ethanol-related diseases. Chronic plus binge ethanol intake induces a more pronounced gut dysbiosis in male mice. Conversely, chronic ethanol is linked to a greater degree of gut dysbiosis in female mice. The changed gut microbiome may be potentially targeted to prevent, mitigate, or treat alcohol use disorders.NEW & NOTEWORTHY Ethanol alters the mouse gut microbiome in a drinking pattern- and sex-dependent manner. Chronic plus binge ethanol intake induces a more severe gut dysbiosis in male mice, whereas chronic ethanol consumption appears to be a more potent inductor of gut dysbiosis in female mice. Ethanol-induced gut dysbiosis alters several pathways linked to metabolism, genetic and environmental information processing, cellular processes, organism systems, and neurological human diseases.
Collapse
Affiliation(s)
| | | | - Lívia Soares Zaramela
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Bruno Ruiz Brandão da Costa
- Department of Clinical, Toxicological and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Vanessa Fernandes Rodrigues
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Bruno Spinosa De Martinis
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Daniela Carlos
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
2
|
Júnior REM, Pedersen ASB, Ferreira RM, de Asevedo GH, Mendes GL, Ribeiro K, Maioli TU, de Faria AMC, Brunialti-Godard AL. Behavioral changes and transcriptional regulation of mesolimbic dopaminergic genes in a mouse model of binge eating disorder by diet intermittent access. J Nutr Biochem 2025; 135:109784. [PMID: 39426552 DOI: 10.1016/j.jnutbio.2024.109784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 08/02/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024]
Abstract
Binge Eating Disorder (BED) is among the most prevalent eating disorders worldwide. It is characterized by recurrent episodes of excessive consumption of palatable foods in short periods, accompanied by a sense of loss of control and distress around the episode, which tends to worsen over time. The mesolimbic dopaminergic system influences on reinforcement and reward-seeking behaviors is implicated in the disorder's pathogenesis. Animal models that replicate the clinical conditions observed in humans, including the disorder progression, are essential for understanding the underlying physiological mechanisms of BED. This study aimed to evaluate binge eating behavior induced by intermittent High Sugar and Butter (HSB) diet access in mice, their phenotypes, transcriptional regulation of mesolimbic dopaminergic system genes, and behavior. Thus, mice were subdivided into three groups: CHOW (maintenance diet only), HSB-i (maintenance diet with thrice-weekly access to HSB), and HSB (continuous access to HSB). Animals were subjected to marble-burying and light-dark box behavioral tests, and transcriptional regulation was evaluated by RT-qPCR. The results indicated that the HSB-i group established a feeding pattern of significantly more kilocalories on days when HSB was available and reduced intake on non-HSB days similar to human binge eating. Over time, binge episodes intensified, potentially indicating a tolerance effect. Additionally, these animals behave differently towards preferring the HSB diet and exhibited altered transcriptional regulation of the Drd1, Slc6a3, and Lrrk2 genes. Our study provides a mouse model that reflects human BED, showing a progression in binge episodes and mesolimbic dopamine pathway involvement, suggesting targets for future therapeutic interventions.
Collapse
Affiliation(s)
- Renato Elias Moreira Júnior
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Agatha Sondertoft Braga Pedersen
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Raquel Mary Ferreira
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Guilherme Henrique de Asevedo
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Grazielle Laudares Mendes
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Karine Ribeiro
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tatiani Uceli Maioli
- Laboratório de Imunobiologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Maria Caetano de Faria
- Laboratório de Imunobiologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Lúcia Brunialti-Godard
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
3
|
Zhu Y, Chen B, Zhang X, Akbar MT, Wu T, Zhang Y, Zhi L, Shen Q. Exploration of the Muribaculaceae Family in the Gut Microbiota: Diversity, Metabolism, and Function. Nutrients 2024; 16:2660. [PMID: 39203797 PMCID: PMC11356848 DOI: 10.3390/nu16162660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
The gut microbiota are mainly composed of Bacteroidetes and Firmicutes and are crucial for metabolism and immunity. Muribaculaceae are a family of bacteria within the order Bacteroidetes. Muribaculaceae produce short-chain fatty acids via endogenous (mucin glycans) and exogenous polysaccharides (dietary fibres). The family exhibits a cross-feeding relationship with probiotics, such as Bifidobacterium and Lactobacillus. The alleviating effects of a plant-based diet on inflammatory bowel disease, obesity, and type 2 diabetes are associated with an increased abundance of Muribaculaceae, a potential probiotic bacterial family. This study reviews the current findings related to Muribaculaceae and systematically introduces their diversity, metabolism, and function. Additionally, the mechanisms of Muribaculaceae in the alleviation of chronic diseases and the limitations in this field of research are introduced.
Collapse
Affiliation(s)
- Yiqing Zhu
- College of Food Science and Nutritional Engineering, China Agricultural University, National Center of Technology Innovation (Deep Processing of Highland Barley) in Food Industry, National Engineering Research Center for Fruit and Vegetable Processing, Beijing 100083, China; (Y.Z.); (B.C.); (X.Z.); (M.T.A.); (T.W.); (Y.Z.); (L.Z.)
| | - Borui Chen
- College of Food Science and Nutritional Engineering, China Agricultural University, National Center of Technology Innovation (Deep Processing of Highland Barley) in Food Industry, National Engineering Research Center for Fruit and Vegetable Processing, Beijing 100083, China; (Y.Z.); (B.C.); (X.Z.); (M.T.A.); (T.W.); (Y.Z.); (L.Z.)
| | - Xinyu Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, National Center of Technology Innovation (Deep Processing of Highland Barley) in Food Industry, National Engineering Research Center for Fruit and Vegetable Processing, Beijing 100083, China; (Y.Z.); (B.C.); (X.Z.); (M.T.A.); (T.W.); (Y.Z.); (L.Z.)
| | - Muhammad Toheed Akbar
- College of Food Science and Nutritional Engineering, China Agricultural University, National Center of Technology Innovation (Deep Processing of Highland Barley) in Food Industry, National Engineering Research Center for Fruit and Vegetable Processing, Beijing 100083, China; (Y.Z.); (B.C.); (X.Z.); (M.T.A.); (T.W.); (Y.Z.); (L.Z.)
- Department of Meat Science and Technology, University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Tong Wu
- College of Food Science and Nutritional Engineering, China Agricultural University, National Center of Technology Innovation (Deep Processing of Highland Barley) in Food Industry, National Engineering Research Center for Fruit and Vegetable Processing, Beijing 100083, China; (Y.Z.); (B.C.); (X.Z.); (M.T.A.); (T.W.); (Y.Z.); (L.Z.)
| | - Yiyun Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, National Center of Technology Innovation (Deep Processing of Highland Barley) in Food Industry, National Engineering Research Center for Fruit and Vegetable Processing, Beijing 100083, China; (Y.Z.); (B.C.); (X.Z.); (M.T.A.); (T.W.); (Y.Z.); (L.Z.)
| | - Li Zhi
- College of Food Science and Nutritional Engineering, China Agricultural University, National Center of Technology Innovation (Deep Processing of Highland Barley) in Food Industry, National Engineering Research Center for Fruit and Vegetable Processing, Beijing 100083, China; (Y.Z.); (B.C.); (X.Z.); (M.T.A.); (T.W.); (Y.Z.); (L.Z.)
| | - Qun Shen
- College of Food Science and Nutritional Engineering, China Agricultural University, National Center of Technology Innovation (Deep Processing of Highland Barley) in Food Industry, National Engineering Research Center for Fruit and Vegetable Processing, Beijing 100083, China; (Y.Z.); (B.C.); (X.Z.); (M.T.A.); (T.W.); (Y.Z.); (L.Z.)
| |
Collapse
|
4
|
Cheng Q, Wang K, Lu R, Xiong Y, Luo X, Li X, Liu W, Wang J, Li Y, Yan J. Effect of white jade snail secretion on antioxidant capacity and intestinal microbial diversity in mouse model of acute gastric ulcer. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:1723-1731. [PMID: 37851602 DOI: 10.1002/jsfa.13059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 09/21/2023] [Accepted: 10/19/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND In the present work, acute gastric ulcer models were constructed by administering hydrochloric acid/ethanol. The mice ingested white jade snail secretion (WJSS) through gastric infusion. Ulcer areas in gastric tissue were recorded, and malondialdehyde (MDA) and superoxide dismutase (SOD) were also measured. Notably, high-throughput 16S rDNA analysis of intestinal flora and determination of amino acid composition in feces were performed to understand the effect of WJSS on model mice. RESULTS Compared with the control group, the ulcer area in the WJSS low-, medium- and high-concentration groups declined by 28.02%, 39.57% and 77.85%, respectively. MDA content decreased by 24.71%, 49.58% and 64.25%, and SOD relative enzyme activity fell by 28.19%, 43.37% and 9.60%, respectively. The amounts of amino acids in the low-, medium- and high-concentration groups were slightly lower, and probiotic bacteria such as Bacteroidetes and Lactobacillales increased in different-concentration WJSS groups. Adding WJSS contributes to the establishment of beneficial intestinal flora and the absorption of amino acids. CONCLUSION Our results showed that WJSS has a beneficial effect on inhibiting hydrochloric acid-ethanolic gastric ulcers, suggesting that WJSS has excellent potential as a novel anti-ulcer agent. Combined with ulcer area, MDA content, SOD content, gut probiotics and other indicators, a high concentration of WJSS had the best protective effect on acute gastric ulcer. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Qian Cheng
- Medical College, Guangxi University, Nanning, China
| | - Kaidi Wang
- Medical College, Guangxi University, Nanning, China
| | - Rui Lu
- Medical College, Guangxi University, Nanning, China
| | - Yi Xiong
- Guangxi Center for Animal Disease Control and Prevention, Nanning, China
| | - Xianqing Luo
- Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Buffalo Genetics, Breeding and Reproduction Technology, Ministry of Agriculture and Guangxi, Nanning, China
| | - Xian Li
- Medical College, Guangxi University, Nanning, China
| | - Wei Liu
- Medical College, Guangxi University, Nanning, China
| | - Jiayu Wang
- Medical College, Guangxi University, Nanning, China
| | - Yixiang Li
- Medical College, Guangxi University, Nanning, China
| | - Jianhua Yan
- Medical College, Guangxi University, Nanning, China
| |
Collapse
|
5
|
Shi B, Li H, He X. Advancing lifelong precision medicine for cardiovascular diseases through gut microbiota modulation. Gut Microbes 2024; 16:2323237. [PMID: 38411391 PMCID: PMC10900281 DOI: 10.1080/19490976.2024.2323237] [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: 12/19/2023] [Accepted: 02/21/2024] [Indexed: 02/28/2024] Open
Abstract
The gut microbiome is known as the tenth system of the human body that plays a vital role in the intersection between health and disease. The considerable inter-individual variability in gut microbiota poses both challenges and great prospects in promoting precision medicine in cardiovascular diseases (CVDs). In this review, based on the development, evolution, and influencing factors of gut microbiota in a full life circle, we summarized the recent advances on the characteristic alteration in gut microbiota in CVDs throughout different life stages, and depicted their pathological links in mechanism, as well as the highlight achievements of targeting gut microbiota in CVDs prevention, diagnosis and treatment. Personalized strategies could be tailored according to gut microbiota characteristics in different life stages, including gut microbiota-blood metabolites combined prediction and diagnosis, dietary interventions, lifestyle improvements, probiotic or prebiotic supplements. However, to fulfill the promise of a lifelong cardiovascular health, more mechanism studies should progress from correlation to causality and decipher novel mechanisms linking specific microbes and CVDs. It is also promising to use the burgeoning artificial intelligence and machine learning to target gut microbiota for developing diagnosis system and screening for new therapeutic interventions.
Collapse
Affiliation(s)
- Bozhong Shi
- Department of Cardiothoracic Surgery, Shanghai Children’s Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoyu Li
- Department of Cardiothoracic Surgery, Shanghai Children’s Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaomin He
- Department of Cardiothoracic Surgery, Shanghai Children’s Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center, National Children’s Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Campanella A, Bonfiglio C, Cuccaro F, Donghia R, Tatoli R, Giannelli G. High Adherence to a Mediterranean Alcohol-Drinking Pattern and Mediterranean Diet Can Mitigate the Harmful Effect of Alcohol on Mortality Risk. Nutrients 2023; 16:59. [PMID: 38201889 PMCID: PMC10780794 DOI: 10.3390/nu16010059] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/18/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Alcohol is a psychoactive substance with deleterious effects on human health and mortality. This study aims to investigate the joint associations between the Mediterranean Diet (MedDiet), alcohol- consumption patterns and mortality from the following: all causes, cardiovascular, neoplastic, the digestive system, and other causes. METHODS A sample of 3411 alcohol consumers aged ≥18 years was selected from two prospective cohort studies: the MICOL and NUTRIHEP Study. Cohorts were enrolled in 2005-2006, and followed up until December 2022, capturing data on alcohol consumption, diet, and mortality. Adherence to the MedDiet was measured by the relative Mediterranean score (rMED), and alcohol consumption by the Mediterranean Alcohol-drinking Pattern index (MADP). Statistical analyses included flexible parametric survival models and subdistribution hazard ratios, to consider different causes of death. RESULTS a significant increase in digestive-system (SHR 2.77, 95% CI 1.16; 63) and cancer mortality risk (SHR 2.25, 95% CI 1.08; 4.70) was observed among individuals with low adherence to the MADP. Low adherence to the Mediterranean pattern of alcohol consumption, combined with low adherence to the MedDiet, was associated with higher overall mortality (HR 2.29, 95% CI 1.04, 5.04), and, in particular, with higher mortality from digestive system diseases (SHR 4.38, 95% CI 1.22, 15.8). CONCLUSIONS This study suggests that deleterious effects of alcohol on mortality vary, depending on alcohol consumption patterns and dietary context. Higher adherence to the MedDiet appears to mitigate the adverse effects of moderate alcohol consumption, particularly for wine drinkers.
Collapse
Affiliation(s)
- Angelo Campanella
- National Institute of Gastroenterology—IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (C.B.); (R.D.); (R.T.); (G.G.)
| | - Caterina Bonfiglio
- National Institute of Gastroenterology—IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (C.B.); (R.D.); (R.T.); (G.G.)
| | | | - Rossella Donghia
- National Institute of Gastroenterology—IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (C.B.); (R.D.); (R.T.); (G.G.)
| | - Rossella Tatoli
- National Institute of Gastroenterology—IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (C.B.); (R.D.); (R.T.); (G.G.)
| | - Gianluigi Giannelli
- National Institute of Gastroenterology—IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (C.B.); (R.D.); (R.T.); (G.G.)
| |
Collapse
|
7
|
Liang T, Kota J, Williams KE, Saxena R, Gawrieh S, Zhong X, Zimmers TA, Chalasani N. Dynamic Alterations to Hepatic MicroRNA-29a in Response to Long-Term High-Fat Diet and EtOH Feeding. Int J Mol Sci 2023; 24:14564. [PMID: 37834011 PMCID: PMC10572557 DOI: 10.3390/ijms241914564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
MicroRNA-29a (miR-29a) is a well characterized fibro-inflammatory molecule and its aberrant expression is linked to a variety of pathological liver conditions. The long-term effects of a high-fat diet (HFD) in combination with different levels of EtOH consumption on miR-29a expression and liver pathobiology are unknown. Mice at 8 weeks of age were divided into five groups (calorie-matched diet plus water (CMD) as a control group, HFD plus water (HFD) as a liver disease group, HFD plus 2% EtOH (HFD + 2% E), HFD + 10% E, and HFD + 20% E as intervention groups) and fed for 4, 13, 26, or 39 weeks. At each time point, analyses were performed for liver weight/body weight (BW) ratio, AST/ALT ratio, as well as liver histology assessments, which included inflammation, estimated fat deposition, lipid area, and fibrosis. Hepatic miR-29a was measured and correlations with phenotypic traits were determined. Four-week feeding produced no differences between the groups on all collected phenotypic traits or miR-29a expression, while significant effects were observed after 13 weeks, with EtOH concentration-specific induction of miR-29a. A turning point for most of the collected traits was apparent at 26 weeks, and miR-29a was significantly down-regulated with increasing liver injury. Overall, miR-29a up-regulation was associated with a lower liver/BW ratio, fat deposition, inflammation, and fibrosis, suggesting a protective role of miR-29a against liver disease progression. A HFD plus increasing concentrations of EtOH produces progressive adverse effects on the liver, with no evidence of beneficial effects of low-dose EtOH consumption. Moreover, miR-29a up-regulation is associated with less severe liver injury.
Collapse
Affiliation(s)
- Tiebing Liang
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (K.E.W.); (S.G.); (N.C.)
| | - Janaiah Kota
- Ultragenyx Pharmaceuticals, Novato, CA 94949, USA;
| | - Kent E. Williams
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (K.E.W.); (S.G.); (N.C.)
| | - Romil Saxena
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Samer Gawrieh
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (K.E.W.); (S.G.); (N.C.)
| | - Xiaoling Zhong
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.Z.); (T.A.Z.)
| | - Teresa A. Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (X.Z.); (T.A.Z.)
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, USA
- Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, USA
| | - Naga Chalasani
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (K.E.W.); (S.G.); (N.C.)
| |
Collapse
|
8
|
Moreira-Júnior RE, Guimarães MADF, Etcheverria da Silva M, Maioli TU, Faria AMC, Brunialti-Godard AL. Animal model for high consumption and preference of ethanol and its interplay with high sugar and butter diet, behavior, and neuroimmune system. Front Nutr 2023; 10:1141655. [PMID: 37063320 PMCID: PMC10097969 DOI: 10.3389/fnut.2023.1141655] [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: 01/10/2023] [Accepted: 03/10/2023] [Indexed: 03/31/2023] Open
Abstract
Introduction Mechanisms that dictate the preference for ethanol and its addiction are not only restricted to the central nervous system (CNS). An increasing body of evidence has suggested that abusive ethanol consumption directly affects the immune system, which in turn interacts with the CNS, triggering neuronal responses and changes, resulting in dependence on the drug. It is known that neuroinflammation and greater immune system reactivity are observed in behavioral disorders and that these can regulate gene transcription. However, there is little information about these findings of the transcriptional profile of reward system genes in high consumption and alcohol preference. In this regard, there is a belief that, in the striatum, an integrating region of the brain reward system, the interaction of the immune response and the transcriptional profile of the Lrrk2 gene that is associated with loss of control and addiction to ethanol may influence the alcohol consumption and preference. Given this information, this study aimed to assess whether problematic alcohol consumption affects the transcriptional profile of the Lrrk2 gene, neuroinflammation, and behavior and whether these changes are interconnected. Methods An animal model developed by our research group has been used in which male C57BL/6 mice and knockouts for the Il6 and Nfat genes were subjected to a protocol of high fat and sugar diet intake and free choice of ethanol in the following stages: Stage 1 (T1)-Dietary treatment, for 8 weeks, in which the animals receive high-calorie diet, High Sugar and Butter (HSB group), or standard diet, American Institute of Nutrition 93-Growth (AIN93G group); and Stage 2 (T2)-Ethanol consumption, in which the animals are submitted, for 4 weeks, to alcohol within the free choice paradigm, being each of them divided into 10 groups, four groups continued with the same diet and in the other six the HSB diet is substituted by the AIN93G diet. Five groups had access to only water, while the five others had a free choice between water and a 10% ethanol solution. The weight of the animals was evaluated weekly and the consumption of water and ethanol daily. At the end of the 12-week experiment, anxiety-like behavior was evaluated by the light/dark box test; compulsive-like behavior by Marble burying, transcriptional regulation of genes Lrrk2, Tlr4, Nfat, Drd1, Drd2, Il6, Il1β, Il10, and iNOS by RT-qPCR; and inflammatory markers by flow cytometry. Animals that the diet was replaced had an ethanol high preference and consumption. Results and discussion We observed that high consumption and preference for ethanol resulted in (1) elevation of inflammatory cells in the brain, (2) upregulation of genes associated with cytokines (Il6 and Il1β) and pro-inflammatory signals (iNOS and Nfat), downregulation of anti-inflammatory cytokine (Il10), dopamine receptor (Drd2), and the Lrrk2 gene in the striatum, and (3) behavioral changes such as decreased anxiety-like behavior, and increased compulsive-like behavior. Our findings suggest that interactions between the immune system, behavior, and transcriptional profile of the Lrrk2 gene influence the ethanol preferential and abusive consumption.
Collapse
Affiliation(s)
- Renato Elias Moreira-Júnior
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Andrade de Freitas Guimarães
- Laboratório de Imunobiologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Miguel Etcheverria da Silva
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tatiani Uceli Maioli
- Laboratório de Imunobiologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Maria Caetano Faria
- Laboratório de Imunobiologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Lúcia Brunialti-Godard
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
9
|
Wu Z, Du Z, Tian Y, Liu M, Zhu K, Zhao Y, Wang H. Inulin accelerates weight loss in obese mice by regulating gut microbiota and serum metabolites. Front Nutr 2022; 9:980382. [PMID: 36245535 PMCID: PMC9554005 DOI: 10.3389/fnut.2022.980382] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
Several studies indicated that the gut microbiota might participate in the beneficial effect of inulin on obesity. However, the mechanisms involved were still largely unknown. Sixteen high-fat diets (HFDs)-induced obese C57BL/6 mice were converted to a normal diet and then randomized into two groups, OND (obese mice + normal diet) group gavage-fed for 10 weeks with normal saline and ONDI (obese mice + normal diet + inulin) group with inulin at 10 g/kg/day. The body weight of HFD-induced obese mice showed different degrees of decrease in both groups. However, the ONDI group lost more weight and returned to normal earlier. Compared to the OND group, inulin supplementation significantly shifted the composition and structure of gut microbiota, such as higher α diversity. The β diversity analysis also confirmed the changes in gut microbiota composition between groups. At the genus level, the abundance of Alistipes was considerably increased, and it was significantly correlated with inulin supplementation (r = 0.72, P = 0.002). Serum metabolite levels were distinctly altered after inulin supplementation, and 143 metabolites were significantly altered in the ONDI group. Among them, indole-3-acrylic acid level increased more than 500-fold compared to the OND group. It was also strongly positive correlation with Alistipes (r = 0.72, P = 0.002) and inulin supplementation (r = 0.99, P = 9.2e−13) and negatively correlated with obesity (r = −0.72, P = 0.002). In conclusion, inulin supplementation could accelerate body weight loss in obese mice by increasing Alistipes and indole-3-acrylic acid level.
Collapse
Affiliation(s)
- Zeang Wu
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Zhenzhu Du
- Analysis and Testing Center, Shihezi University, Shihezi, China
| | - Yuanyuan Tian
- School of Medicine, Shihezi University, Shihezi, China
| | - Miao Liu
- School of Medicine, Shihezi University, Shihezi, China
| | - Kailong Zhu
- School of Medicine, Shihezi University, Shihezi, China
| | - Yufan Zhao
- School of Medicine, Shihezi University, Shihezi, China
| | - Haixia Wang
- School of Medicine, Shihezi University, Shihezi, China
- *Correspondence: Haixia Wang,
| |
Collapse
|
10
|
Kang J, Liu Y, Chen X, Xu F, Xiong W, Li X. Shifts of Antibiotic Resistomes in Soil Following Amendments of Antibiotics-Contained Dairy Manure. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:10804. [PMID: 36078515 PMCID: PMC9517759 DOI: 10.3390/ijerph191710804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 06/15/2023]
Abstract
Dairy manure is a nutrition source for cropland soils and also simultaneously serves as a contamination source of antibiotic resistance genes (ARGs). In this study, five classes of antibiotics including aminoglycosides, beta-lactams, macrolides, sulfonamides, and tetracyclines, were spiked in dairy manure and incubated with soil for 60 days. The high throughput qPCR and 16S rRNA amplicon sequencing were used to detect temporal shifts of the soil antibiotic resistomes and bacterial community. Results indicated dairy manure application increased the ARG abundance by 0.5-3.7 times and subtype numbers by 2.7-3.7 times and changed the microbial community structure in soils. These effects were limited to the early incubation stage. Selection pressure was observed after the addition of sulfonamides. Bacterial communities played an important role in the shifts of ARG profiles and accounted for 44.9% of the resistome variation. The incubation period, but not the different antibiotic treatments, has a strong impact on the bacteria community. Firmicutes and Bacteroidetes were the dominant bacterial hosts for individual ARGs. This study advanced our understanding of the effect of dairy manure and antibiotics on the antibiotic resistome in soils and provided a reference for controlling ARG dissemination from dairy farms to the environment.
Collapse
Affiliation(s)
- Jijun Kang
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yiming Liu
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xiaojie Chen
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Fei Xu
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Wenguang Xiong
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutic Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Xiubo Li
- Key Laboratory of Animal Antimicrobial Resistance Surveillance, Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
11
|
Abstract
Alcohol is part of the usual diet of millions of individuals worldwide. However, not all individuals who drink alcohol experience the same effects, nor will everyone develop an alcohol use disorder. Here we propose that the intestinal microbiota (IMB) helps explain the different consumption patterns of alcohol among individuals. 507 humans participated in this study and alcohol consumption and IMB composition were analyzed. On the other hand, in 80 adult male Wistar rats, behavioral tests, alcohol intoxication, fecal transplantation, administration of antibiotics and collection of fecal samples were performed. For identification and relative quantification of bacterial taxa was used the bacterial 16 S ribosomal RNA gene. In humans, we found that heavy episodic drinking is associated with a specific stool type phenotype (type 1, according to Bristol Stool Scale; p < 0.05) and with an increase in the abundance of Actinobacteria (p < 0.05). Next, using rats, we demonstrate that the transfer of IMB from alcohol-intoxicated animals causes an increase in voluntary alcohol consumption in transplant-recipient animals (p < 0.001). The relative quantification data indicate that the genus Porphyromonas could be associated with the effect on voluntary alcohol consumption. We also show that gut microbiota depletion by antibiotics administration causes a reduction in alcohol consumption (p < 0.001) and altered the relative abundance of relevant phyla such as Firmicutes, Bacteroidetes or Cyanobacteria (p < 0.05), among others. Benjamini-Hochberg false discovery rate (FDR) correction was performed for multiple comparisons. These studies reveal some of the consequences of alcohol on the IMB and provide evidence that manipulation of IMB may alter voluntary alcohol consumption.
Collapse
|
12
|
Schugar RC, Gliniak CM, Osborn LJ, Massey W, Sangwan N, Horak A, Banerjee R, Orabi D, Helsley RN, Brown AL, Burrows A, Finney C, Fung KK, Allen FM, Ferguson D, Gromovsky AD, Neumann C, Cook K, McMillan A, Buffa JA, Anderson JT, Mehrabian M, Goudarzi M, Willard B, Mak TD, Armstrong AR, Swanson G, Keshavarzian A, Garcia-Garcia JC, Wang Z, Lusis AJ, Hazen SL, Brown JM. Gut microbe-targeted choline trimethylamine lyase inhibition improves obesity via rewiring of host circadian rhythms. eLife 2022; 11:e63998. [PMID: 35072627 PMCID: PMC8813054 DOI: 10.7554/elife.63998] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity has repeatedly been linked to reorganization of the gut microbiome, yet to this point obesity therapeutics have been targeted exclusively toward the human host. Here, we show that gut microbe-targeted inhibition of the trimethylamine N-oxide (TMAO) pathway protects mice against the metabolic disturbances associated with diet-induced obesity (DIO) or leptin deficiency (Lepob/ob). Small molecule inhibition of the gut microbial enzyme choline TMA-lyase (CutC) does not reduce food intake but is instead associated with alterations in the gut microbiome, improvement in glucose tolerance, and enhanced energy expenditure. We also show that gut microbial CutC inhibition is associated with reorganization of host circadian control of both phosphatidylcholine and energy metabolism. This study underscores the relationship between microbe and host metabolism and provides evidence that gut microbe-derived trimethylamine (TMA) is a key regulator of the host circadian clock. This work also demonstrates that gut microbe-targeted enzyme inhibitors have potential as anti-obesity therapeutics.
Collapse
Affiliation(s)
- Rebecca C Schugar
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Christy M Gliniak
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Lucas J Osborn
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - William Massey
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Naseer Sangwan
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Anthony Horak
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Rakhee Banerjee
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Danny Orabi
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Robert N Helsley
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Amanda L Brown
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Amy Burrows
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Chelsea Finney
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Kevin K Fung
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Frederick M Allen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Daniel Ferguson
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Anthony D Gromovsky
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Chase Neumann
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Kendall Cook
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Amy McMillan
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Jennifer A Buffa
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - James T Anderson
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Margarete Mehrabian
- Departments of Medicine, Microbiology, and Human Genetics, University of California, Los AngelesLos AngelesUnited States
| | - Maryam Goudarzi
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Proteomics and Metabolomics Core, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Belinda Willard
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Proteomics and Metabolomics Core, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Tytus D Mak
- Mass Spectrometry Data Center, National Institute of Standards and Technology (NIST)GaithersburgUnited States
| | - Andrew R Armstrong
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical CenterChicagoUnited States
| | - Garth Swanson
- Departments of Medicine, Microbiology, and Human Genetics, University of California, Los AngelesLos AngelesUnited States
| | - Ali Keshavarzian
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical CenterChicagoUnited States
| | | | - Zeneng Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Aldons J Lusis
- Departments of Medicine, Microbiology, and Human Genetics, University of California, Los AngelesLos AngelesUnited States
| | - Stanley L Hazen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
- Department of Cardiovascular Medicine, Heart Vascular and Thoracic Institute, Cleveland ClinicClevelandUnited States
| | - Jonathan Mark Brown
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland ClinicClevelandUnited States
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| |
Collapse
|
13
|
Zhao Q, Hou D, Fu Y, Xue Y, Guan X, Shen Q. Adzuki Bean Alleviates Obesity and Insulin Resistance Induced by a High-Fat Diet and Modulates Gut Microbiota in Mice. Nutrients 2021; 13:nu13093240. [PMID: 34579118 PMCID: PMC8466346 DOI: 10.3390/nu13093240] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/11/2021] [Accepted: 09/14/2021] [Indexed: 12/23/2022] Open
Abstract
Adzuki bean consumption has many health benefits, but its effects on obesity and regulating gut microbiota imbalances induced by a high-fat diet (HFD) have not been thoroughly studied. Mice were fed a low-fat diet, a HFD, and a HFD supplemented with 15% adzuki bean (HFD-AB) for 12 weeks. Adzuki bean supplementation significantly reduced obesity, lipid accumulation, and serum lipid and lipopolysaccharide (LPS) levels induced by HFD. It also mitigated liver function damage and hepatic steatosis. In particular, adzuki bean supplementation improved glucose homeostasis by increasing insulin sensitivity. In addition, it significantly reversed HFD-induced gut microbiota imbalances. Adzuki bean significantly reduced the ratio of Firmicutes/Bacteroidetes (F/B); enriched the occurrence of Bifidobacterium, Prevotellaceae, Ruminococcus_1, norank_f_Muribaculaceae, Alloprevotella, Muribaculum, Turicibacter, Lachnospiraceae_NK4A136_group, and Lachnoclostridium; and returned HFD-dependent taxa (Desulfovibrionaceae, Bilophila, Ruminiclostridium_9, Blautia, and Ruminiclostridium) back to normal status. PICRUSt2 analysis showed that the changes in gut microbiota induced by adzuki bean supplementation may be associated with the metabolism of carbohydrates, lipids, sulfur, and cysteine and methionine; and LPS biosynthesis; and valine, leucine, and isoleucine degradation.
Collapse
Affiliation(s)
- Qingyu Zhao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Q.Z.); (D.H.); (Y.F.); (Y.X.)
- National Center of Technology Innovation (Deep Processing of Highland Barley) in Food Industry, Beijing 100083, China
- National Engineering Research Center for Fruit and Vegetable Processing, Beijing 100083, China
- Key Laboratory of Plant Protein and Grain Processing, Beijing 100083, China
| | - Dianzhi Hou
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Q.Z.); (D.H.); (Y.F.); (Y.X.)
- National Center of Technology Innovation (Deep Processing of Highland Barley) in Food Industry, Beijing 100083, China
- National Engineering Research Center for Fruit and Vegetable Processing, Beijing 100083, China
- Key Laboratory of Plant Protein and Grain Processing, Beijing 100083, China
| | - Yongxia Fu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Q.Z.); (D.H.); (Y.F.); (Y.X.)
- National Center of Technology Innovation (Deep Processing of Highland Barley) in Food Industry, Beijing 100083, China
- National Engineering Research Center for Fruit and Vegetable Processing, Beijing 100083, China
- Key Laboratory of Plant Protein and Grain Processing, Beijing 100083, China
| | - Yong Xue
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Q.Z.); (D.H.); (Y.F.); (Y.X.)
- National Center of Technology Innovation (Deep Processing of Highland Barley) in Food Industry, Beijing 100083, China
- National Engineering Research Center for Fruit and Vegetable Processing, Beijing 100083, China
- Key Laboratory of Plant Protein and Grain Processing, Beijing 100083, China
| | - Xiao Guan
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China;
| | - Qun Shen
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Q.Z.); (D.H.); (Y.F.); (Y.X.)
- National Center of Technology Innovation (Deep Processing of Highland Barley) in Food Industry, Beijing 100083, China
- National Engineering Research Center for Fruit and Vegetable Processing, Beijing 100083, China
- Key Laboratory of Plant Protein and Grain Processing, Beijing 100083, China
- Correspondence: ; Tel.: +86-010-6273-7524
| |
Collapse
|
14
|
Silva CBP, Elias-Oliveira J, McCarthy CG, Wenceslau CF, Carlos D, Tostes RC. Ethanol: striking the cardiovascular system by harming the gut microbiota. Am J Physiol Heart Circ Physiol 2021; 321:H275-H291. [PMID: 34142885 DOI: 10.1152/ajpheart.00225.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ethanol consumption represents a significant public health problem, and excessive ethanol intake is a risk factor for cardiovascular disease (CVD), one of the leading causes of death and disability worldwide. The mechanisms underlying the effects of ethanol on the cardiovascular system are complex and not fully comprehended. The gut microbiota and their metabolites are indispensable symbionts essential for health and homeostasis and therefore, have emerged as potential contributors to ethanol-induced cardiovascular system dysfunction. By mechanisms that are not completely understood, the gut microbiota modulates the immune system and activates several signaling pathways that stimulate inflammatory responses, which in turn, contribute to the development and progression of CVD. This review summarizes preclinical and clinical evidence on the effects of ethanol in the gut microbiota and discusses the mechanisms by which ethanol-induced gut dysbiosis leads to the activation of the immune system and cardiovascular dysfunction. The cross talk between ethanol consumption and the gut microbiota and its implications are detailed. In summary, an imbalance in the symbiotic relationship between the host and the commensal microbiota in a holobiont, as seen with ethanol consumption, may contribute to CVD. Therefore, manipulating the gut microbiota, by using antibiotics, probiotics, prebiotics, and fecal microbiota transplantation might prove a valuable opportunity to prevent/mitigate the deleterious effects of ethanol and improve cardiovascular health and risk prevention.
Collapse
Affiliation(s)
- Carla B P Silva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jefferson Elias-Oliveira
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Cameron G McCarthy
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Camilla F Wenceslau
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Daniela Carlos
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
15
|
Moreira Júnior RE, de Carvalho LM, Dos Reis DC, Cassali GD, Faria AMC, Maioli TU, Brunialti-Godard AL. Diet-induced obesity leads to alterations in behavior and gut microbiota composition in mice. J Nutr Biochem 2021; 92:108622. [PMID: 33705942 DOI: 10.1016/j.jnutbio.2021.108622] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/30/2020] [Accepted: 02/04/2021] [Indexed: 12/17/2022]
Abstract
The high prevalence of obesity and associated metabolic disorders are one of the major public health problems worldwide. Among the main causal factors of obesity, excessive consumption of food rich in sugar and fat stands out due to its high energy density. The regulation of food intake relies on hypothalamic control by the action of several neuropeptides. Excessive consumption of hypercaloric diets has impact in the behavior and in the gut microbiota. In the present study, we used a high-sugar and fat (HSB) diet for 12 weeks to induce obesity in C57BL/6 mice and to investigate its effects on the gut microbiota, hypothalamic peptides, and behavior. We hypothesize that chronic consumption of HSB diet can change the behavior. Additionally, we also hypothesize that changes in gut microbiota can be associated with changes in the transcriptional regulation of hypothalamic peptides and behavior. To evaluate the gut microbiota, we performed the sequencing of 16S rRNA gene, which demonstrate that HSB diet modulates the gut microbiota with an increase in the Firmicutes and Actinobacteria phylum and a decrease of Bacteroidetes phylum. The real time qPCR revealed that HSB-fed mice presented changes in the transcriptional regulation of hypothalamic neuropeptides genes such as Npy, Gal and Galr1. The Marble-burying and Light/dark box tests also showed an alteration in anxiety and impulsive behaviors for the HSB-fed mice. Our data provides evidence that obesity induced by HSB diet consumption is associated with alterations in gut microbiota and behavior, highlighting the multifactorial characteristics of this disease.
Collapse
Affiliation(s)
- Renato Elias Moreira Júnior
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luana Martins de Carvalho
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Diego Carlos Dos Reis
- Laboratório de Patologia Comparada, Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Geovanni Dantas Cassali
- Laboratório de Patologia Comparada, Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Ana Maria Caetano Faria
- Departmento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tatiani Uceli Maioli
- Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Ana Lúcia Brunialti-Godard
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
16
|
LeBrun ES, Nighot M, Dharmaprakash V, Kumar A, Lo CC, Chain PSG, Ma TY. The Gut Microbiome and Alcoholic Liver Disease: Ethanol Consumption Drives Consistent and Reproducible Alteration in Gut Microbiota in Mice. Life (Basel) 2020; 11:7. [PMID: 33374112 PMCID: PMC7823357 DOI: 10.3390/life11010007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/22/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023] Open
Abstract
Phenotypic health effects, both positive and negative, have been well studied in association with the consumption of alcohol in humans as well as several other mammals including mice. Many studies have also associated these same health effects and phenotypes to specific members of gut microbiome communities. Here we utilized a chronic plus binge ethanol feed model (Gao-binge model) to explore microbiome community changes across three independent experiments performed in mice. We found significant and reproducible differences in microbiome community assemblies between ethanol-treated mice and control mice on the same diet absent of ethanol. We also identified significant differences in gut microbiota occurring temporally with ethanol treatment. Peak shift in communities was observed 4 days after the start of daily alcohol consumption. We quantitatively identified many of the bacterial genera indicative of these ethanol-induced shifts including 20 significant genera when comparing ethanol treatments with controls and 14 significant genera based on temporal investigation. Including overlap of treatment with temporal shifts, we identified 25 specific genera of interest in ethanol treatment microbiome shifts. Shifts coincide with observed presentation of fatty deposits in the liver tissue, i.e., Alcoholic Liver Disease-associated phenotype. The evidence presented herein, derived from three independent experiments, points to the existence of a common, reproducible, and characterizable "mouse ethanol gut microbiome".
Collapse
Affiliation(s)
- Erick S. LeBrun
- Biosecurity and Public Health, Los Alamos National Laboratory, Los Alamos, NM 87545, USA; (E.S.L.); (A.K.); (C.-C.L.)
| | - Meghali Nighot
- Department of Medicine, Division of Gastroenterology and Hepatology, Penn State College of Medicine, Hershey, PA 17033, USA; (M.N.); (V.D.)
| | - Viszwapriya Dharmaprakash
- Department of Medicine, Division of Gastroenterology and Hepatology, Penn State College of Medicine, Hershey, PA 17033, USA; (M.N.); (V.D.)
| | - Anand Kumar
- Biosecurity and Public Health, Los Alamos National Laboratory, Los Alamos, NM 87545, USA; (E.S.L.); (A.K.); (C.-C.L.)
| | - Chien-Chi Lo
- Biosecurity and Public Health, Los Alamos National Laboratory, Los Alamos, NM 87545, USA; (E.S.L.); (A.K.); (C.-C.L.)
| | - Patrick S. G. Chain
- Biosecurity and Public Health, Los Alamos National Laboratory, Los Alamos, NM 87545, USA; (E.S.L.); (A.K.); (C.-C.L.)
| | - Thomas Y. Ma
- Department of Medicine, Division of Gastroenterology and Hepatology, Penn State College of Medicine, Hershey, PA 17033, USA; (M.N.); (V.D.)
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| |
Collapse
|
17
|
Introducing Murine Microbiome Database (MMDB): A Curated Database with Taxonomic Profiling of the Healthy Mouse Gastrointestinal Microbiome. Microorganisms 2019; 7:microorganisms7110480. [PMID: 31652812 PMCID: PMC6920994 DOI: 10.3390/microorganisms7110480] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/17/2019] [Accepted: 10/21/2019] [Indexed: 12/15/2022] Open
Abstract
The gut microbiota modulates overall metabolism, the immune system and brain development of the host. The majority of mammalian gut microbiota consists of bacteria. Among various model animals, the mouse has been most widely used in pre-clinical biological experiments. The significant compositional differences in taxonomic profiles among different mouse strains due to gastrointestinal locations, genotypes and vendors have been well documented. However, details of such variations are yet to be elucidated. This study compiled and analyzed 16S rRNA gene-based taxonomic profiles of 554 healthy mouse samples from 14 different projects to construct a comprehensive database of the microbiome of a healthy mouse gastrointestinal tract. The database, named Murine Microbiome Database, should provide researchers with useful taxonomic information and better biological insight about how each taxon, such as genus and species, is associated with locations in the gastrointestinal tract, genotypes and vendors. The database is freely accessible over the Internet.
Collapse
|