1
|
Silva CLM, Puthanveetil PN, Oliveira SD. Liver bypass in the development of pathogen-associated pulmonary vascular disease: contribution of mesocaval and portosystemic shunts. Am J Physiol Gastrointest Liver Physiol 2025; 328:G791-G800. [PMID: 40323787 DOI: 10.1152/ajpgi.00409.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/18/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Portosystemic and mesocaval shunts are aberrant vascular connections that bypass hepatic detoxification process, directly linking the portal to the systemic circulation. These shunts, whether congenital or acquired, might play a pivotal role in the pathogenesis of systemic inflammatory diseases, such as schistosomiasis-associated pulmonary hypertension (Sch-PH) by facilitating the dissemination of pathogen-derived eggs and antigens from the gut and mesentery into the lungs. Beyond the translocation of Schistosoma mansoni eggs, emerging evidence implicates that gut-lung microbiome dysbiosis contributes to the development of pulmonary hypertension (PH) in the preclinical animal model of Sch-PH. Sch-PH emerges as a chronic complication of schistosomiasis and evolves silently, progressively increasing the mean pulmonary arterial pressure and vascular resistance, leading to right heart hypertrophy, failure, and significant morbidity and mortality. Chronic schistosomiasis is often linked to the development of portal hypertension, which significantly contributes to the formation of the porto/mesocaval shunt as a compensatory response that can have far-reaching implications on pulmonary vascular physiology. In addition, portal hypertension compromises the integrity of the intestinal barrier, exacerbating peritoneal and mesenteric inflammation, potentially facilitating microbial and metabolite entrance into the systemic circulation. This article briefly discusses the mechanisms by which porto/mesocaval shunts contribute to PH, especially Group I PH, focusing on the interplay between portosystemic shunting, microbial translocation, and systemic dissemination of proinflammatory metabolites.
Collapse
Affiliation(s)
- Claudia Lucia Martins Silva
- Biochemical and Molecular Pharmacology Laboratory, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Prasanth N Puthanveetil
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Illinois, United States
- Department of Pharmacology, College of Graduate Studies, Midwestern University, Downers Grove, Illinois, United States
| | - Suellen Darc Oliveira
- Vascular Immunobiology Laboratory, Department of Anesthesiology, College of Medicine, University of Illinois Chicago, Chicago, Illinois, United States
- Vascular Immunobiology Laboratory, Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, Illinois, United States
| |
Collapse
|
2
|
Shen H, Liangpunsakul S, Iwakiri Y, Szabo G, Wang H. Immunological mechanisms and emerging therapeutic targets in alcohol-associated liver disease. Cell Mol Immunol 2025:10.1038/s41423-025-01291-w. [PMID: 40399593 DOI: 10.1038/s41423-025-01291-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Accepted: 04/19/2025] [Indexed: 05/23/2025] Open
Abstract
Alcohol-associated liver disease (ALD) is a major global health challenge, with inflammation playing a central role in its progression. As inflammation emerges as a critical therapeutic target, ongoing research aims to unravel its underlying mechanisms. This review explores the immunological pathways of ALD, highlighting the roles of immune cells and their inflammatory mediators in disease onset and progression. We also examine the complex interactions between inflammatory cells and non-parenchymal liver cells, as well as their crosstalk with extra-hepatic organs, including the gut, adipose tissue, and nervous system. Furthermore, we summarize current clinical research on anti-inflammatory therapies and discuss promising therapeutic targets. Given the heterogeneity of ALD-associated inflammation, we emphasize the need for precision medicine to optimize treatment strategies and improve patient outcomes.
Collapse
Affiliation(s)
- Haiyuan Shen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - Yasuko Iwakiri
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Gyongyi Szabo
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China.
| |
Collapse
|
3
|
Li X, Ni Z, Shi W, Zhao K, Zhang Y, Liu L, Wang Z, Chen J, Yu Z, Gao X, Qin Y, Zhao J, Peng W, Shi J, Kosten TR, Lu L, Su L, Xue Y, Sun H. Nitrate ameliorates alcohol-induced cognitive impairment via oral microbiota. J Neuroinflammation 2025; 22:106. [PMID: 40234914 PMCID: PMC12001487 DOI: 10.1186/s12974-025-03439-x] [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: 01/29/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025] Open
Abstract
Alcohol use is associated with cognitive impairment and dysregulated inflammation. Oral nitrate may benefit cognitive impairment in aging through altering the oral microbiota. Similarly, the beneficial effects of nitrate on alcohol-induced cognitive decline and the roles of the oral microbiota merit investigation. Here we found that nitrate supplementation effectively mitigated cognitive impairment induced by chronic alcohol exposure in mice, reducing both systemic and neuroinflammation. Furthermore, nitrate restored the dysbiosis of the oral microbiota caused by alcohol consumption. Notably, removing the oral microbiota led to a subsequent loss of the beneficial effects of nitrate. Oral microbiota from donor alcohol use disordered humans who had been taking the nitrate intervention were transplanted into germ-free mice which then showed increased cognitive function and reduced neuroinflammation. Finally, we examined 63 alcohol drinkers with varying levels of cognitive impairment and found that lower concentrations of nitrate metabolism-related bacteria were associated with higher cognitive impairment and lower nitrate levels in plasma. These findings highlight the protective role of nitrate against alcohol-induced cognition impairment and neuroinflammation and suggest that the oral microbiota associated with nitrate metabolism and brain function may form part of a "microbiota-mouth-brain axis".
Collapse
Affiliation(s)
- Xiangxue Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Zhaojun Ni
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Weixiong Shi
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, 100021, China
| | - Kangqing Zhao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Yanjie Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Lina Liu
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Zhong Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Jie Chen
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Zhoulong Yu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Xuejiao Gao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Ying Qin
- Addiction Medicine Department, The Second People's Hospital of Guizhou Province, Guizhou, China
| | - Jingwen Zhao
- Addiction Medicine Department, The Second People's Hospital of Guizhou Province, Guizhou, China
| | - Wenjuan Peng
- Addiction Medicine Department, The Second People's Hospital of Guizhou Province, Guizhou, China
| | - Jie Shi
- National Institute On Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, China
| | - Thomas R Kosten
- Department of Psychiatry, Pharmacology, Neuroscience, Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China
- National Institute On Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, China
| | - Lei Su
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, 100021, China.
| | - Yanxue Xue
- National Institute On Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, 100191, China.
- Chinese Institute for Brain Research, Beijing, China.
| | - Hongqiang Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), No.51 Huayuan North Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
4
|
Zhang Y, Li H, Li B, Li Y, Chai X, Li S, Xue X, Li H, Zhao Y, Tang Y, Yin B, Zhao P, Li E, Feng P. Dachaihu decoction ameliorates abnormal behavior by regulating gut microbiota in rats with propionic acid-induced autism. Front Microbiol 2025; 16:1535451. [PMID: 40018671 PMCID: PMC11867326 DOI: 10.3389/fmicb.2025.1535451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/23/2025] [Indexed: 03/01/2025] Open
Abstract
Background Autism spectrum disorder (ASD) is an early-onset neurodevelopmental disorder, usually accompanied by gut microbiota dysregulation. Gut microbiota homeostasis is considered effective for ASD. Reportedly, Dachaihu decoction (DCHD) can efficiently regulate gut microbiota and inflammation. However, the mechanisms underlying the effects of DCHD in the treatment of ASD remain unclear. Objective This study investigated the potential effects and mechanisms of DCHD in treating ASD. Methods In the animal experiment, propionic acid was administered to construct an ASD rat model. The ASD rats were treated with DCHD, and the efficacy was assessed using the behavioral detections, such as open field test, elevated plus maze test, novel object recognition test. Additionally, the levels of IL-6, TNF-α, IL-10, T-SOD, MDA, GSH and CAT were determined using kits, and histological staining was used to evaluate brain morphology. Moreover, tight junction proteins (ZO-1 and occludin) expression levels were evaluated using RT-qPCR, whereas Iba1 expression level was assessed by immunofluorescence staining. The 16S rRNA sequencing and metabolomic analysis of feces revealed the potential targets of DCHD against ASD. In a small human trail, the clinical scales ADOS-2 and Autism Behavior Checklist (ABC) assessed autism severity. Gastrointestinal problems and brain function were evaluated based on food intolerance and event-related potential, respectively. Results DCHD significantly improved autism-like behaviors and increased antioxidant enzyme activity, decreased inflammation and enhanced the intestinal barrier by the animal experiment. Furthermore, the DCHD treatment altered the gut microbiota profile, with increased probiotics Adlercreutzia, Parvibacter, Turicibacter, and Christensenellaceae. Further, DCHD increased the beneficial metabolite indole-3-acetate and decreased the cognitive impairment-related metabolites asymmetric dimethylarginine and homogentisic acid. Meanwhile, the small clinical trial revealed that DCHD significantly alleviated the core symptoms of ASD, with decreased ADOS-2 and ABC scale scores. DCHD also decreased the levels of specific egg white/yolk and milk IgG antibodies and shortened the MMN and P3b latencies. Conclusion This study demonstrated that DCHD may alleviate ASD via inhibiting oxidative stress, reducing inflammation, and modulating the gut microbiota in rats. Combined with human trial, DCHD may be a promising drug for treating ASD. This study provides a scientific rationale for treating mental disorders related to gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Yangyang Zhang
- Department of Children Rehabilitation, Henan Key Laboratory of Rehabilitation Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hang Li
- Henan Key Laboratory for Helicobacter Pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bolin Li
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yizhuang Li
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuejun Chai
- School of Basic Medical Science, The Shaanxi Key Laboratory of Brain Disorders, Xi’an Medical University, Xi’an, China
| | - Sheng Li
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Xia Xue
- Henan Key Laboratory for Helicobacter Pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Honglei Li
- Department of Children Rehabilitation, Henan Key Laboratory of Rehabilitation Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yonghong Zhao
- Department of Children Rehabilitation, Henan Key Laboratory of Rehabilitation Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Youcai Tang
- Department of Children Rehabilitation, Henan Key Laboratory of Rehabilitation Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Outstanding Overseas Scientists Chronic Liver Injury Workshop, Zhengzhou Key Laboratory of Metabolism-Associated Fatty Liver Disease, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Baoqi Yin
- Department of Children Rehabilitation, Henan Key Laboratory of Rehabilitation Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengju Zhao
- Department of Children Rehabilitation, Henan Key Laboratory of Rehabilitation Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Enyao Li
- Department of Children Rehabilitation, Henan Key Laboratory of Rehabilitation Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengya Feng
- Department of Children Rehabilitation, Henan Key Laboratory of Rehabilitation Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Helicobacter Pylori and Digestive Tract Microecology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
5
|
Qi Z, Liu J, Xu Y, Sun H, Qi X, Cong M, Zhang X, Yan Y, Liu T. Protective effects of phenylethanol glycosides from Cistanche tubulosa against ALD through modulating gut microbiota homeostasis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118925. [PMID: 39395767 DOI: 10.1016/j.jep.2024.118925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/14/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cistanche tubulosa (Schenk) Wight, a Chinese herbal medicine (Rou Cong Rong) with Xinjiang characteristics, was recorded in many medical books in ancient China and often used as a tonic medicine. Supported by the traditional Chinese medicine theory of "homology of liver and kidney," C. tubulosa (Schenk) Wight has many clinical applications in tonifying the kidney and protecting the liver. Modern pharmacological studies have also found that the protective effects of phenylethanol glycosides from C. tubulosa (Schenk) Wight (CPhGs) play an important role in ameliorating alcoholic liver injury. AIM OF THE STUDY We aimed to investigate whether CPhGs can enhance the therapeutic outcome of alcoholic liver disease (ALD) by targeting the "gut-liver axis," thus contributing to the knowledge of how Chinese herbs alleviate disease by influencing the gut microbiota. MATERIALS AND METHODS An ALD mouse model was established using the Lieber-DeCarli alcohol liquid diet, and the effects of CPhGs on the intestinal barrier and gut microbiota of ALD mice were investigated in a pseudo-sterile mouse model and fecal microbiota transplantation (FMT) mouse model. We fed female C57BL/6N mice with Lieber-DeCarli ethanol liquid diet, according to the NIAAA model. Animal experiment of long-term, ethanol diet intervention for 6W, and short-term for 11d. The FMT experiments were also performed. RESULTS CPhGs significantly improved ALD manifestations. ALD mice demonstrated significant gut microbiota dysbiosis and significantly abnormal proliferation of Allobaculum compared with the control diet group in long-term NIAAA mouse model (L-Pair). In mice that received the long-term intervention, the improvement in gut barrier function in the CPhGs-treated group was accompanied by a significant decrease in the abundance of Allobaculum and a significant increase in the abundance of Akkermansia. Furthermore, compared with the mouse were gavaged fecal microbiota from the long-term NIAAA mouse donors (FMT-EtOH), the number of goblet cells, abundance of Akkermansia, and the intestinal short-chain fatty acid concentrations were significantly increased in the mouse were gavaged fecal microbiota from high (700 mg/kg) doses of CPhGs orally in long-term NIAAA model donors (FMT-EtOH-H). Network analysis and species distribution results demonstrated that Akkermansia and Allobaculum were the genera with the highest abundances in the gut microbiota and that their interaction was related to propionic acid metabolism. CONCLUSIONS The results suggest that CPhGs exert a protective effect against ALD by modulating the abundance and composition of Akkermansia and Allobaculum in the intestine, maintaining the intestinal mucus balance, and safeguarding intestinal barrier integrity.
Collapse
Affiliation(s)
- Zhaoyao Qi
- School of Public Health, Xinjiang Medical University, Xinjiang, Urumqi, 830011, China.
| | - Jincun Liu
- School of Public Health, Xinjiang Medical University, Xinjiang, Urumqi, 830011, China.
| | - Yuanhui Xu
- School of Public Health, Xinjiang Medical University, Xinjiang, Urumqi, 830011, China.
| | - Hongguang Sun
- School of Public Health, Xinjiang Medical University, Xinjiang, Urumqi, 830011, China.
| | - Xinxin Qi
- School of Public Health, Xinjiang Medical University, Xinjiang, Urumqi, 830011, China.
| | - Meili Cong
- School of Public Health, Xinjiang Medical University, Xinjiang, Urumqi, 830011, China.
| | - Xinxuan Zhang
- School of Public Health, Xinjiang Medical University, Xinjiang, Urumqi, 830011, China.
| | - Yuxin Yan
- School of Public Health, Xinjiang Medical University, Xinjiang, Urumqi, 830011, China.
| | - Tao Liu
- School of Public Health, Xinjiang Medical University, Xinjiang, Urumqi, 830011, China.
| |
Collapse
|
6
|
Liu X, Liu H, Wu X, Zhao Z, Wang S, Wang H, Qin X. Xiaoyaosan against depression through suppressing LPS mediated TLR4/NLRP3 signaling pathway in "microbiota-gut-brain" axis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118683. [PMID: 39121928 DOI: 10.1016/j.jep.2024.118683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Depression impairs not only central nervous system, but also peripheral systems of the host. Gut microbiota have been proved to be involved in the pathogenesis of depression. Xiaoyaosan (XYS) has a history of over a thousand years in China for treating depression, dramatically alleviating anxiety, cognitive disorders, and especially gastrointestinal dysfunctions. Yet, it still just scratches the surface of the anti-depression mechanisms of XYS. AIM OF THE STUDY This study aims to elucidate the mechanism of actions of XYS from the perspective of "microbiota-gut-brain" axis. MATERIALS AND METHODS We firstly evaluated the effects of XYS on the macroscopic behaviors of depressed rats that induced by chronic unpredictable mild stress (CUMS). Secondly, the effects of XYS on intestinal homeostasis of depressed rats were revealed by using dysbacteriosis model. Subsequently, the underlying mechanisms were demonstrated by 16S rRNA gene sequencing technology and molecular biology methods. Finally, correlation analysis and visualization of the anti-depression effects of XYS were performed from the "microbiota - gut - brain" perspective. RESULTS Our data indicated that XYS ameliorated the depression-like symptoms of CUMS rats, partly depending on the presence of gut microbiota. Furthermore, we illustrated that XYS reversed CUMS-induced gut dysbiosis of depressed rats in terms of decreasing the Bacteroidetes/Firmicutes ratio and the abundances of Bacteroides, and Corynebacterium, while increasing the abundances of Lactobacillus and Adlercreutzia. The significant enrichment of Bacteroides and the level of lipopolysaccharides (LPS) suggested that depression damaged the immune responses and gut barrier. Mechanistically, XYS significantly down-regulated the expression levels of factors that involved in TLR4/NLRP3 signaling pathway in the colon and brain tissues of depressed rats. In addition, XYS significantly increased the levels of claudin 1 and ZO-1, showing that XYS positively maintained the integrity of gut and blood-brain barriers (BBB). CONCLUSION Our study offers insights into the anti-depression effects of XYS through a lens of "microbiota-TLR4/NLRP3 signaling pathway-barriers", providing a foundation for enhancing clinical efficiency and enriching drug selection, and contributing to our understanding of the mechanisms of traditional Chinese medicines (TCMs) in treating depression.
Collapse
Affiliation(s)
- Xiaojie Liu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, 030006, Shanxi, China.
| | - Huimin Liu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, 030006, Shanxi, China
| | - Xiaoling Wu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, 030006, Shanxi, China
| | - Ziyu Zhao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, 030006, Shanxi, China
| | - Senyan Wang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, 030006, Shanxi, China
| | - Huimin Wang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, 030006, Shanxi, China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, 030006, Shanxi, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No. 92, Wucheng Rd. Xiaodian Dist., Taiyuan, 030006, Shanxi, China
| |
Collapse
|
7
|
Raya Tonetti F, Eguileor A, Mrdjen M, Pathak V, Travers J, Nagy LE, Llorente C. Gut-liver axis: Recent concepts in pathophysiology in alcohol-associated liver disease. Hepatology 2024; 80:1342-1371. [PMID: 38691396 PMCID: PMC11801230 DOI: 10.1097/hep.0000000000000924] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/20/2024] [Indexed: 05/03/2024]
Abstract
The growing recognition of the role of the gut microbiome's impact on alcohol-associated diseases, especially in alcohol-associated liver disease, emphasizes the need to understand molecular mechanisms involved in governing organ-organ communication to identify novel avenues to combat alcohol-associated diseases. The gut-liver axis refers to the bidirectional communication and interaction between the gut and the liver. Intestinal microbiota plays a pivotal role in maintaining homeostasis within the gut-liver axis, and this axis plays a significant role in alcohol-associated liver disease. The intricate communication between intestine and liver involves communication between multiple cellular components in each organ that enable them to carry out their physiological functions. In this review, we focus on novel approaches to understanding how chronic alcohol exposure impacts the microbiome and individual cells within the liver and intestine, as well as the impact of ethanol on the molecular machinery required for intraorgan and interorgan communication.
Collapse
Affiliation(s)
| | - Alvaro Eguileor
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Marko Mrdjen
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH
| | - Vai Pathak
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jared Travers
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH
- Department of Gastroenterology and Hepatology, University Hospital, Cleveland OH
| | - Laura E Nagy
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland OH
| | - Cristina Llorente
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
8
|
Shen H, Zhang C, Zhang Q, Lv Q, Liu H, Yuan H, Wang C, Meng F, Guo Y, Pei J, Yu C, Tie J, Chen X, Yu H, Zhang G, Wang X. Gut microbiota modulates depressive-like behaviors induced by chronic ethanol exposure through short-chain fatty acids. J Neuroinflammation 2024; 21:290. [PMID: 39508236 PMCID: PMC11539449 DOI: 10.1186/s12974-024-03282-6] [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: 09/11/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Chronic ethanol exposure (CEE) is recognized as an important risk factor for depression, and the gut-brain axis has emerged as a key mechanism underlying chronic ethanol exposure-induced anxiety and depression-like behaviors. Short-chain fatty acids (SCFAs), which are the key metabolites generated by gut microbiota from insoluble dietary fiber, exert protective roles on the central nervous system, including the reduction of neuroinflammation. However, the link between gut microbial disturbances caused by chronic ethanol exposure, production of SCFAs, and anxiety and depression-like behaviors remains unclear. METHODS Initially, a 90-day chronic ethanol exposure model was established, followed by fecal microbiota transplantation model, which was supplemented with SCFAs via gavage. Anxiety and depression-like behaviors were determined by open field test, forced swim test, and elevated plus-maze. Serum and intestinal SCFAs levels were quantified using GC-MS. Changes in related indicators, including the intestinal barrier, intestinal inflammation, neuroinflammation, neurotrophy, and nerve damage, were detected using Western blotting, immunofluorescence, and Nissl staining. RESULTS Chronic ethanol exposure disrupted with gut microbial homeostasis, reduced the production of SCFAs, and led to anxiety and depression-like behaviors. Recipient mice transplanted with fecal microbiota that had been affected by chronic ethanol exposure exhibited impaired intestinal structure and function, low levels of SCFAs, intestinal inflammation, activation of neuroinflammation, a compromised blood-brain barrier, neurotrophic defects, alterations in the GABA system, anxiety and depression-like behaviors. Notably, the negative effects observed in these recipient mice were significantly alleviated through the supplementation of SCFAs. CONCLUSION SCFAs not only mitigate damage to intestinal structure and function but also alleviate various lesions in the central nervous system, such as neuroinflammation, and reduce anxiety and depression-like behaviors, which were triggered by transplantation with fecal microbiota that had been affected by chronic ethanol exposure, adding more support that SCFAs serve as a bridge between the gut and the brain.
Collapse
Affiliation(s)
- Hui Shen
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, P. R. China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, Liaoning, 110122, P. R. China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, 110122, P. R. China
| | - Chaoxu Zhang
- Department of Hematology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, P. R. China
| | - Qian Zhang
- Department of Health Statistics, School of Public Health, China Medical University, Shenyang, Liaoning, 110001, P. R. China
- Department of Reproductive Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning, 110016, P. R. China
| | - Qing Lv
- Department of Clinical Nutrition, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110032, P. R. China
| | - Hao Liu
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, P. R. China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, Liaoning, 110122, P. R. China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, 110122, P. R. China
| | - Huiya Yuan
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, Liaoning, 110122, P. R. China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, 110122, P. R. China
- Department of Forensic Analytical Toxicology, China Medical University School of Forensic Medicine, Shenyang, Liaoning, 110122, P. R. China
| | - Changliang Wang
- The People's Procuratorate of Liaoning Province Judicial Authentication Center, Shenyang, Liaoning, 110122, P. R. China
- Collaborative Laboratory of Intelligentized Forensic Science (CLIFS), Shenyang, Liaoning, 110032, P. R. China
| | - Fanyue Meng
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, P. R. China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, Liaoning, 110122, P. R. China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, 110122, P. R. China
| | - Yufu Guo
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, P. R. China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, Liaoning, 110122, P. R. China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, 110122, P. R. China
| | - Jiaxin Pei
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, P. R. China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, Liaoning, 110122, P. R. China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, 110122, P. R. China
| | - Chenyang Yu
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, P. R. China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, Liaoning, 110122, P. R. China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, 110122, P. R. China
| | - Jinming Tie
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, P. R. China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, Liaoning, 110122, P. R. China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, 110122, P. R. China
| | - Xiaohuan Chen
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, P. R. China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, Liaoning, 110122, P. R. China
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, 110122, P. R. China
| | - Hao Yu
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, P. R. China.
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, Liaoning, 110122, P. R. China.
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, 110122, P. R. China.
| | - Guohua Zhang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, P. R. China.
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, Liaoning, 110122, P. R. China.
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, 110122, P. R. China.
| | - Xiaolong Wang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, P. R. China.
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, Liaoning, 110122, P. R. China.
- China Medical University Center of Forensic Investigation, Shenyang, Liaoning, 110122, P. R. China.
| |
Collapse
|
9
|
Pan X, Guo A, Guan K, Chen C, Xu S, Tang Y, Li X, Huang Z. Lactobacillus rhamnosus GG attenuates depression-like behaviour and cognitive deficits in chronic ethanol exposure mice by down-regulating systemic inflammatory factors. Addict Biol 2024; 29:e13445. [PMID: 39585236 PMCID: PMC11587820 DOI: 10.1111/adb.13445] [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: 04/16/2024] [Revised: 07/31/2024] [Accepted: 09/20/2024] [Indexed: 11/26/2024]
Abstract
Ethanol can directly or indirectly lead to cognitive and mental disorders. The long-term intake of alcohol can directly affect the distribution of gut microbiota. Lactobacillus rhamnosus GG (LGG) is a natural bacterium isolated from healthy human intestines that has the function of preventing cytokine-induced cell apoptosis and protecting cell barriers. However, the regulatory effect of LGG on cognitive and mental disorders caused by chronic ethanol exposure (CEE) is still unclear. In this study, we established a CEE mouse model through free alcohol consumption and added LGG or antibiotics in the later stages of the model. Sequencing analysis of the 16S rRNA gene showed that CEE resulted in a decrease in the abundance and diversity of mouse gut microbial communities accompanied by alterations in the relative abundance of multiple enterobacterial genera. The use of LGG and antibiotics alleviated the depression-like behaviour and cognitive impairment of CEE-induced mice, reduced expression of inflammatory factors such as interleukin (IL)-6, IL-1β and tumour necrosis factor (TNF)-α in the ileum, serum and brain and increased the expression of synaptophysin (SYN), postsynaptic density protein-95 (PSD-95) and brain-derived neurotrophic factor (BDNF) in the hippocampus. Together, LGG can alleviate depression-like behaviour caused by CEE in mice while also improving cognitive and memory functions through reducing peripheral and nervous system inflammation factors and balancing gut microbiota.
Collapse
Affiliation(s)
- Xiaoyu Pan
- The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Anqi Guo
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental DisordersWenzhouZhejiangChina
| | - Kaiyu Guan
- Peking University Sixth Hospital, Peking University Institute of Mental HealthBeijingChina
| | - Congcong Chen
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental DisordersWenzhouZhejiangChina
| | - Shengnan Xu
- The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Yali Tang
- Institute of Brain ScienceWenzhou Medical UniversityWenzhouZhejiangChina
| | - Xi Li
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental DisordersWenzhouZhejiangChina
| | - Zhengwei Huang
- The Affiliated Kangning Hospital of Wenzhou Medical University, Zhejiang Provincial Clinical Research Center for Mental DisordersWenzhouZhejiangChina
| |
Collapse
|
10
|
Samulėnaitė S, García-Blanco A, Mayneris-Perxachs J, Domingo-Rodríguez L, Cabana-Domínguez J, Fernàndez-Castillo N, Gago-García E, Pineda-Cirera L, Burokas A, Espinosa-Carrasco J, Arboleya S, Latorre J, Stanton C, Hosomi K, Kunisawa J, Cormand B, Fernández-Real JM, Maldonado R, Martín-García E. Gut microbiota signatures of vulnerability to food addiction in mice and humans. Gut 2024; 73:1799-1815. [PMID: 38926079 PMCID: PMC11503113 DOI: 10.1136/gutjnl-2023-331445] [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: 11/01/2023] [Accepted: 04/01/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE Food addiction is a multifactorial disorder characterised by a loss of control over food intake that may promote obesity and alter gut microbiota composition. We have investigated the potential involvement of the gut microbiota in the mechanisms underlying food addiction. DESIGN We used the Yale Food Addiction Scale (YFAS) 2.0 criteria to classify extreme food addiction in mouse and human subpopulations to identify gut microbiota signatures associated with vulnerability to this disorder. RESULTS Both animal and human cohorts showed important similarities in the gut microbiota signatures linked to food addiction. The signatures suggested possible non-beneficial effects of bacteria belonging to the Proteobacteria phylum and potential protective effects of Actinobacteria against the development of food addiction in both cohorts of humans and mice. A decreased relative abundance of the species Blautia wexlerae was observed in addicted humans and of Blautia genus in addicted mice. Administration of the non-digestible carbohydrates, lactulose and rhamnose, known to favour Blautia growth, led to increased relative abundance of Blautia in mice faeces in parallel with dramatic improvements in food addiction. A similar improvement was revealed after oral administration of Blautia wexlerae as a beneficial microbe. CONCLUSION By understanding the crosstalk between this behavioural alteration and gut microbiota, these findings constitute a step forward to future treatments for food addiction and related eating disorders.
Collapse
Affiliation(s)
- Solveiga Samulėnaitė
- Laboratory of Neuropharmacology-Neurophar, Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Alejandra García-Blanco
- Laboratory of Neuropharmacology-Neurophar, Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Jordi Mayneris-Perxachs
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Diabetes, Endocrinology and Nutrition, Dr Josep Trueta University Hospital, Girona, Spain
| | - Laura Domingo-Rodríguez
- Laboratory of Neuropharmacology-Neurophar, Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Judit Cabana-Domínguez
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona, (IBUB), Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Barcelona, Spain
| | - Noèlia Fernàndez-Castillo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona, (IBUB), Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Barcelona, Spain
| | - Edurne Gago-García
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona, (IBUB), Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Barcelona, Spain
| | - Laura Pineda-Cirera
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona, (IBUB), Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Barcelona, Spain
| | - Aurelijus Burokas
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | | | - Silvia Arboleya
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Asturias, Spain
| | - Jessica Latorre
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Diabetes, Endocrinology and Nutrition, Dr Josep Trueta University Hospital, Girona, Spain
| | - Catherine Stanton
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Co, Cork, Ireland
| | - Koji Hosomi
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan. (NIBIOHN), Ibaraki, Osaka, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan. (NIBIOHN), Ibaraki, Osaka, Japan
| | - Bru Cormand
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, (CIBERER), Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona, (IBUB), Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Barcelona, Spain
| | - Jose Manuel Fernández-Real
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Diabetes, Endocrinology and Nutrition, Dr Josep Trueta University Hospital, Girona, Spain
- Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology-Neurophar, Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Elena Martín-García
- Laboratory of Neuropharmacology-Neurophar, Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
11
|
Li C, Zhu C, Tu G, Chen Z, Mo Z, Luo C. Impact of Altered Gut Microbiota on Ketamine-Induced Conditioned Place Preference in Mice. Neuropsychiatr Dis Treat 2024; 20:1725-1740. [PMID: 39318552 PMCID: PMC11421448 DOI: 10.2147/ndt.s476420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024] Open
Abstract
Objects Ketamine is a drug of abuse worldwide and current treatments for ketamine abuse are inadequate. It is an urgent need to develop novel anti-addictive strategy. Since gut microbiota plays a crucial role in drug abuse, the present study investigates the impact and mechanisms of the gut microbiota in addictive behaviors induced by ketamine addiction. Methods Conditioned place preference (CPP) was employed to assess addiction, followed by 16S rRNA gene sequencing to elucidate alterations in the gut microbiota. Furthermore, qRT-PCR, ELISA, and immunohistochemistry were conducted to evaluate the expression levels of crucial genes and proteins associated with the gut-brain axis. Additionally, we investigated whether ketamine addiction is regulated through the gut microbiota by orally administering antibiotics to establish pseudo-germ-free mice. Results We found that repeated ketamine administration (20 mg/kg) induced CPP and significantly altered gut microbiota diversity and composition, as revealed by 16S rRNA gene sequencing. Compared to the control group, ketamine exposure exhibited differences in the relative abundance of 5 microbial families, with 4 (Lachnospiraceae, Ruminococcaceae, Desulfovibrionaceae and Family-XIII) showing increases, while one (Prevotellaceae) displayed a decrease. At the genus level, five genera were upregulated, while one was downregulated. Furthermore, COG analysis revealed significant differences in protein functionality between the two groups. Additionally, axis series studies showed that ketamine dependence reduced levels of tight junction proteins, GABA and GABRA1, while increasing BDNF and 5-HT. Moreover, an oral antibiotic cocktail simulating pseudo germ-free conditions in mice did not enhance the addictive behavior induced by ketamine. Conclusion Our study supports the hypothesis that ketamine-induced CPP is mediated through the gut microbiota. The present study provides new insights into improvement of efficient strategy for addiction treatment.
Collapse
Affiliation(s)
- Chan Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, People's Republic of China
- School of Life Sciences, Guangzhou University, Guangzhou, People's Republic of China
| | - Chen Zhu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Genghong Tu
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong, People's Republic of China
| | - Zhijie Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, People's Republic of China
| | - Zhixian Mo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, People's Republic of China
| | - Chaohua Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
12
|
Camarini R, Marianno P, Hanampa-Maquera M, Oliveira SDS, Câmara NOS. Prenatal Stress and Ethanol Exposure: Microbiota-Induced Immune Dysregulation and Psychiatric Risks. Int J Mol Sci 2024; 25:9776. [PMID: 39337263 PMCID: PMC11431796 DOI: 10.3390/ijms25189776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/22/2024] [Accepted: 08/25/2024] [Indexed: 09/30/2024] Open
Abstract
Changes in maternal gut microbiota due to stress and/or ethanol exposure can have lasting effects on offspring's health, particularly regarding immunity, inflammation response, and susceptibility to psychiatric disorders. The literature search for this review was conducted using PubMed and Scopus, employing keywords and phrases related to maternal stress, ethanol exposure, gut microbiota, microbiome, gut-brain axis, diet, dysbiosis, progesterone, placenta, prenatal development, immunity, inflammation, and depression to identify relevant studies in both preclinical and human research. Only a limited number of reviews were included to support the arguments. The search encompassed studies from the 1990s to the present. This review begins by exploring the role of microbiota in modulating host health and disease. It then examines how disturbances in maternal microbiota can affect the offspring's immune system. The analysis continues by investigating the interplay between stress and dysbiosis, focusing on how prenatal maternal stress influences both maternal and offspring microbiota and its implications for susceptibility to depression. The review also considers the impact of ethanol consumption on gut dysbiosis, with an emphasis on the effects of prenatal ethanol exposure on both maternal and offspring microbiota. Finally, it is suggested that maternal gut microbiota dysbiosis may be significantly exacerbated by the combined effects of stress and ethanol exposure, leading to immune system dysfunction and chronic inflammation, which could increase the risk of depression in the offspring. These interactions underscore the potential for novel mental health interventions that address the gut-brain axis, especially in relation to maternal and offspring health.
Collapse
Affiliation(s)
- Rosana Camarini
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Priscila Marianno
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Maylin Hanampa-Maquera
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Samuel Dos Santos Oliveira
- Department of Immunology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Niels Olsen Saraiva Câmara
- Department of Immunology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| |
Collapse
|
13
|
Fu H, Ge Y, Liu X, Deng S, Li J, Tan P, Yang Y, Wu Z. Exposure to the environmental pollutant chlorpyrifos induces hepatic toxicity through activation of the JAK/STAT and MAPK pathways. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 928:171711. [PMID: 38494025 DOI: 10.1016/j.scitotenv.2024.171711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 02/16/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024]
Abstract
Chlorpyrifos (CHP) is an inexpensive highly effective organophosphate insecticide used worldwide. The unguided and excessive use of CHP by farmers has led to its significant accumulation in crops as well as contamination of water sources, causing health problems for humans and animals. Therefore, this study evaluated the toxicological effects of exposure to the environmental pollutant CHP at low, medium, and high (2.5, 5, and 10 mg·kg-1 BW) levels on rat liver by examining antioxidant levels, inflammation, and apoptosis based on the no observed adverse effect levels (NOAEL) (1 mg·kg-1 BW) and the CHP dose that does not cause any visual symptoms (5 mg·kg-1 BW). Furthermore, the involvement of the JAK/STAT and MAPK pathways in CHP-induced toxic effects was identified. The relationship between the expression levels of key proteins (p-JAK/JAK, p-STAT/STAT, p-JNK/JNK, p-P38/P38, and p-ERK/ERK) in the pathways and changes in the expression of markers associated with inflammation [inflammatory factors (IL-1β, IL-6, IL-10, TNF-α), chemokines (GCLC and GCLM), and inflammatory signaling pathways (NF-кB, TLR2, TLR4, NLRP3, ASC, MyD88, IFN-γ, and iNOS)] and apoptosis [Bad, Bax, Bcl-2, Caspase3, Caspase9, and the cleavage substrate of Caspase PARP1] were also determined. The results suggest that CHP exposure disrupts liver function and activates the JAK/STAT and MAPK pathways via oxidative stress, exacerbating inflammation and apoptosis. Meanwhile, the JAK/STAT and MAPK pathways are involved in CHP-induced hepatotoxicity. These findings provide a novel direction for effective prevention and amelioration of health problems caused by CHP abuse in agriculture and households.
Collapse
Affiliation(s)
- Huiyang Fu
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China
| | - Yao Ge
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Xiyuan Liu
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China
| | - Siwei Deng
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China
| | - Jun Li
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China
| | - Peng Tan
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China; Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
14
|
Yu Z, Guo M, Yu B, Wang Y, Yan Z, Gao R. Anorexia nervosa and bulimia nervosa: a Mendelian randomization study of gut microbiota. Front Microbiol 2024; 15:1396932. [PMID: 38784806 PMCID: PMC11111991 DOI: 10.3389/fmicb.2024.1396932] [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: 03/06/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Background Anorexia nervosa (AN) and bulimia nervosa (BN) poses a significant challenge to global public health. Despite extensive research, conclusive evidence regarding the association between gut microbes and the risk of AN and BN remains elusive. Mendelian randomization (MR) methods offer a promising avenue for elucidating potential causal relationships. Materials and methods Genome-wide association studies (GWAS) datasets of AN and BN were retrieved from the OpenGWAS database for analysis. Independent single nucleotide polymorphisms closely associated with 196 gut bacterial taxa from the MiBioGen consortium were identified as instrumental variables. MR analysis was conducted utilizing R software, with outlier exclusion performed using the MR-PRESSO method. Causal effect estimation was undertaken employing four methods, including Inverse variance weighted. Sensitivity analysis, heterogeneity analysis, horizontal multivariate analysis, and assessment of causal directionality were carried out to assess the robustness of the findings. Results A total of 196 bacterial taxa spanning six taxonomic levels were subjected to analysis. Nine taxa demonstrating potential causal relationships with AN were identified. Among these, five taxa, including Peptostreptococcaceae, were implicated as exerting a causal effect on AN risk, while four taxa, including Gammaproteobacteria, were associated with a reduced risk of AN. Similarly, nine taxa exhibiting potential causal relationships with BN were identified. Of these, six taxa, including Clostridiales, were identified as risk factors for increased BN risk, while three taxa, including Oxalobacteraceae, were deemed protective factors. Lachnospiraceae emerged as a common influence on both AN and BN, albeit with opposing effects. No evidence of heterogeneity or horizontal pleiotropy was detected for significant estimates. Conclusion Through MR analysis, we revealed the potential causal role of 18 intestinal bacterial taxa in AN and BN, including Lachnospiraceae. It provides new insights into the mechanistic basis and intervention targets of gut microbiota-mediated AN and BN.
Collapse
Affiliation(s)
- Zongliang Yu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Manping Guo
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Postdoctoral Research Station, China Academy of Chinese Medical Sciences, Beijing, China
- Postdoctoral Works Station, Yabao Pharmaceutical Group Co., Ltd., Yuncheng, China
| | - Binyang Yu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yiming Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zian Yan
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rui Gao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
15
|
Zhou Y, Zhang L, Li Q, Wang P, Wang H, Shi H, Lu W, Zhang Y. Prenatal PFAS exposure, gut microbiota dysbiosis, and neurobehavioral development in childhood. JOURNAL OF HAZARDOUS MATERIALS 2024; 469:133920. [PMID: 38457972 DOI: 10.1016/j.jhazmat.2024.133920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 01/17/2024] [Accepted: 02/27/2024] [Indexed: 03/10/2024]
Abstract
Studies on the role of the gut microbiota in the associations between per- and polyfluoroalkyl substance (PFAS) exposure and adverse neurodevelopment are limited. Umbilical cord serum and faeces samples were collected from children, and the Strengths and Difficulties Questionnaire (SDQ) was conducted. Generalized linear models, linear mixed-effects models, multivariate analysis by linear models and microbiome regression-based kernel association tests were used to evaluate the associations among PFAS exposure, the gut microbiota, and neurobehavioural development. Perfluorohexane sulfonic acid (PFHxS) exposure was associated with increased scores for conduct problems and externalizing problems, as well as altered gut microbiota alpha and beta diversity. PFHxS concentrations were associated with higher relative abundances of Enterococcus spp. but lower relative abundances of several short-chain fatty acid-producing genera (e.g., Ruminococcus gauvreauii group spp.). PFHxS exposure was also associated with increased oxidative phosphorylation. Alpha and beta diversity were found significantly associated with conduct problems and externalizing problems. Ruminococcus gauvreauii group spp. abundance was positively correlated with prosocial behavior scores. Increased alpha diversity played a mediating role in the associations of PFHxS exposure with conduct problems. Our results suggest that the gut microbiota might play an important role in PFAS neurotoxicity, which may have implications for PFAS control.
Collapse
Affiliation(s)
- Yuhan Zhou
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China, Fudan University, Shanghai 200032, China; School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Liyi Zhang
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China, Fudan University, Shanghai 200032, China
| | - Qiang Li
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China, Fudan University, Shanghai 200032, China; Putuo District Center for Disease Control & Prevention, Shanghai 200333, China
| | - Pengpeng Wang
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China, Fudan University, Shanghai 200032, China
| | - Hang Wang
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China, Fudan University, Shanghai 200032, China
| | - Huijing Shi
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China, Fudan University, Shanghai 200032, China
| | - Wenwei Lu
- School of Science and Technology, Jiangnan University, Jiangsu 214122, China
| | - Yunhui Zhang
- Key Lab of Health Technology Assessment, National Health Commission of the People's Republic of China, Fudan University, Shanghai 200032, China.
| |
Collapse
|
16
|
Koutromanos I, Legaki E, Gazouli M, Vasilopoulos E, Kouzoupis A, Tzavellas E. Gut microbiome in alcohol use disorder: Implications for health outcomes and therapeutic strategies-a literature review. World J Methodol 2024; 14:88519. [PMID: 38577203 PMCID: PMC10989405 DOI: 10.5662/wjm.v14.i1.88519] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/22/2023] [Accepted: 01/24/2024] [Indexed: 03/07/2024] Open
Abstract
Alcohol use disorder (AUD) represents a major public health issue which affects millions of people globally and consist a chronic relapsing condition associated with substantial morbidity and mortality. The gut microbiome plays a crucial role in maintaining overall health and has emerged as a significant contributor to the pathophysiology of various psychiatric disorders. Recent evidence suggests that the gut microbiome is intimately linked to the development and progression of AUD, with alcohol consumption directly impacting its composition and function. This review article aims to explore the intricate relationship between the gut microbiome and AUD, focusing on the implications for mental health outcomes and potential therapeutic strategies. We discuss the bidirectional communication between the gut microbiome and the brain, highlighting the role of microbiota-derived metabolites in neuroinflammation, neurotransmission, and mood regulation. Furthermore, we examine the influence of AUD-related factors, such as alcohol-induced gut dysbiosis and increased intestinal permeability, on mental health outcomes. Finally, we explore emerging therapeutic avenues targeting the gut microbiome in the management of AUD, including prebiotics, probiotics, and fecal microbiota transplantation. Understanding the complex interplay between the gut microbiome and AUD holds promise for developing novel interventions that could improve mental health outcomes in individuals with AUD.
Collapse
Affiliation(s)
- Ilias Koutromanos
- First Department of Psychiatry, "Aiginition" Hospital, School of Medicine, National and Kapodistrian University of Athens, School of Medicine, National and Kapodistrian University of Athens, Athens 11528, Greece
| | - Evangelia Legaki
- Department of Basic Biological Science, School of Medicine, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Maria Gazouli
- Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Efthimios Vasilopoulos
- First Department of Psychiatry, "Aiginition" Hospital, School of Medicine, National and Kapodistrian University of Athens, School of Medicine, National and Kapodistrian University of Athens, Athens 11528, Greece
| | - Anastasios Kouzoupis
- First Department of Psychiatry, "Aiginition" Hospital, School of Medicine, National and Kapodistrian University of Athens, School of Medicine, National and Kapodistrian University of Athens, Athens 11528, Greece
| | - Elias Tzavellas
- First Department of Psychiatry, "Aiginition" Hospital, School of Medicine, National and Kapodistrian University of Athens, School of Medicine, National and Kapodistrian University of Athens, Athens 11528, Greece
| |
Collapse
|
17
|
Chen S, Li M, Tong C, Wang Y, He J, Shao Q, Liu Y, Wu Y, Song Y. Regulation of miRNA expression in the prefrontal cortex by fecal microbiota transplantation in anxiety-like mice. Front Psychiatry 2024; 15:1323801. [PMID: 38410679 PMCID: PMC10894985 DOI: 10.3389/fpsyt.2024.1323801] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/29/2024] [Indexed: 02/28/2024] Open
Abstract
Background The gut-brain axis and gut microbiota have emerged as key players in emotional disorders. Recent studies suggest that alterations in gut microbiota may impact psychiatric symptoms through brain miRNA along the gut-brain axis. However, direct evidence linking gut microbiota to the pathophysiology of generalized anxiety disorder (GAD) via brain miRNA is limited. In this study, we explored the effects of fecal microbiota transplantation (FMT) from GAD donors on gut microbiota and prefrontal cortex miRNA in recipient mice, aiming to understand the relationship between these two factors. Methods Anxiety scores and gut microbiota composition were assessed in GAD patients, and their fecal samples were utilized for FMT in C57BL/6J mice. Anxiety-like behavior in mice was evaluated using open field and elevated plus maze tests. High-throughput sequencing of gut microbiota 16S rRNA and prefrontal cortex miRNA was performed. Results The fecal microbiota of GAD patients exhibited a distinct microbial structure compared to the healthy group, characterized by a significant decrease in Verrucomicrobia and Akkermansia, and a significant increase in Actinobacteria and Bacteroides. Subsequent FMT from GAD patients to mice induced anxiety-like behavior in recipients. Detailed analysis of gut microbiota composition revealed lower abundances of Verrucomicrobia, Akkermansia, Bifidobacterium, and Butyricimonas, and higher abundances of Deferribacteres, Allobaculum, Bacteroides, and Clostridium in mice that received FMT from GAD patients. MiRNA analysis identified five key miRNAs affecting GAD pathogenesis, including mmu-miR-10a-5p, mmu-miR-1224-5p, mmu-miR-218-5p, mmu-miR-10b-5p, and mmu-miR-488-3p. Notably, mmu-miR-488-3p showed a strong negative correlation with Verrucomicrobia and Akkermansia. Conclusion This study demonstrates that anxiety-like behavior induced by human FMT can be transmitted through gut microbiota and is associated with miRNA expression in the prefrontal cortex. It is inferred that the reduction of Akkermansia caused by FMT from GAD patients leads to the upregulation of mmu-miR-488-3p expression, resulting in the downregulation of its downstream target gene Creb1 and interference with its related signaling pathway. These findings highlight the gut microbiota's crucial role in the GAD pathophysiology.
Collapse
Affiliation(s)
- Simin Chen
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Mengjia Li
- College of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Changqing Tong
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yanan Wang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jiahui He
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qi Shao
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Liu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Wu
- Liuzhou Key laboratory of infection disease and immunology, Research Center of Medical Sciences, Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
| | - Yuehan Song
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
18
|
Wigren MA, Johnson TA, Griffitt RJ, Hay AG, Knott JA, Sepúlveda MS. Limited impact of weathered residues from the Deepwater Horizon oil spill on the gut-microbiome and foraging behavior of sheepshead minnows ( Cyprinodon variegatus). JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2024; 87:1-21. [PMID: 37830742 DOI: 10.1080/15287394.2023.2265413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
The Deepwater Horizon disaster of April 2010 was the largest oil spill in U.S. history and exerted catastrophic effects on several ecologically important fish species in the Gulf of Mexico (GoM). Within fish, the microbiome plays a key symbiotic role in maintaining host health and aids in acquiring nutrients, supporting immune function, and modulating behavior. The aim of this study was to examine if exposure to weathered oil might produce significant shifts in fish gut-associated microbial communities as determined from taxa and genes known for hydrocarbon degradation, and whether foraging behavior was affected. The gut microbiome (16S rRNA and shotgun metagenomics) of sheepshead minnow (Cyprinodon variegatus) was characterized after fish were exposed to oil in High Energy Water Accommodated Fractions (HEWAF; tPAH = 81.1 ± 12.4 µg/L) for 7 days. A foraging behavioral assay was used to determine feeding efficiency before and after oil exposure. The fish gut microbiome was not significantly altered in alpha or beta diversity. None of the most abundant taxa produced any significant shifts as a result of oil exposure, with only rare taxa showing significant shifts in abundance between treatments. However, several bioindicator taxa known for hydrocarbon degradation were detected in the oil treatment, primarily Sphingomonas and Acinetobacter. Notably, the genus Stenotrophomonas was detected in high abundance in 16S data, which previously was not described as a core member of fish gut microbiomes. Data also demonstrated that behavior was not significantly affected by oil exposure. Potential low bioavailability of the oil may have been a factor in our observation of minor shifts in taxa and no behavioral effects. This study lays a foundation for understanding the microbiome of captive sheepshead minnows and indicates the need for further research to elucidate the responses of the fish gut-microbiome under oil spill conditions.
Collapse
Affiliation(s)
- Maggie A Wigren
- Department of Forestry and Natural Resources, Purdue University, West Lafayette, IN, USA
| | - Timothy A Johnson
- Department of Animal Sciences, Purdue University, West Lafayette, IN, USA
| | - Robert J Griffitt
- Division of Coastal Sciences, School of Ocean Science and Engineering, University of Southern Mississippi, Ocean Springs, MS, USA
| | - Anthony G Hay
- Department of Microbiology, Cornell University, Ithaca, NY, USA
| | - Jonathan A Knott
- Department of Forestry and Natural Resources, Purdue University, West Lafayette, IN, USA
| | - Maria S Sepúlveda
- Department of Forestry and Natural Resources, Purdue University, West Lafayette, IN, USA
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
19
|
Wang C, Yan J, Du K, Liu S, Wang J, Wang Q, Zhao H, Li M, Yan D, Zhang R, Yang F. Intestinal microbiome dysbiosis in alcohol-dependent patients and its effect on rat behaviors. mBio 2023; 14:e0239223. [PMID: 37962470 PMCID: PMC10746284 DOI: 10.1128/mbio.02392-23] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/03/2023] [Indexed: 11/15/2023] Open
Abstract
IMPORTANCE Intestinal microbiome dysbiosis is associated with psychiatric disease through the "microbiota-gut-brain" axis. Here, we revealed that there was obvious intestinal microbiome (including bacterial and fungal) dysbiosis in alcohol-dependent patients. Alcohol consumption seriously disturbs the gut equilibrium between bacteria and fungi, reduces the interactions among bacterial-fungal trans-kingdom, and increases intestinal permeability. Gut microbiota should be considered as a whole to study the development of alcohol dependence. The gut microbiome of alcohol-dependent patients increased the anxiety- and depression-like behavior in rats. The gut microbiota dysbiosis may promote the development of alcohol dependence by regulating the endogenous cholecystokinin (CCK) and related receptors. Hence, regulating the balance of gut microbiota and the endogenous CCK may be a potential strategy for reducing the risk of relapse in alcohol addiction patients.
Collapse
Affiliation(s)
- Chuansheng Wang
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - Junli Yan
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - Keda Du
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - Shuai Liu
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - Jiali Wang
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - Qi Wang
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - Huajie Zhao
- Department of Pathogeny, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Min Li
- Department of Pathogeny, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Dong Yan
- Department of Pathogeny, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Ruiling Zhang
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - Fan Yang
- Department of Pathogeny, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
20
|
Yin Y, Fan CF. The Effects of Mukbang Watching on Enteral Feeding Intolerance Among Critically Ill Patients: Study Protocol for a Randomized Controlled Trail. Patient Prefer Adherence 2023; 17:2891-2897. [PMID: 37965439 PMCID: PMC10642568 DOI: 10.2147/ppa.s438190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/02/2023] [Indexed: 11/16/2023] Open
Abstract
Purpose With an estimated prevalence of 38%, enteral feeding intolerance (EFI) is common in critically ill patients receiving enteral nutrition (EN), and is associated with higher mortality and longer duration of mechanical ventilation. Various methods have been reported to decrease the incidence of EFI during EN, such as post-pyloric feeding, continuous EN delivery, abdominal massage, and probiotic supplementation. However, several studies reported conflicting results. Inappropriate interventions may cause gastrointestinal (GI) injury. This study aims to design a protocol based on Mukbang videos, which are widely watched online, to detect their effects on the incidence of EFI, nutritional status, incidence of infectious complications, and activities of daily living. Patients and Methods We will conduct a three-arm, parallel-design, randomized controlled trial that will be implemented in 273 patients from intensive care units. Participants will be randomized into one of the three intervention arms (1:1:1), which will be performed by a research assistant. Participants were allocated to three groups: (a) watching mukbang video, (b) watching a cooking show, and (c) watching a non-food content video. Prior to EN initiation, each participant will watch a ten-minute mukbang video, cookery show, or non-food content video. Conclusion Mukbang videos show food, expressions of mukbangers and eating sounds. If it effectively reduces the incidence of EFI, leads to greater nutritional status, lower incidence of infectious complications, and a higher level of independence among patients compared with watching cooking videos or non-food content videos, it has broad dissemination potential as a non-invasive, easily assessing, and using method.
Collapse
Affiliation(s)
- Yao Yin
- Department of Neurosurgery, West China Hospital, West China School of Nursing, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Chao-Feng Fan
- Department of Neurosurgery, West China Hospital, West China School of Nursing, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
21
|
Wen X, Yang H, Li Z, Chu W. Alcohol degradation, learning, and memory-enhancing effect of Acetobacter pasteurianus BP2201 in Caenorhabditis elegans model. J Appl Microbiol 2023; 134:lxad253. [PMID: 37934610 DOI: 10.1093/jambio/lxad253] [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: 07/23/2023] [Revised: 08/18/2023] [Accepted: 10/31/2023] [Indexed: 11/09/2023]
Abstract
AIMS This study aimed to investigate the probiotic effects of Acetobacter pasteurianus BP2201, isolated from brewing mass, for the treatment of alcohol-induced learning and memory ability impairments in a Caenorhabditis elegans model. METHODS AND RESULTS Acetobacter pasteurianus BP2201 was examined for probiotic properties, including acid and bile salt resistance, ethanol degradation, antioxidant efficacy, hemolytic activity, and susceptibility to antibiotics. The strain displayed robust acid and bile salt tolerance, efficient ethanol degradation, potent antioxidant activity, and susceptibility to specific antibiotics. Additionally, in the C. elegans model, administering A. pasteurianus BP2201 significantly improved alcohol-induced learning and memory impairments. CONCLUSIONS Acetobacter pasteurianus BP2201 proves to be a promising candidate strain for the treatment of learning and memory impairments induced by alcohol intake.
Collapse
Affiliation(s)
- Xin Wen
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Huazhong Yang
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Zhongqi Li
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Weihua Chu
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
22
|
Wang M, Jiang Y, Wang S, Fu L, Liang Z, Zhang Y, Huang X, Li X, Feng M, Long D. Yak milk protects against alcohol-induced liver injury in rats. Food Funct 2023; 14:9857-9871. [PMID: 37853817 DOI: 10.1039/d3fo03675h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
The protective effects of yak milk (YM) against chronic alcoholic liver injury in rats were investigated in this study. Histologic and biochemical analyses demonstrated that YM consumption ameliorates alcohol-induced liver injury by increasing the liver antioxidant enzyme activity and reducing inflammation. Furthermore, microbiome and metabolomic analyses exploring YM's impact on gut microbiota and metabolism found that YM administration regulates gut microbiota composition. Specifically, there was a decrease in the relative abundance of Helicobacter, Streptococcus, Peptococcus and Tyzzerella, along with an increase in Turisibacter and Intestinimonas. Moreover, Pearson analysis indicated positive correlations between Peptococcus and Tyzzerella with ALT and AST levels, while showing a negative correlation with ADH levels. Furthermore, differential metabolite analysis of fecal samples from the YM group identified significant increases in the taurine (2-Aminoethanesulfonic acid), hypotaurine (2-Aminoethanesulfonic Acid) and isethionic acid levels. Finally, KEGG topology analysis highlighted taurine and hypotaurine metabolism as the primary pathways influenced by YM intervention. Therefore, these findings collectively suggest that YM may protect alcohol-exposed rats against liver injury by modulating oxidative stress, inflammatory response, gut microbiota disorder, and metabolic regulation.
Collapse
Affiliation(s)
- Man Wang
- School of Public Health, Lanzhou University, Lanzhou, China.
| | - Yanshi Jiang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Siying Wang
- School of Public Health, Lanzhou University, Lanzhou, China.
| | - Lin Fu
- School of Public Health, Lanzhou University, Lanzhou, China.
| | - Zujin Liang
- School of Public Health, Lanzhou University, Lanzhou, China.
| | - Ying Zhang
- School of Public Health, Lanzhou University, Lanzhou, China.
| | - Xiaodan Huang
- School of Public Health, Lanzhou University, Lanzhou, China.
| | - Xin Li
- School of Public Health, Lanzhou University, Lanzhou, China.
| | - Meiying Feng
- School of Public Health, Lanzhou University, Lanzhou, China.
| | - Danfeng Long
- School of Public Health, Lanzhou University, Lanzhou, China.
| |
Collapse
|
23
|
Watanabe C, Oyanagi E, Aoki T, Hamada H, Kawashima M, Yamagata T, Kremenik MJ, Yano H. Antidepressant properties of voluntary exercise mediated by gut microbiota. Biosci Biotechnol Biochem 2023; 87:1407-1419. [PMID: 37667506 DOI: 10.1093/bbb/zbad115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/05/2023] [Indexed: 09/06/2023]
Abstract
Although regular exercise has been reported to prevent depression, it has not been clarified whether the gut microbiota is involved in the factors that prevent depression through exercise. We investigated the effects of voluntary exercise on the gut microbiota and the prevention of depression-like behaviors using mice. C57BL/6 J male mice were subjected to 10 weeks of sedentary control or wheel running, then they were subjected to social defeat stress (SDS). Exercise attenuated that sucrose drinking was decreased by SDS treatment. Exercise increased the expression of Bdnf and decreased expression of Zo-1 and Claudin5 in the brain. Fecal Turicibacter, Allobaculum, and Clostridium sensu stricto, and propionate in the cecum were decreased by the exercise. Voluntary exercise-induced antidepressant properties might be partially caused by suppression of serotonin uptake into gut microbiota and increase the permeability of the blood-brain barrier via reduced propionate production.
Collapse
Affiliation(s)
- Chihiro Watanabe
- Graduate School of Health Science and Technology, Kawasaki University of Medical Welfare, Kurashiki, Okayama, Japan
| | - Eri Oyanagi
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki, Okayama, Japan
| | - Takafumi Aoki
- Department of Clinical Nutrition, Kawasaki University of Medical Welfare, Kurashiki, Okayama, Japan
| | - Hiroki Hamada
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki, Okayama, Japan
| | - Masato Kawashima
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki, Okayama, Japan
| | - Takashi Yamagata
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki, Okayama, Japan
| | - Michel J Kremenik
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki, Okayama, Japan
| | - Hiromi Yano
- Graduate School of Health Science and Technology, Kawasaki University of Medical Welfare, Kurashiki, Okayama, Japan
- Department of Health and Sports Science, Kawasaki University of Medical Welfare, Kurashiki, Okayama, Japan
| |
Collapse
|
24
|
Khan MAS, Chang SL. Alcohol and the Brain-Gut Axis: The Involvement of Microglia and Enteric Glia in the Process of Neuro-Enteric Inflammation. Cells 2023; 12:2475. [PMID: 37887319 PMCID: PMC10605902 DOI: 10.3390/cells12202475] [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: 07/16/2023] [Revised: 09/25/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023] Open
Abstract
Binge or chronic alcohol consumption causes neuroinflammation and leads to alcohol use disorder (AUD). AUD not only affects the central nervous system (CNS) but also leads to pathologies in the peripheral and enteric nervous systems (ENS). Thus, understanding the mechanism of the immune signaling to target the effector molecules in the signaling pathway is necessary to alleviate AUD. Growing evidence shows that excessive alcohol consumption can activate neuroimmune cells, including microglia, and change the status of neurotransmitters, affecting the neuroimmune system. Microglia, like peripheral macrophages, are an integral part of the immune defense and represent the reticuloendothelial system in the CNS. Microglia constantly survey the CNS to scavenge the neuronal debris. These cells also protect parenchymal cells in the brain and spinal cord by repairing nerve circuits to keep the nervous system healthy against infectious and stress-derived agents. In an activated state, they become highly dynamic and mobile and can modulate the levels of neurotransmitters in the CNS. In several ways, microglia, enteric glial cells, and macrophages are similar in terms of causing inflammation. Microglia also express most of the receptors that are constitutively present in macrophages. Several receptors on microglia respond to the inflammatory signals that arise from danger-associated molecular patterns (DAMPs), pathogen-associated molecular patterns (PAMPs), endotoxins (e.g., lipopolysaccharides), and stress-causing molecules (e.g., alcohol). Therefore, this review article presents the latest findings, describing the roles of microglia and enteric glial cells in the brain and gut, respectively, and their association with neurotransmitters, neurotrophic factors, and receptors under the influence of binge and chronic alcohol use, and AUD.
Collapse
Affiliation(s)
- Mohammed A. S. Khan
- Department of Neurosurgery, Brigham Hospital for Children, Harvard Medical School, Boston, MA 02115, USA;
| | - Sulie L. Chang
- Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, NJ 07079, USA
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07079, USA
| |
Collapse
|
25
|
Wen X, Wang Z, Liu Q, Lessing DJ, Chu W. Acetobacter pasteurianus BP2201 alleviates alcohol-induced hepatic and neuro-toxicity and modulate gut microbiota in mice. Microb Biotechnol 2023; 16:1834-1857. [PMID: 37354051 PMCID: PMC10443346 DOI: 10.1111/1751-7915.14303] [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: 04/19/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/26/2023] Open
Abstract
The excessive consumption of alcohol results in a dysbiosis of the gut microbiota, which subsequently impairs the gut microbiota-brain/liver axes and induces cognitive dysfunction and hepatic injury. This study aimed to investigate the potential effect of Acetobacter pasteurianus BP2201 in reducing the negative effects of alcohol consumption on cognitive function and liver health by modulating the gut microbiota-brain/liver axes. Treatment with A. pasteurianus BP2201 improved alcohol-induced hippocampal damage, suppressed neuroinflammation, promoted neuroprotein expression in the hippocampus and enhanced cognitive function. At the same time, A. pasteurianus BP2201 can also reduce serum lipid levels, relieve oxidative stress, inhibit TLR4/MyD88/NF-κB pathway, reduce the secretion of TNF-α and IL-1β, so as to improve alcoholic liver injury. Concomitantly, the treatment with A. pasteurianus BP2201 leads to a shift in the intestinal microbiota structure towards that of healthy individuals, inhibiting the proliferation of harmful bacteria and promoting the recovery of beneficial bacteria. In addition, it also improves brain cognitive dysfunction and liver health by affecting the gut microbiota-brain/liver axes by promoting the synthesis of relevant amino acids and the metabolism of nucleotide base components. These findings demonstrate the potential of regulating the gut microbiome and gut microbiota-brain/liver axes to mitigate alcohol-induced disease.
Collapse
Affiliation(s)
- Xin Wen
- Department of Pharmaceutical Microbiology, School of Life Science and TechnologyChina Pharmaceutical UniversityNanjingChina
| | - Zheng Wang
- Department of Pharmaceutical Microbiology, School of Life Science and TechnologyChina Pharmaceutical UniversityNanjingChina
| | - Qi Liu
- Department of Pharmaceutical Microbiology, School of Life Science and TechnologyChina Pharmaceutical UniversityNanjingChina
| | - Duncan James Lessing
- Department of Pharmaceutical Microbiology, School of Life Science and TechnologyChina Pharmaceutical UniversityNanjingChina
| | - Weihua Chu
- Department of Pharmaceutical Microbiology, School of Life Science and TechnologyChina Pharmaceutical UniversityNanjingChina
- State Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjingChina
| |
Collapse
|
26
|
Zhu L, Wang Y, Pan CQ, Xing H. Gut microbiota in alcohol-related liver disease: pathophysiology and gut-brain cross talk. Front Pharmacol 2023; 14:1258062. [PMID: 37601074 PMCID: PMC10436520 DOI: 10.3389/fphar.2023.1258062] [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: 07/13/2023] [Accepted: 07/27/2023] [Indexed: 08/22/2023] Open
Abstract
Alcohol-related liver disease (ALD) from excessive alcohol intake has a unique gut microbiota profile. The disease progression-free survival in ALD patients has been associated with the degree of gut dysbiosis. The vicious cycles between gut dysbiosis and the disease progression in ALD including: an increase of acetaldehyde production and bile acid secretion, impaired gut barrier, enrichment of circulating microbiota, toxicities of microbiota metabolites, a cascade of pro-inflammatory chemokines or cytokines, and augmentation in the generation of reactive oxygen species. The aforementioned pathophysiology process plays an important role in different disease stages with a spectrum of alcohol hepatitis, ALD cirrhosis, neurological dysfunction, and hepatocellular carcinoma. This review aims to illustrate the pathophysiology of gut microbiota and clarify the gut-brain crosstalk in ALD, which may provide the opportunity of identifying target points for future therapeutic intervention in ALD.
Collapse
Affiliation(s)
- Lin Zhu
- Center of Liver Diseases Division 3, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yixuan Wang
- Division of Gastroenterology and Hepatology, BaoJi Central Hospital, Shaanxi, China
| | - Calvin Q. Pan
- Center of Liver Diseases Division 3, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Division of Gastroenterology and Hepatology, NYU Langone Health, New York University School of Medicine, New York, NY, United States
| | - Huichun Xing
- Center of Liver Diseases Division 3, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Center of Liver Diseases, Peking University Ditan Teaching Hospital, Beijing, China
| |
Collapse
|
27
|
Kong D, Sun JX, Yang JQ, Li YS, Bi K, Zhang ZY, Wang KH, Luo HY, Zhu M, Xu Y. Ketogenic diet: a potential adjunctive treatment for substance use disorders. Front Nutr 2023; 10:1191903. [PMID: 37575322 PMCID: PMC10414993 DOI: 10.3389/fnut.2023.1191903] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
Substance use disorders (SUD) can lead to serious health problems, and there is a great interest in developing new treatment methods to alleviate the impact of substance abuse. In recent years, the ketogenic diet (KD) has shown therapeutic benefits as a dietary therapy in a variety of neurological disorders. Recent studies suggest that KD can compensate for the glucose metabolism disorders caused by alcohol use disorder by increasing ketone metabolism, thereby reducing withdrawal symptoms and indicating the therapeutic potential of KD in SUD. Additionally, SUD often accompanies increased sugar intake, involving neural circuits and altered neuroplasticity similar to substance addiction, which may induce cross-sensitization and increased use of other abused substances. Reducing carbohydrate intake through KD may have a positive effect on this. Finally, SUD is often associated with mitochondrial damage, oxidative stress, inflammation, glia dysfunction, and gut microbial disorders, while KD may potentially reverse these abnormalities and serve a therapeutic role. Although there is much indirect evidence that KD has a positive effect on SUD, the small number of relevant studies and the fact that KD leads to side effects such as metabolic abnormalities, increased risk of malnutrition and gastrointestinal symptoms have led to the limitation of KD in the treatment of SUD. Here, we described the organismal disorders caused by SUD and the possible positive effects of KD, aiming to provide potential therapeutic directions for SUD.
Collapse
Affiliation(s)
- Deshenyue Kong
- General Hospital of Eastern Theater Command, Nanjing, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jia-xue Sun
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ji-qun Yang
- Third People’s Hospital of Kunming City/Drug Rehabilitation Hospital of Kunming City, Kunming, China
| | - Yuan-sen Li
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ke Bi
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zun-yue Zhang
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
| | - Kun-hua Wang
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
| | - Hua-you Luo
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Mei Zhu
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yu Xu
- General Hospital of Eastern Theater Command, Nanjing, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
28
|
Kim CS, Shin DM. Gut microbiota and cognitive development in infant mice: Quantity and source of potable water. PLoS One 2023; 18:e0286951. [PMID: 37315057 DOI: 10.1371/journal.pone.0286951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/29/2023] [Indexed: 06/16/2023] Open
Abstract
Not only the water quantity consumed but also the source of drinking water has been considered for their health benefits, but there is limited evidence. We aimed to determine whether the amount and type of drinking water affect physiological and biological functions, including brain function, by confirming how it affects gut microbiota which has an important regulatory role in host physiology. Three-week-old infant mice were subjected to 1) a water restriction experiment (control group, ad libitum consumption of distilled water; dehydration group, time-limited access to distilled water [15 min/day]) and 2) different water source experiment (distilled water, purified water, spring water, and tap water groups). The gut microbiota and cognitive development were analyzed using the 16S ribosomal ribonucleic acid sequencing method and the Barnes maze, respectively. The relative abundance of Firmicutes and Bacteroidetes and the Firmicutes-to-Bacteroidetes ratio (F/B ratio) changed depending on age (juveniles vs. infants). Insufficient water intake reversed these developmental changes, showing that the relative abundances of Bacteroidetes and Firmicutes and the F/B ratio in dehydrated juvenile mice were similar to those in normal infant mice. Additionally, clustering analysis revealed no significant differences in the intestinal flora in the mice from the different drinking water sources; however, dehydration significantly altered the composition of the genera compared to the other water source groups wherein water was provided ad libitum. Moreover, cognitive development was significantly disrupted by insufficient water intake, although the type of drinking water had no significant influence. Cognitive decline, measured by relative latency, was positively associated with the relative abundance of unclassified Erysipelotrichaceae that were in significantly high relative abundance in the dehydration group. These results suggest that the water quantity consumed, rather than the mineral content of drinking water, is imperative for shaping the early gut microbiota associated with cognitive development during infancy.
Collapse
Affiliation(s)
- Chong-Su Kim
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul, Republic of Korea
- Research Institute of Human Ecology, Seoul National University, Seoul, Republic of Korea
| | - Dong-Mi Shin
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul, Republic of Korea
- Research Institute of Human Ecology, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
29
|
Yao H, Zhang D, Yu H, Yuan H, Shen H, Lan X, Liu H, Chen X, Meng F, Wu X, Zhang G, Wang X. Gut microbiota regulates chronic ethanol exposure-induced depressive-like behavior through hippocampal NLRP3-mediated neuroinflammation. Mol Psychiatry 2023; 28:919-930. [PMID: 36280756 PMCID: PMC9908543 DOI: 10.1038/s41380-022-01841-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022]
Abstract
Chronic ethanol exposure (CEE), which can lead to neuroinflammation, is an increasing risk factor for depression disorder, but the underlying mechanism is not clear. Recent observations have revealed the associations among psychiatric disorders, ethanol exposure and alterations of the gut microbiota. Here, we found that CEE induced depressive-like behavior, which could be alleviated by probiotics and transferred from donor to recipient mice by fecal microbiota transplantation (FMT). Neuroinflammation and the activation of the NLRP3 inflammasome were also observed in recipient mice. The downregulation of NLRP3 in the hippocampus mitigated CEE-induced depressive-like behavior and neuroinflammation but had no significant effect on FMT recipient mice. Moreover, elevated serum inflammatory factors in recipient mice showed a significant mediation effect between the gut microbiota and depressive-like behavior. Together, our study findings indicate that the gut microbiota contributes to both hippocampal NLRP3-mediated neuroinflammation and depressive-like behavior induced by CEE, which may open avenues for potential interventions against CEE-associated psychiatric disorders.
Collapse
Affiliation(s)
- Hui Yao
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China ,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China
| | - Dalin Zhang
- grid.412636.40000 0004 1757 9485Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001 Liaoning PR China
| | - Hao Yu
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China ,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China
| | - Huiya Yuan
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884Department of Forensic Analytical Toxicology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China
| | - Hui Shen
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China ,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China
| | - Xinze Lan
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China ,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China
| | - Hao Liu
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China ,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China
| | - Xiaohuan Chen
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China ,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China
| | - Fanyue Meng
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China ,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China
| | - Xu Wu
- grid.412449.e0000 0000 9678 1884Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122 Liaoning PR China ,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122 Liaoning PR China ,grid.412449.e0000 0000 9678 1884China Medical University Center of Forensic Investigation, Shenyang, 110122 Liaoning PR China
| | - Guohua Zhang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122, Liaoning, PR China. .,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122, Liaoning, PR China. .,China Medical University Center of Forensic Investigation, Shenyang, 110122, Liaoning, PR China.
| | - Xiaolong Wang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang, 110122, Liaoning, PR China. .,Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122, Liaoning, PR China. .,China Medical University Center of Forensic Investigation, Shenyang, 110122, Liaoning, PR China.
| |
Collapse
|
30
|
Li X, Zhao K, Chen J, Ni Z, Yu Z, Hu L, Qin Y, Zhao J, Peng W, Lu L, Gao X, Sun H. Diurnal changes of the oral microbiome in patients with alcohol dependence. Front Cell Infect Microbiol 2022; 12:1068908. [PMID: 36579346 PMCID: PMC9791055 DOI: 10.3389/fcimb.2022.1068908] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
Background Saliva secretion and oral microbiota change in rhythm with our biological clock. Dysbiosis of the oral microbiome and alcohol consumption have a two-way interactive impact, but little is known about whether the oral microbiome undergoes diurnal changes in composition and function during the daytime in patients with alcohol dependence (AD). Methods The impact of alcohol consumption on the diurnal salivary microbiome was examined in a case-control study of 32 AD patients and 21 healthy control (HC) subjects. We tested the changes in microbial composition and individual taxon abundance by 16S rRNA gene sequencing. Results The present study is the first report showing that alcohol consumption enhanced the richness of the salivary microbiome and lowered the evenness. The composition of the oral microbiota changed significantly in alcohol-dependent patients. Additionally, certain genera were enriched in the AD group, including Actinomyces, Leptotrichia, Sphaerochaeta and Cyanobacteria, all of which have pathogenic effects on the host. There is a correlation between liver enzymes and oral microbiota. KEGG function analysis also showed obvious alterations during the daytime. Conclusion Alcohol drinking influences diurnal changes in the oral microbiota, leading to flora disturbance and related functional impairment. In particular, the diurnal changes of the oral microbiota may open avenues for potential interventions that can relieve the detrimental consequences of AD.
Collapse
Affiliation(s)
- Xiangxue Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Kangqing Zhao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Jie Chen
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Zhaojun Ni
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Zhoulong Yu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Lingming Hu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Ying Qin
- Addiction Medicine Department, The Second People’s Hospital of Guizhou Province, Guizhou, China
| | - Jingwen Zhao
- Addiction Medicine Department, The Second People’s Hospital of Guizhou Province, Guizhou, China
| | - Wenjuan Peng
- Addiction Medicine Department, The Second People’s Hospital of Guizhou Province, Guizhou, China
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Xuejiao Gao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China,*Correspondence: Xuejiao Gao, ; Hongqiang Sun,
| | - Hongqiang Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China,*Correspondence: Xuejiao Gao, ; Hongqiang Sun,
| |
Collapse
|
31
|
Chen L, Yuan F, Chen S, Li X, Kong L, Zhang W. Potential Role of Host Microbiome in Areca Nut-Associated Carcinogenesis and Addiction. Molecules 2022; 27:8171. [PMID: 36500264 PMCID: PMC9739811 DOI: 10.3390/molecules27238171] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/21/2022] [Accepted: 08/30/2022] [Indexed: 11/25/2022] Open
Abstract
Areca nut (AN) is widely consumed all over the world, bringing great harm to human health and economy. Individuals with AN chewing are at high risk of cardiovascular disease and impaired immune system and metabolic system. Despite a growing number of studies having reported on the adverse effects brought by AN chewing, the exact mechanism of it is limited and the need for additional exploration remains. In recent years, the interaction between microorganisms, especially intestinal microorganism and host, has been extensively studied. AN chewing might disrupt the oral and intestinal microbiota communities through direct connect with the microbes it contains, altering PH, oxygen of oral and intestinal microenvironment, and disturbing the immune homeostasis. These mechanisms provide insights into the interplay between areca nut and host microbiota. Emerging studies have proposed that bidirectional interaction between polyphenols and intestinal microbes might play a potential role in the divergence of polyphenol, extracted from AN, among individuals with or without AN-induced cancer development and progression. Although some AN chewers have been aware of the harmful effects brought by AN, they cannot abolish this habit because of the addiction of AN. Increasing studies have tried to revealed that gut microbiota might influence the onset/development of addictive behaviors. Altogether, this review summarizes the possible reasons for the disturbance of host microbiota caused by areca nut chewing and clarifies the complex interaction between human microbiome and major constituents and the addiction and carcinogenicity of AN, tempting to provide novel insights into the development and utilization of it, and to control the adverse consequences caused by AN chewing.
Collapse
Affiliation(s)
- Lihui Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410078, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, China
| | - Fulai Yuan
- Health Management Center, Xiangya Hospital, Central South University, Changsha 410078, China
| | - Sifang Chen
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, Xiamen 361000, China
| | - Xiong Li
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510060, China
| | - Lingyu Kong
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410078, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, Changsha 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, China
| |
Collapse
|
32
|
Hou X, Rong C, Zhang Q, Song S, Cong Y, Zhang HT. Cyclic Nucleotide Phosphodiesterases in Alcohol Use Disorders: Involving Gut Microbiota. Int J Neuropsychopharmacol 2022; 26:70-79. [PMID: 36087271 PMCID: PMC9850663 DOI: 10.1093/ijnp/pyac060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 08/18/2022] [Accepted: 09/09/2022] [Indexed: 01/22/2023] Open
Abstract
Alcohol abuse is 1 of the most significant public health problems in the world. Chronic, excessive alcohol consumption not only causes alcohol use disorder (AUD) but also changes the gut and lung microbiota, including bacterial and nonbacterial types. Both types of microbiota can release toxins, further damaging the gastrointestinal and respiratory tracts; causing inflammation; and impairing the functions of the liver, lung, and brain, which in turn deteriorate AUD. Phosphodiesterases (PDEs) are critical in the control of intracellular cyclic nucleotides, including cyclic adenosine monophosphate and cyclic guanosine monophosphate. Inhibition of certain host PDEs reduces alcohol consumption and attenuates alcohol-related impairment. These PDEs are also expressed in the microbiota and may play a role in controlling microbiota-associated inflammation. Here, we summarize the influences of alcohol on gut/lung bacterial and nonbacterial microbiota as well as on the gut-liver/brain/lung axis. We then discuss the relationship between gut and lung microbiota-mediated PDE signaling and AUD consequences in addition to highlighting PDEs as potential targets for treatment of AUD.
Collapse
Affiliation(s)
- Xueqin Hou
- Correspondence: Xueqin Hou, PhD, Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China ()
| | | | - Qiwei Zhang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China
| | - Shuangshuang Song
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China
| | - Yifan Cong
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong 271016, P.R. China
| | - Han-Ting Zhang
- Han-Ting Zhang, MD, PhD, Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong 266073, P.R. China ()
| |
Collapse
|
33
|
Yao H, Zhang D, Yu H, Shen H, Lan X, Liu H, Chen X, Wu X, Zhang G, Wang X. Chronic ethanol exposure induced anxiety‐like behaviour by altering gut microbiota and GABA system. Addict Biol 2022; 27:e13203. [DOI: 10.1111/adb.13203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/23/2022] [Accepted: 06/09/2022] [Indexed: 12/16/2022]
Affiliation(s)
- Hui Yao
- Department of Forensic Pathology China Medical University School of Forensic Medicine Shenyang Liaoning China
- Liaoning Province Key Laboratory of Forensic Bio‐evidence Sciences Shenyang Liaoning China
- China Medical University Center of Forensic Investigation Shenyang Liaoning China
| | - Dalin Zhang
- Department of Thyroid Surgery The 1st Affiliated Hospital of China Medical University Shenyang China
| | - Hao Yu
- Department of Forensic Pathology China Medical University School of Forensic Medicine Shenyang Liaoning China
- Liaoning Province Key Laboratory of Forensic Bio‐evidence Sciences Shenyang Liaoning China
- China Medical University Center of Forensic Investigation Shenyang Liaoning China
| | - Hui Shen
- Department of Forensic Pathology China Medical University School of Forensic Medicine Shenyang Liaoning China
- Liaoning Province Key Laboratory of Forensic Bio‐evidence Sciences Shenyang Liaoning China
- China Medical University Center of Forensic Investigation Shenyang Liaoning China
| | - Xinze Lan
- Department of Forensic Pathology China Medical University School of Forensic Medicine Shenyang Liaoning China
- Liaoning Province Key Laboratory of Forensic Bio‐evidence Sciences Shenyang Liaoning China
- China Medical University Center of Forensic Investigation Shenyang Liaoning China
| | - Hao Liu
- Department of Forensic Pathology China Medical University School of Forensic Medicine Shenyang Liaoning China
- Liaoning Province Key Laboratory of Forensic Bio‐evidence Sciences Shenyang Liaoning China
- China Medical University Center of Forensic Investigation Shenyang Liaoning China
| | - Xiaohuan Chen
- Department of Forensic Pathology China Medical University School of Forensic Medicine Shenyang Liaoning China
- Liaoning Province Key Laboratory of Forensic Bio‐evidence Sciences Shenyang Liaoning China
- China Medical University Center of Forensic Investigation Shenyang Liaoning China
| | - Xu Wu
- Department of Forensic Pathology China Medical University School of Forensic Medicine Shenyang Liaoning China
- Liaoning Province Key Laboratory of Forensic Bio‐evidence Sciences Shenyang Liaoning China
- China Medical University Center of Forensic Investigation Shenyang Liaoning China
| | - Guohua Zhang
- Department of Forensic Pathology China Medical University School of Forensic Medicine Shenyang Liaoning China
- Liaoning Province Key Laboratory of Forensic Bio‐evidence Sciences Shenyang Liaoning China
- China Medical University Center of Forensic Investigation Shenyang Liaoning China
| | - Xiaolong Wang
- Department of Forensic Pathology China Medical University School of Forensic Medicine Shenyang Liaoning China
- Liaoning Province Key Laboratory of Forensic Bio‐evidence Sciences Shenyang Liaoning China
- China Medical University Center of Forensic Investigation Shenyang Liaoning China
| |
Collapse
|
34
|
Li J, Zou C, Liu Y. Amelioration of Ovalbumin-Induced Food Allergy in Mice by Targeted Rectal and Colonic Delivery of Cyanidin-3-O-Glucoside. Foods 2022; 11:foods11111542. [PMID: 35681291 PMCID: PMC9180400 DOI: 10.3390/foods11111542] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/19/2022] [Accepted: 05/21/2022] [Indexed: 02/04/2023] Open
Abstract
Targeted rectal and colonic delivery is an effective strategy to exploit the biological functions of polyphenols. This work investigated the anti-food allergy (FA) activity of cyanidin-3-O-glucoside (C3G) delivered by enteric sodium alginate in vivo. The results showed that through targeted rectal and colonic delivery, the C3G showed better results in ameliorating clinical allergic symptoms, diarrhea, and serological indicators including ovalbumin-specific IgE, histamine, and mast cell protease-1. The C3G was more efficient in enhancing the intestinal epithelial barrier by up-regulating the tight junction protein expression and promoting secretory IgA and β-defensin secretion. The improved bioactivity in regulating T helper (Th)1/Th2 immune balance in the intestinal mucosa was also observed. Compared with the intestinal microbiota structure of the model group, targeted rectal and colonic delivery of C3G was able to bring the abundance of Bacteroidota and Firmicutes close to the levels found in normal mice. Furthermore, there was an evident increase in beneficial bacteria in the intestinal flora, such as Lactobacillus and Odoribacter, and a decrease in pathogenic bacteria like Helicobacter and Turicibacter. Therefore, the anti-FA activity of C3G could be increased via targeted rectal and colonic delivery, while the mechanism might be attributed to the regulation of intestinal microecological homeostasis.
Collapse
Affiliation(s)
- Jie Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China;
| | - Chao Zou
- Gaoan Public Inspection and Testing Center, Gao’an 330800, China;
| | - Yixiang Liu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China;
- Collaborative Innovation Center of Provincial and Ministerial Co-Construction for Marine Food Deep Processing, Dalian Polytechnic University, Dalian 116034, China
- Correspondence:
| |
Collapse
|
35
|
Supplementation with sodium butyrate protects against antibiotic-induced increases in ethanol consumption behavior in mice. Alcohol 2022; 100:1-9. [PMID: 34999234 PMCID: PMC8983552 DOI: 10.1016/j.alcohol.2021.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/15/2021] [Accepted: 12/21/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND We have recently reported that oral treatment of adult male C57BL/6J mice with a non-absorbable antibiotic cocktail resulted in an increase in ethanol intake and in significant reductions in butyrate-producing gut microbiota populations. This work led us to hypothesize that reduction in butyrate levels within the gut is linked to antibiotic-induced increases in voluntary ethanol consumption. OBJECTIVE This study tested whether ad libitum sodium butyrate supplementation can prevent antibiotic-induced ethanol consumption in mice. METHODS Sodium butyrate was provided to adult male C57BL/6J mice in drinking water alone or in combination with antibiotic cocktail. Effects on ethanol (20%) intake were measured using drinking in the dark and modified 2-bottle choice paradigms. Body parameters, food and liquid intake, cecum, and adipose tissues were measured during and/or at the conclusion of the drinking in the dark study. Cecal 16s rRNA was analyzed for microbiota diversity and changes in specific bacterial phyla/species. RESULTS In drinking in the dark, sodium butyrate supplementation prevented antibiotic-induced increases in ethanol intake without altering basal ethanol consumption. Furthermore, sodium butyrate supplementation lowered ethanol preference in the 2-bottle choice study. Ethanol intake was correlated to specific bacterial phyla/species. Sodium butyrate did not affect the changes in microbiota diversity and composition induced by antibiotic cocktail. CONCLUSIONS The findings support a role of gut microbiota-derived butyrate in regulating alcohol-induced behaviors. Additionally, the work contributes to efforts in development of novel microbiome-based strategies as novel preventative and intervention-based therapeutics to address alcohol use disorder.
Collapse
|
36
|
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
|
37
|
Herlihy B, Roy S. Gut-Microbiome Implications in Opioid Use Disorder and Related Behaviors. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2022; 2:10311. [PMID: 38390617 PMCID: PMC10880781 DOI: 10.3389/adar.2022.10311] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/25/2022] [Indexed: 02/24/2024]
Abstract
Substance use disorder (SUD) is a prevalent disease that has caused hundreds of thousands of deaths and affected the lives of even more. Despite its global impact, there is still no known cure for SUD, or the psychological symptoms associated with drug use. Many of the behavioral consequences of drug use prevent people from breaking the cycle of addiction or cause them to relapse back into the cycle due to the physical and psychological consequences of withdrawal. Current research is aimed at understanding the cause of these drug related behaviors and therapeutically targeting them as a mechanism to break the addiction cycle. Research on opioids suggests that the changes in the microbiome during drug use modulated drug related behaviors and preventing these microbial changes could attenuate behavioral symptoms. This review aims to highlight the relationship between the changes in the microbiome and behavior during opioid treatment, as well as highlight the additional research needed to understand the mechanism in which the microbiome modulates behavior to determine the best therapeutic course of action.
Collapse
Affiliation(s)
- Bridget Herlihy
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neuroscience, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sabita Roy
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
38
|
Yao H, Zhang D, Yu H, Shen H, Lan X, Liu H, Chen X, Wu X, Zhang G, Wang X. AMPAkine CX516 alleviated chronic ethanol exposure-induced neurodegeneration and depressive-like behavior in mice. Toxicol Appl Pharmacol 2022; 439:115924. [PMID: 35181401 DOI: 10.1016/j.taap.2022.115924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 12/15/2022]
Abstract
Chronic ethanol exposure (CEE) is associated with greater neurodegenerative effects and an increased risk of depression disorder. The AMPAR is thought to be involved in depression and a reduction in its GluA1 subunit was observed in the mouse hippocampus after CEE. AMPAkines are positive allosteric modulators of the AMPA receptor and have improved depressive-like behavior. However, the role of AMPARs in CEE-induced depressive-like behavior is not clear. It is unclear whether AMPAkines, positive allosteric agonists of AMPARs, protect against ethanol-induced depression. We investigated the effects of CX516 on ethanol-induced depressive-like behavior in a mouse model. CX516 (5 mg/kg) administration alleviated 20% (m/V) ethanol-induced depressive-like behavior in mice. Furthermore, CX516 significantly diminished the inhibition of the ERK1/2-BDNF-TrkB pathway in the hippocampus of ethanol-exposed mice. In addition, CX516 attenuated the levels of pro-inflammatory (IL-6, IL-1β), apoptosis (BAX, BCL-2), and neurodegeneration (FJC) in the mouse hippocampus induced by CEE.
Collapse
Affiliation(s)
- Hui Yao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Dalin Zhang
- Department of Thyroid Surgery, the 1st Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, PR China
| | - Hao Yu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Hui Shen
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Xinze Lan
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Hao Liu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Xiaohuan Chen
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Xu Wu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Guohua Zhang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China.
| | - Xiaolong Wang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, PR China.
| |
Collapse
|
39
|
Gu Y, Han Y, Ren S, Zhang B, Zhao Y, Wang X, Zhang S, Qin J. Correlation among gut microbiota, fecal metabolites and autism-like behavior in an adolescent valproic acid-induced rat autism model. Behav Brain Res 2022; 417:113580. [PMID: 34555431 DOI: 10.1016/j.bbr.2021.113580] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/18/2021] [Accepted: 09/08/2021] [Indexed: 12/26/2022]
Abstract
This study aims to understand the relationship between fecal metabolites and gut microbiota in an adolescent valproic acid-induced rat autism model (VPA-exposed offspring). We analyzed the fecal samples of VPA-exposed offspring using 16S rRNA gene sequencing and untargeted metabolomics. Autism-like behavior was evaluated by a three-chamber sociability test and a self-grooming test. Based on these data, we analyzed the association among fecal metabolites, gut microbiota and autism-like behavior. Behavioral tests showed that VPA-exposed offspring displayed typical autism-like behavior. Forty-nine named differential fecal metabolites and 14 enriched KEGG pathways were identified between the VPA and control groups. Five fecal metabolites may be used as characteristic metabolites. The richness and diversity of gut microbiota did not differ between the two groups, while the overall composition of gut microbiota was significantly different. Candidatus_Saccharimonas, Desulfovibrio, [Eubacterium]_xylanophilum_group and Ruminococcus_2 were the characteristic genera of VPA-exposed offspring. Correlation analysis revealed a tight relationship among gut microbiota, fecal metabolites and autistic behavior in VPA-exposed offspring. This study illustrates that specific alterations in gut microbiota and fecal metabolites may be regarded as characteristics of VPA-exposed offspring. The characteristic gut microbiota and fecal metabolites as well as their relationship may play a crucial role in autism-like behavior caused by prenatal exposure to VPA.
Collapse
Affiliation(s)
- Youyu Gu
- Department of Pediatrics, Peking University People's Hospital, Beijing 100044, China
| | - Ying Han
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China.
| | - Shimeng Ren
- Department of Pediatrics, Peking University People's Hospital, Beijing 100044, China
| | - Bi Zhang
- Department of Pediatrics, Peking University People's Hospital, Beijing 100044, China
| | - Yihan Zhao
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - Xiaoxi Wang
- Neuroscience Research Institute, Peking University, Beijing 100191, China
| | - Shaobin Zhang
- Beijing Gutgene Technology Co. Ltd, Beijing 100085, China
| | - Jiong Qin
- Department of Pediatrics, Peking University People's Hospital, Beijing 100044, China.
| |
Collapse
|
40
|
Neuroprotective effect of fucoidan by regulating gut-microbiota-brain axis in alcohol withdrawal mice. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
41
|
Fu X, Chen T, Cai J, Liu B, Zeng Y, Zhang X. The Microbiome-Gut-Brain Axis, a Potential Therapeutic Target for Substance-Related Disorders. Front Microbiol 2021; 12:738401. [PMID: 34690981 PMCID: PMC8526971 DOI: 10.3389/fmicb.2021.738401] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/31/2021] [Indexed: 12/31/2022] Open
Abstract
Substance addiction is a complex worldwide public health problem. It endangers both personal life and social stability, causing great loss on economy. Substance-related disorder is considered to be a complicated chronic brain disorder. It resulted from interactions among pharmacological properties of addictive substances, individual susceptibility, and social–environmental factors. Unfortunately, there is still no ideal treatment for this disorder. Recent lines of evidence suggest that gut microbiome may play an important role in the pathogenesis of neuropsychiatric disorders, including substance-related disorders. This review summarizes the research on the relationship between gut microbiome and substance-related disorders, including different types of substance, different individual susceptibility, and the occurrence and development of substance-induced mental disorders. We also discuss the potentiation of gut microbiome in the treatment of substance-related disorders, especially in the treatment of substance-induced mental disorders and manipulation on individuals’ responsiveness to addictive substances.
Collapse
Affiliation(s)
- Xuan Fu
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center on Mental Disorders, Changsha, China.,National Technology Institute on Mental Disorders, Changsha, China.,Hunan Medical Center for Mental Health, Changsha, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China.,Mental Health Institute of Central South University, Changsha, China
| | - Ti Chen
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jingda Cai
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center on Mental Disorders, Changsha, China.,National Technology Institute on Mental Disorders, Changsha, China.,Hunan Medical Center for Mental Health, Changsha, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China.,Mental Health Institute of Central South University, Changsha, China
| | - Bo Liu
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center on Mental Disorders, Changsha, China.,National Technology Institute on Mental Disorders, Changsha, China.,Hunan Medical Center for Mental Health, Changsha, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China.,Mental Health Institute of Central South University, Changsha, China
| | - Yaohui Zeng
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center on Mental Disorders, Changsha, China.,National Technology Institute on Mental Disorders, Changsha, China.,Hunan Medical Center for Mental Health, Changsha, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China.,Mental Health Institute of Central South University, Changsha, China
| | - Xiaojie Zhang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center on Mental Disorders, Changsha, China.,National Technology Institute on Mental Disorders, Changsha, China.,Hunan Medical Center for Mental Health, Changsha, China.,Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China.,Mental Health Institute of Central South University, Changsha, China
| |
Collapse
|
42
|
Ge T, Yao X, Zhao H, Yang W, Zou X, Peng F, Li B, Cui R. Gut microbiota and neuropsychiatric disorders: Implications for neuroendocrine-immune regulation. Pharmacol Res 2021; 173:105909. [PMID: 34543739 DOI: 10.1016/j.phrs.2021.105909] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/08/2021] [Accepted: 09/15/2021] [Indexed: 12/17/2022]
Abstract
Recently, increasing evidence has shown gut microbiota dysbiosis might be implicated in the physiological mechanisms of neuropsychiatric disorders. Altered microbial community composition, diversity and distribution traits have been reported in neuropsychiatric disorders. However, the exact pathways by which the intestinal microbiota contribute to neuropsychiatric disorders remain largely unknown. Given that the onset and progression of neuropsychiatric disorders are characterized with complicated alterations of neuroendocrine and immunology, both of which can be continually affected by gut microbiota via "microbiome-gut-brain axis". Thus, we assess the complicated crosstalk between neuroendocrine and immunological regulation might underlie the mechanisms of gut microbiota associated with neuropsychiatric disorders. In this review, we summarized clinical and preclinical evidence on the role of the gut microbiota in neuropsychiatry disorders, especially in mood disorders and neurodevelopmental disorders. This review may elaborate the potential mechanisms of gut microbiota implicating in neuroendocrine-immune regulation and provide a comprehensive understanding of physiological mechanisms for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Tongtong Ge
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Xiaoxiao Yao
- Department of Hepatopancreatobiliary Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Haisheng Zhao
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Xiaohan Zou
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Fanzhen Peng
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
43
|
Specific Changes in the Mammalian Gut Microbiome as a Biomarker for Oxytocin-Induced Behavioral Changes. Microorganisms 2021; 9:microorganisms9091938. [PMID: 34576833 PMCID: PMC8465812 DOI: 10.3390/microorganisms9091938] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/22/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
Prolonged exposure to psychiatric pharmacological agents is often associated with marked gastrointestinal phenomena, including changes in food intake, bowel motility, gastric emptying, and transit time. Those changes are reflected in the gut microbiota composition of the patient and can, therefore, be objectively measured. This is in contrast to the standard psychiatric evaluation of patients, which includes symptoms that are subjectively assessed (i.e., mood, anxiety level, perception, thought disorders, etc.). The association between a drug’s effect on the microbiota and psychiatric symptoms may allow for quantifiable surrogate markers of treatment effectiveness. Changes in the levels of specific drug-sensitive bacterial species can, thus, potentially serve as biomarkers for the intake and effectiveness of psychiatric drugs. Here, we show substantial microbiota changes that were associated with oxytocin administration and the decreased anxiety/depression-like behaviors it conferred in a rat model of corticosterone-induced stress. Compared with oxytocin, citalopram produced more minor effects on the rats’ microbiota. Alterations in the gut microbiota may, therefore, reflect the consumption and effectiveness of some psychiatric drugs.
Collapse
|
44
|
Yang C, Fu X, Hao W, Xiang X, Liu T, Yang B, Zhang X. Gut dysbiosis associated with the rats' responses in methamphetamine-induced conditioned place preference. Addict Biol 2021; 26:e12975. [PMID: 33094505 DOI: 10.1111/adb.12975] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/11/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023]
Abstract
Methamphetamine (MA) is a potent stimulant and notoriously addictive. Individuals respond to MA effects differently and thus have a varying susceptible risk of developing MA use disorder. Cumulative evidence has indicated that gut dysbiosis contributes to behavioral response to drug effects. However, the role of gut microbiota in the susceptible risk of developing MA use disorder has remained elusive. Using an MA-induced conditioned place preference (CPP) rat model, we administrated the same dose of MA to rats, which then showed distinct preferences in drug-related place, indicating their different responses to MA. From all of the MA-exposed rats, the eight with the highest CPP scores were labeled as group high CPP (H-CPP), and the eight with the lowest were labeled as group low CPP (L-CPP). By 16S ribosomal RNA (rRNA) sequencing, we found that the gut microbiota compositions differed between H-CPP and L-CPP. Specifically, Akkermansia was significantly higher in H-CPP and positively correlated with the CPP scores. Notably, H-CPP and L-CPP differed in the gut microbiota composition prior to the CPP training; Ruminococcus was the dominant phylotype in H-CPP at baseline. More importantly, rats pretreated by antibiotics showed a significantly stronger MA-induced CPP than did the controls. Our study demonstrates that the gut dysbiosis was associated with the MA-induced CPP, indicating that the gut microbiota might be important modulators for MA-induced behavior and vulnerability to MA use disorder.
Collapse
Affiliation(s)
- Cheng Yang
- Department of Psychiatry, The Second Xiangya Hospital Central South University Changsha China
- National Clinical Research Center on Mental Disorders (Xiangya) Changsha China
- National Technology Institute on Mental Disorders Changsha China
- Hunan Key Laboratory of Psychiatry and Mental Health Changsha China
- Mental Health Institute of Central South University Changsha China
| | - Xiaoya Fu
- Department of Psychiatry, The Second Xiangya Hospital Central South University Changsha China
- National Clinical Research Center on Mental Disorders (Xiangya) Changsha China
- National Technology Institute on Mental Disorders Changsha China
- Hunan Key Laboratory of Psychiatry and Mental Health Changsha China
- Mental Health Institute of Central South University Changsha China
| | - Wei Hao
- Department of Psychiatry, The Second Xiangya Hospital Central South University Changsha China
- National Clinical Research Center on Mental Disorders (Xiangya) Changsha China
- National Technology Institute on Mental Disorders Changsha China
- Hunan Key Laboratory of Psychiatry and Mental Health Changsha China
- Mental Health Institute of Central South University Changsha China
| | - Xiaojun Xiang
- Department of Psychiatry, The Second Xiangya Hospital Central South University Changsha China
- National Clinical Research Center on Mental Disorders (Xiangya) Changsha China
- National Technology Institute on Mental Disorders Changsha China
- Hunan Key Laboratory of Psychiatry and Mental Health Changsha China
- Mental Health Institute of Central South University Changsha China
| | - Tieqiao Liu
- Department of Psychiatry, The Second Xiangya Hospital Central South University Changsha China
- National Clinical Research Center on Mental Disorders (Xiangya) Changsha China
- National Technology Institute on Mental Disorders Changsha China
- Hunan Key Laboratory of Psychiatry and Mental Health Changsha China
- Mental Health Institute of Central South University Changsha China
| | - Bao‐Zhu Yang
- Department of Psychiatry Yale University School of Medicine New Haven CT USA
| | - Xiaojie Zhang
- Department of Psychiatry, The Second Xiangya Hospital Central South University Changsha China
- National Clinical Research Center on Mental Disorders (Xiangya) Changsha China
- National Technology Institute on Mental Disorders Changsha China
- Hunan Key Laboratory of Psychiatry and Mental Health Changsha China
- Mental Health Institute of Central South University Changsha China
| |
Collapse
|
45
|
Guo Y, Zhu X, Zeng M, Qi L, Tang X, Wang D, Zhang M, Xie Y, Li H, Yang X, Chen D. A diet high in sugar and fat influences neurotransmitter metabolism and then affects brain function by altering the gut microbiota. Transl Psychiatry 2021; 11:328. [PMID: 34045460 PMCID: PMC8160265 DOI: 10.1038/s41398-021-01443-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 05/05/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota (GM) metabolites can modulate the physiology of the host brain through the gut-brain axis. We wished to discover connections between the GM, neurotransmitters, and brain function using direct and indirect methods. A diet with increased amounts of sugar and fat (high-sugar and high-fat (HSHF) diet) was employed to disturb the host GM. Then, we monitored the effect on pathology, neurotransmitter metabolism, transcription, and brain circularRNAs (circRNAs) profiles in mice. Administration of a HSHF diet-induced dysbacteriosis, damaged the intestinal tract, changed the neurotransmitter metabolism in the intestine and brain, and then caused changes in brain function and circRNA profiles. The GM byproduct trimethylamine-n-oxide could degrade some circRNAs. The basal level of the GM decided the conversion rate of choline to trimethylamine-n-oxide. A change in the abundance of a single bacterial strain could influence neurotransmitter secretion. These findings suggest that a new link between metabolism, brain circRNAs, and GM. Our data could enlarge the "microbiome-transcriptome" linkage library and provide more information on the gut-brain axis. Hence, our findings could provide more information on the interplay between the gut and brain to aid the identification of potential therapeutic markers and mechanistic solutions to complex problems encountered in studies of pathology, toxicology, diet, and nutrition development.
Collapse
Affiliation(s)
- Yinrui Guo
- grid.411866.c0000 0000 8848 7685School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangdong, Guangzhou 510120 China
| | - Xiangxiang Zhu
- grid.464309.c0000 0004 6431 5677State Key Laboratory of Applied Microbiology Southern China; Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application; Guangdong Open Laboratory of Applied Microbiology; Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070 China ,grid.258164.c0000 0004 1790 3548Academy of Life Sciences, Jinan University, Guangdong Province, Guangzhou, 510000 China
| | - Miao Zeng
- grid.464309.c0000 0004 6431 5677State Key Laboratory of Applied Microbiology Southern China; Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application; Guangdong Open Laboratory of Applied Microbiology; Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070 China ,grid.411304.30000 0001 0376 205XChengdu University of Traditional Chinese Medicine, Chengdu, 610075 China
| | - Longkai Qi
- grid.464309.c0000 0004 6431 5677State Key Laboratory of Applied Microbiology Southern China; Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application; Guangdong Open Laboratory of Applied Microbiology; Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070 China
| | - Xiaocui Tang
- grid.464309.c0000 0004 6431 5677State Key Laboratory of Applied Microbiology Southern China; Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application; Guangdong Open Laboratory of Applied Microbiology; Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070 China
| | - Dongdong Wang
- grid.464309.c0000 0004 6431 5677State Key Laboratory of Applied Microbiology Southern China; Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application; Guangdong Open Laboratory of Applied Microbiology; Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070 China
| | - Mei Zhang
- grid.411304.30000 0001 0376 205XChengdu University of Traditional Chinese Medicine, Chengdu, 610075 China
| | - Yizhen Xie
- grid.464309.c0000 0004 6431 5677State Key Laboratory of Applied Microbiology Southern China; Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application; Guangdong Open Laboratory of Applied Microbiology; Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070 China
| | - Hongye Li
- grid.258164.c0000 0004 1790 3548Academy of Life Sciences, Jinan University, Guangdong Province, Guangzhou, 510000 China
| | - Xin Yang
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China.
| | - Diling Chen
- State Key Laboratory of Applied Microbiology Southern China; Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application; Guangdong Open Laboratory of Applied Microbiology; Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China.
| |
Collapse
|
46
|
Nasef NA, Keshk WA, El-Meligy SM, Allah AAA, Ibrahim WM. Modulatory effect of simvastatin on redox status, caspase-3 expression, p-protein kinase B (p-Akt), and brain-derived neurotrophic factor (BDNF) in an ethanol-induced neurodegeneration model. Can J Physiol Pharmacol 2021; 99:478-489. [PMID: 33002367 DOI: 10.1139/cjpp-2020-0360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neurodegenerative diseases are a common cause of morbidity and mortality worldwide, with oxidative stress, inflammation, and protein aggregation representing the main underlying mechanisms that ultimately lead to cell death. Ethanol has shown strong neurodegenerative consequences in experimental animal brains. Statins are a class of lipid-lowering drugs with many pleotropic effects. Therefore, the aim of the present study was to explore the modulatory effect of simvastatin (10 mg·kg-1·day-1) before and after the development of neurodegeneration (for 55 and 25 days, respectively) on redox state, caspase-3 expression, p-protein kinase B (p-Akt), and brain-derived neurotrophic factor (BDNF) in ethanol-induced (15% ethanol solution for 55 days) neurodegeneration. Seventy female Albino Swiss mice were included and randomly divided into five groups: C, control group; E, ethanol group; ES, group treated with simvastatin from the first day of ethanol intake; E + S, group treated with simvastatin after neurodegeneration development; and S, simvastatin group. Administration of simvastatin from the first day improved the biochemical changes, suppressed apoptosis, and induced autophagy and neurogenesis; however, its administration after the development of neurodegeneration resulted in partial improvement. The histopathological findings confirmed the biochemical changes. In conclusion, simvastatin has a neuroprotective effect against the development of ethanol-induced neurodegeneration and its progression.
Collapse
Affiliation(s)
- Nahla A Nasef
- Department of Medical Biochemistry & Molecular Biology Faculty of Medicine, Tanta University, Tanta, Egypt
- Department of Medical Biochemistry & Molecular Biology Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Walaa A Keshk
- Department of Medical Biochemistry & Molecular Biology Faculty of Medicine, Tanta University, Tanta, Egypt
- Department of Medical Biochemistry & Molecular Biology Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Salwa M El-Meligy
- Department of Medical Biochemistry & Molecular Biology Faculty of Medicine, Tanta University, Tanta, Egypt
- Department of Medical Biochemistry & Molecular Biology Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Ahmed A Abd Allah
- Department of Medical Biochemistry & Molecular Biology Faculty of Medicine, Tanta University, Tanta, Egypt
- Department of Medical Biochemistry & Molecular Biology Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Wafaa M Ibrahim
- Department of Medical Biochemistry & Molecular Biology Faculty of Medicine, Tanta University, Tanta, Egypt
- Department of Medical Biochemistry & Molecular Biology Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
47
|
Angoa-Pérez M, Kuhn DM. Evidence for Modulation of Substance Use Disorders by the Gut Microbiome: Hidden in Plain Sight. Pharmacol Rev 2021; 73:571-596. [PMID: 33597276 PMCID: PMC7896134 DOI: 10.1124/pharmrev.120.000144] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The gut microbiome modulates neurochemical function and behavior and has been implicated in numerous central nervous system (CNS) diseases, including developmental, neurodegenerative, and psychiatric disorders. Substance use disorders (SUDs) remain a serious threat to the public well-being, yet gut microbiome involvement in drug abuse has received very little attention. Studies of the mechanisms underlying SUDs have naturally focused on CNS reward circuits. However, a significant body of research has accumulated over the past decade that has unwittingly provided strong support for gut microbiome participation in drug reward. β-Lactam antibiotics have been employed to increase glutamate transporter expression to reverse relapse-induced release of glutamate. Sodium butyrate has been used as a histone deacetylase inhibitor to prevent drug-induced epigenetic alterations. High-fat diets have been used to alter drug reward because of the extensive overlap of the circuitry mediating them. This review article casts these approaches in a different light and makes a compelling case for gut microbiome modulation of SUDs. Few factors alter the structure and composition of the gut microbiome more than antibiotics and a high-fat diet, and butyrate is an endogenous product of bacterial fermentation. Drugs such as cocaine, alcohol, opiates, and psychostimulants also modify the gut microbiome. Therefore, their effects must be viewed on a complex background of cotreatment-induced dysbiosis. Consideration of the gut microbiome in SUDs should have the beneficial effects of expanding the understanding of SUDs and aiding in the design of new therapies based on opposing the effects of abused drugs on the host's commensal bacterial community. SIGNIFICANCE STATEMENT: Proposed mechanisms underlying substance use disorders fail to acknowledge the impact of drugs of abuse on the gut microbiome. β-Lactam antibiotics, sodium butyrate, and high-fat diets are used to modify drug seeking and reward, overlooking the notable capacity of these treatments to alter the gut microbiome. This review aims to stimulate research on substance abuse-gut microbiome interactions by illustrating how drugs of abuse share with antibiotics, sodium butyrate, and fat-laden diets the ability to modify the host microbial community.
Collapse
Affiliation(s)
- Mariana Angoa-Pérez
- Research and Development Service, John D. Dingell VA Medical Center, and Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan
| | - Donald M Kuhn
- Research and Development Service, John D. Dingell VA Medical Center, and Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan
| |
Collapse
|
48
|
An G, Zhang Y, Fan L, Chen J, Wei M, Li C, Chen X, Zhang L, Yang D, Wang J. Integrative Analysis of Vaginal Microorganisms and Serum Metabolomics in Rats With Estrous Cycle Disorder Induced by Long-Term Heat Exposure Based on 16S rDNA Gene Sequencing and LC/MS-Based Metabolomics. Front Cell Infect Microbiol 2021; 11:595716. [PMID: 33738264 PMCID: PMC7962411 DOI: 10.3389/fcimb.2021.595716] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/11/2021] [Indexed: 12/26/2022] Open
Abstract
Long term heat exposure (HE) leads to estrous cycle disorder (ECD) in female rats and damages reproductive function. However, the regulation mechanism of vaginal microorganisms and serum metabolomics remains unclear. This study aimed to explore the effects of microbes on the vaginal secretions of rats with ECD and describe the serum metabolomics characteristics and their relationship with vaginal microorganisms. The alterations in the serum levels of neurotransmitters were used to verify the possible regulatory pathways. The relative abundance, composition, and colony interaction network of microorganisms in the vaginal secretions of rats with ECD changed significantly. The metabolomics analysis identified 22 potential biomarkers in the serum including lipid metabolism, amino acid metabolism, and mammalian target of rapamycin and gonadotropin-releasing hormone (GnRH) signaling pathways. Further, 52 pairs of vaginal microbiota–serum metabolites correlations (21 positive and 31 negative) were determined. The abundance of Gardnerella correlated positively with the metabolite L-arginine concentration and negatively with the oleic acid concentration. Further, a negative correlation was found between the abundance of Pseudomonas and the L-arginine concentration and between the metabolite benzoic acid concentration and the abundance of Adlercreutzia. These four bacteria–metabolite pairs had a direct or indirect relationship with the estrous cycle and reproduction. The glutamine, glutamate, and dopamine levels were significantly uncontrolled. The former two were closely related to GnRH signaling pathways involved in the development and regulation of HE-induced ECD in rats. Serum neurotransmitters partly reflected the regulatory effect of vaginal microorganisms on the host of HE-induced ECD, and glutamatergic neurotransmitters might be closely related to the alteration in vaginal microorganisms. These findings might help comprehend the mechanism of HE-induced ECD and propose a new intervention based on vaginal microorganisms.
Collapse
Affiliation(s)
- GaiHong An
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Yu Zhang
- Department of Endocrinology, Tianjin Central Hospital of Gynecology and Obstetrics, Tianjin, China
| | - LiJun Fan
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - JiaJun Chen
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - MengFan Wei
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Chao Li
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - XueWei Chen
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Li Zhang
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - DanFeng Yang
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Jing Wang
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| |
Collapse
|
49
|
Han QQ, Fu Y, Le JM, Pilot A, Cheng S, Chen PQ, Wu H, Wan GQ, Gu XF. Electroacupuncture may alleviate behavioral defects via modulation of gut microbiota in a mouse model of Parkinson's disease. Acupunct Med 2021; 39:501-511. [PMID: 33557583 DOI: 10.1177/0964528421990658] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Parkinson's disease (PD) is a chronic neurodegenerative disease involving non-motor symptoms, of which gastrointestinal disorders are the most common. In light of recent results, intestinal dysfunction may be involved in the pathogenesis of PD. Electroacupuncture (EA) has shown potential effects, although the underlying mechanism remains mostly unknown. We speculated that EA could relieve the behavioral defects of PD, and that this effect would be associated with modulation of the gut microbiota. METHODS Mice were randomly divided into three groups: control, PD + MA (manual acupuncture), and PD + EA. MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) was used to establish the mouse model of PD. Rotarod performance tests, open field tests, and pole tests were carried out to assess motor deficiencies. Immunohistochemistry was conducted to examine the survival of dopaminergic neurons. 16S ribosomal RNA (rRNA) gene sequencing was applied to investigate the alterations of the gut microbiome. Quantitative real-time polymerase chain reaction (PCR) was performed to characterize the messenger RNA (mRNA) levels of pro-inflammatory and anti-inflammatory cytokines. RESULTS We found that EA was able to alleviate the behavioral defects in the rotarod performance test and pole test, and partially rescue the significant loss of dopaminergic neurons in the substantia nigra (SN) chemically induced by MPTP in mice. Moreover, the PD + MA mice showed a tendency toward decreased intestinal microbial alpha diversity, while EA significantly reversed it. The abundance of Erysipelotrichaceae was significantly increased in PD + MA mice, and the alteration was also reversed by EA. In addition, the pro-inflammatory cytokines interleukin (IL)-6 and tumor necrosis factor (TNF)-α were substantially increased in the SN of PD + MA mice, an effect that was reversed by EA. CONCLUSION These results suggest that EA may alleviate behavioral defects via modulation of gut microbiota and suppression of inflammation in the SN of mice with PD, which provides new insights into the pathogenesis of PD and its treatment.
Collapse
Affiliation(s)
- Qiu-Qin Han
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yi Fu
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Jia-Mei Le
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Adam Pilot
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
| | - Si Cheng
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Pei-Qing Chen
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Hailong Wu
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Guo-Qing Wan
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Xue-Feng Gu
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|
50
|
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
|