1
|
Jiang C, Zhan Q, Zeng C. The 5-HT-related gut-brain axis in obesity. Life Sci 2024; 358:123171. [PMID: 39447731 DOI: 10.1016/j.lfs.2024.123171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/22/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
AIMS The incidence of obesity increases annually. It is closely related to the occurrence of cardiovascular diseases, malignant tumors, etc., and has become a major global health problem. 5-hydroxytryptamine (5-HT), a multifunctional monoamine neurotransmitter, is dispersed throughout the central nervous system and digestive tract. It is intimately related to the mechanism of obesity. MATERIALS AND METHODS PubMed, Web of Science and Embase were carefully searched. We collected articles that are closely related to 5-HT, the gut-brain axis, and obesity. KEY FINGDINGS The gut microbiota not only influences nutrient metabolism but also centrally meditates appetite and mood regulation. The gut-brain axis, a system connecting the gut and the brain, is known to participate in two-way communication between the gut flora and the central nervous system. SIGNIFICANCE There have been few reports on whether peripheral and central 5-HT interact bidirectionally via the gut-brain axis and jointly play a role in the pathogenesis of obesity. In this review, we summarize the rationale for the contribution of the 5-HT-related gut-brain axis to the development of obesity and explore feasible signaling pathways, which elucidates new targets for preventing and treating obesity.
Collapse
Affiliation(s)
- Chaoyong Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qiong Zhan
- Department of Neurology, the Second Xiangya Hospital, Central South University, Changsha 410011, China; Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, the Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Chang Zeng
- Health Management Center, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
2
|
Yuan Y, Zhuang Y, Cui Y, Liu Y, Zhang Q, Xiao Q, Meng Q, Jiang J, Hao W, Wei X. Effects of 1, 2-bis (2,4, 6-tribromophenoxy) ethane and bis (2-ethylhexyl) tetrabromophthalate on serum metabolic and lipid profiles in male rats. Toxicol Appl Pharmacol 2024; 490:117020. [PMID: 38969211 DOI: 10.1016/j.taap.2024.117020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
This study explored the effects of 1, 2-bis (2,4, 6-tribromophenoxy) ethane (BTBPE) and bis (2-ethylhexyl) tetrabromophthalate (TBPH) on serum metabolites and lipids in male Sprague-Dawley (SD) rats. Rats were orally gavaged 250 mg/kg bw of BTBPE and 500 mg/kg bw of TBPH for 28 consecutive days. Serum samples were collected for metabolomics and lipidomics analysis. Orthogonal partial least squares discriminant analysis (OPLS-DA) was used to explore changes in rat metabolic patterns. Least absolute shrinkage and selection operator (LASSO) regression models were established using serum levels of total thyroxine (TT4), free thyroxine (FT4), and rats' grouping information as variables to screen for robust differential substances. SuperPred was the database to obtain potential targets. The metabolomics and lipidomics results showed that BTBPE and TBPH had an impact on rat metabolic patterns, affecting pathways such as vitamin B6 synthesis. For BTBPE treatment, pyridoxal and ceramide (Cer) 24:0;4O were selected as differential substances related to thyroid hormones. For TBPH treatment, dehydroascorbic acid, acylcarnitine (CAR) 19:0, and diglyceride (DG) 38:4 were selected as differential substances related to thyroid hormones. Serotonin 2c receptor and cyclooxygenase-2 were chosen as potential targets of BTBPE and TBPH, respectively. In conclusion, this study found that BTBPE and TBPH impacted the metabolism of rats, and this effect may be related to changes in thyroid function.
Collapse
Affiliation(s)
- Yuese Yuan
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Yimeng Zhuang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Yuan Cui
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Yuetong Liu
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Qiong Zhang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Qianqian Xiao
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Qinghe Meng
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Jianjun Jiang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Weidong Hao
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Xuetao Wei
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China.
| |
Collapse
|
3
|
Osawa Y, Kawai H, Nakashima K, Nakaseko Y, Suto D, Yanagida K, Hashidate-Yoshida T, Mori T, Yoshio S, Ohtake T, Shindou H, Kanto T. Sphingosine-1-phosphate promotes liver fibrosis in metabolic dysfunction-associated steatohepatitis. PLoS One 2024; 19:e0303296. [PMID: 38753743 PMCID: PMC11098361 DOI: 10.1371/journal.pone.0303296] [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: 12/21/2023] [Accepted: 04/22/2024] [Indexed: 05/18/2024] Open
Abstract
AIM Metabolic dysfunction-associated steatohepatitis (MASH) is one of the most prevalent liver diseases and is characterized by steatosis and the accumulation of bioactive lipids. This study aims to understand the specific lipid species responsible for the progression of liver fibrosis in MASH. METHODS Changes in bioactive lipid levels were examined in the livers of MASH mice fed a choline-deficient diet (CDD). Additionally, sphingosine kinase (SphK)1 mRNA, which generates sphingosine 1 phosphate (S1P), was examined in the livers of patients with MASH. RESULTS CDD induced MASH and liver fibrosis were accompanied by elevated levels of S1P and increased expression of SphK1 in capillarized liver sinusoidal endothelial cells (LSECs) in mice. SphK1 mRNA also increased in the livers of patients with MASH. Treatment of primary cultured mouse hepatic stellate cells (HSCs) with S1P stimulated their activation, which was mitigated by the S1P receptor (S1PR)2 inhibitor, JTE013. The inhibition of S1PR2 or its knockout in mice suppressed liver fibrosis without reducing steatosis or hepatocellular damage. CONCLUSION S1P level is increased in MASH livers and contributes to liver fibrosis via S1PR2.
Collapse
Affiliation(s)
- Yosuke Osawa
- Departments of Gastroenterology, International University of Health and Welfare Hospital, Tochigi, Japan
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Hironari Kawai
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Keigo Nakashima
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
- Departments of Surgery, International University of Health and Welfare Hospital, Tochigi, Japan
| | - Yuichi Nakaseko
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
- Departments of Surgery, International University of Health and Welfare Hospital, Tochigi, Japan
| | - Daisuke Suto
- Departments of Gastroenterology, International University of Health and Welfare Hospital, Tochigi, Japan
| | - Keisuke Yanagida
- Departments of Lipid Life Science, National Center for Global Health and Medicine, Tokyo, Japan
| | | | - Taizo Mori
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Sachiyo Yoshio
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Takaaki Ohtake
- Departments of Gastroenterology, International University of Health and Welfare Hospital, Tochigi, Japan
| | - Hideo Shindou
- Departments of Lipid Life Science, National Center for Global Health and Medicine, Tokyo, Japan
- Departments of Medical Lipid Science, Graduated Scholl of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tatsuya Kanto
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| |
Collapse
|
4
|
Zhang S, Chau HT, Tun HM, Huang FY, Wong DKH, Mak LY, Yuen MF, Seto WK. Virological response to nucleos(t)ide analogues treatment in chronic hepatitis B patients is associated with Bacteroides-dominant gut microbiome. EBioMedicine 2024; 103:105101. [PMID: 38583259 PMCID: PMC11002572 DOI: 10.1016/j.ebiom.2024.105101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND Gut dysbiosis is present in chronic hepatitis B virus (HBV) infection. In this study, we integrated microbiome and metabolome analysis to investigate the role of gut microbiome in virological response to nucleos(t)ide analogues (NAs) treatment. METHODS Chronic HBV patients were prospectively recruited for steatosis and fibrosis assessments via liver elastography, with full-length 16S sequencing performed to identify the compositional gut microbiota differences. Fasting plasma bile acids were quantified by liquid chromatography-tandem mass spectrometry. FINDINGS All patients (n = 110) were characterized into three distinct microbial clusters by their dominant genus: c-Bacteroides, c-Blautia, and c-Prevotella. Patients with c-Bacteroides had a higher plasma ursodeoxycholic acids (UDCA) level and an increase in 7-alpha-hydroxysteroid dehydrogenase (secondary bile acid biotransformation) than other clusters. In NAs-treated patients (n = 84), c-Bacteroides was associated with higher odds of plasma HBV-DNA undetectability when compared with non-c-Bacteroides clusters (OR 3.49, 95% CI 1.43-8.96, p = 0.01). c-Blautia was positively associated with advanced fibrosis (OR 2.74, 95% CI 1.09-7.31, p = 0.04). No such associations were found in treatment-naïve patients. Increased Escherichia coli relative abundance (0.21% vs. 0.03%, p = 0.035) was found in on-treatment patients (median treatment duration 98.1 months) with advanced fibrosis despite HBV DNA undetectability. An enrichment in l-tryptophan biosynthesis was observed in patients with advanced fibrosis, which exhibited a positive correlation with Escherichia coli. INTERPRETATION Collectively, unique bacterial signatures, including c-Bacteroides and c-Blautia, were associated with virological undetectability and fibrosis evolution during NAs therapy in chronic HBV, setting up intriguing possibilities in optimizing HBV treatment. FUNDING This study was supported by the Guangdong Natural Science Fund (2019A1515012003).
Collapse
Affiliation(s)
- Saisai Zhang
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Hau-Tak Chau
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Hein Min Tun
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; System Microbiology and Antimicrobial Resistance (SMART) Lab, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Fung-Yu Huang
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Danny Ka-Ho Wong
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Lung-Yi Mak
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Man-Fung Yuen
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China.
| | - Wai-Kay Seto
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
5
|
Min BH, Devi S, Kwon GH, Gupta H, Jeong JJ, Sharma SP, Won SM, Oh KK, Yoon SJ, Park HJ, Eom JA, Jeong MK, Hyun JY, Stalin N, Park TS, Choi J, Lee DY, Han SH, Kim DJ, Suk KT. Gut microbiota-derived indole compounds attenuate metabolic dysfunction-associated steatotic liver disease by improving fat metabolism and inflammation. Gut Microbes 2024; 16:2307568. [PMID: 38299316 PMCID: PMC10841017 DOI: 10.1080/19490976.2024.2307568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 01/16/2024] [Indexed: 02/02/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease, and its prevalence has increased worldwide in recent years. Additionally, there is a close relationship between MASLD and gut microbiota-derived metabolites. However, the mechanisms of MASLD and its metabolites are still unclear. We demonstrated decreased indole-3-propionic acid (IPA) and indole-3-acetic acid (IAA) in the feces of patients with hepatic steatosis compared to healthy controls. Here, IPA and IAA administration ameliorated hepatic steatosis and inflammation in an animal model of WD-induced MASLD by suppressing the NF-κB signaling pathway through a reduction in endotoxin levels and inactivation of macrophages. Bifidobacterium bifidum metabolizes tryptophan to produce IAA, and B. bifidum effectively prevents hepatic steatosis and inflammation through the production of IAA. Our study demonstrates that IPA and IAA derived from the gut microbiota have novel preventive or therapeutic potential for MASLD treatment.
Collapse
Affiliation(s)
- Byeong Hyun Min
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
| | - Shivani Devi
- Department of Life Science, Gachon University, Sungnam, Republic of Korea
| | - Goo Hyun Kwon
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
| | - Haripriya Gupta
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
| | - Jin-Ju Jeong
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
| | - Satya Priya Sharma
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
| | - Sung-Min Won
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
| | - Ki-Kwang Oh
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
| | - Sang Jun Yoon
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
| | - Hee Jin Park
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
| | - Jung A Eom
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
| | - Min Kyo Jeong
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
| | - Ji Ye Hyun
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
| | - Nattan Stalin
- Department of Life Science, Gachon University, Sungnam, Republic of Korea
| | - Tae-Sik Park
- Department of Life Science, Gachon University, Sungnam, Republic of Korea
| | - Jieun Choi
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute of Agricultural and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Do Yup Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute of Agricultural and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sang Hak Han
- Department of Pathology, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Dong Joon Kim
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
| | - Ki Tae Suk
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
| |
Collapse
|
6
|
Yanko R, Levashov M, Chaka OG, Nosar V, Khasabov SG, Khasabova I. Tryptophan Prevents the Development of Non-Alcoholic Fatty Liver Disease. Diabetes Metab Syndr Obes 2023; 16:4195-4204. [PMID: 38152280 PMCID: PMC10752026 DOI: 10.2147/dmso.s444278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/18/2023] [Indexed: 12/29/2023] Open
Abstract
Purpose The main aim of this research is to study the protective effects of tryptophan on the histomorphological and biochemical abnormalities in the liver caused by a high-calorie diet (HCD), as well as its ability to normalize mitochondrial functions in order to prevent the development of non-alcoholic fatty liver disease (NAFLD). Methods The study was conducted in male Wistar rats aged 3 months at the start of the experiment. Control animals (group I) were fed a standard diet. Group II experimental animals were fed a diet with an excess of fat (45%) and carbohydrates (31%) for 12 weeks. Group III experimental animals also received L-tryptophan at a dose of 80 mg/kg body weight in addition to the HCD. The presence of NAFLD, functional activity, physiological regeneration, and the state of the liver parenchyma and connective tissue were assessed using physiological, morphological, histo-morphometric, biochemical, and biophysical research methods. Results HCD induced the development of NAFLD, which is characterized by an increase in liver weight, hypertrophy of hepatocytes and an increase in the concentration of lipids, cholesterol and triglycerides in liver tissue. Increased alanine aminotransferase activity in the liver of obese rats also confirm hepatocytes damage. Tryptophan added to the diet lowered the severity of NAFLD by reducing fat accumulation and violations of bioelectric properties, and prevented a decrease in mitochondrial ATP synthesis. Conclusion The addition of tryptophan can have a potential positive effect on the liver, reducing the severity of structural, biochemical, mitochondrial and bioelectric damage caused by HCD.
Collapse
Affiliation(s)
- Roman Yanko
- Department of Clinical Physiology of Connective Tissue, Bogomoletz Institute of Physiology National Academy of Sciences of Ukraine, Kiev, Ukraine
| | - Mikhail Levashov
- Department of Clinical Physiology of Connective Tissue, Bogomoletz Institute of Physiology National Academy of Sciences of Ukraine, Kiev, Ukraine
| | - Olena Georgievna Chaka
- Department of Clinical Physiology of Connective Tissue, Bogomoletz Institute of Physiology National Academy of Sciences of Ukraine, Kiev, Ukraine
| | - Valentina Nosar
- Department of Clinical Physiology of Connective Tissue, Bogomoletz Institute of Physiology National Academy of Sciences of Ukraine, Kiev, Ukraine
| | - Sergey G Khasabov
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Iryna Khasabova
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
7
|
Iannone V, Babu AF, Lok J, Gómez-Gallego C, D'Auria G, Vazquez-Uribe R, Vaaben TH, Bongers M, Mikkonen S, Vaittinen M, Tikkanen I, Kettunen M, Klåvus A, Sehgal R, Kaminska D, Pihlajamaki J, Hanhineva K, El-Nezami H, Sommer MOA, Kolehmainen M. Changes in liver metabolic pathways demonstrate efficacy of the combined dietary and microbial therapeutic intervention in MASLD mouse model. Mol Metab 2023; 78:101823. [PMID: 37839774 PMCID: PMC10618820 DOI: 10.1016/j.molmet.2023.101823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023] Open
Abstract
OBJECTIVE Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), is the most prevalent liver disease globally, yet no therapies are approved. The effects of Escherichia coli Nissle 1917 expressing aldafermin, an engineered analog of the intestinal hormone FGF19, in combination with dietary change were investigated as a potential treatment for MASLD. METHODS MASLD was induced in C57BL/6J male mice by American lifestyle-induced obesity syndrome diet and then switched to a standard chow diet for seven weeks. In addition to the dietary change, the intervention group received genetically engineered E. coli Nissle expressing aldafermin, while control groups received either E. coli Nissle vehicle or no treatment. MASLD-related plasma biomarkers were measured using an automated clinical chemistry analyzer. The liver steatosis was assessed by histology and bioimaging analysis using Fiji (ImageJ) software. The effects of the intervention in the liver were also evaluated by RNA sequencing and liquid-chromatography-based non-targeted metabolomics analysis. Pathway enrichment studies were conducted by integrating the differentially expressed genes from the transcriptomics findings with the metabolites from the metabolomics results using Ingenuity pathway analysis. RESULTS After the intervention, E. coli Nissle expressing aldafermin along with dietary changes reduced body weight, liver steatosis, plasma aspartate aminotransferase, and plasma cholesterol levels compared to the two control groups. The integration of transcriptomics with non-targeted metabolomics analysis revealed the downregulation of amino acid metabolism and related receptor signaling pathways potentially implicated in the reduction of hepatic steatosis and insulin resistance. Moreover, the downregulation of pathways linked to lipid metabolism and changes in amino acid-related pathways suggested an overall reduction of oxidative stress in the liver. CONCLUSIONS These data support the potential for using engineered microbial therapeutics in combination with dietary changes for managing MASLD.
Collapse
Affiliation(s)
- Valeria Iannone
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland
| | - Ambrin Farizah Babu
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland; Afekta Technologies Ltd., Microkatu 1, 70210 Kuopio, Finland
| | - Johnson Lok
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland
| | - Carlos Gómez-Gallego
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland.
| | - Giuseppe D'Auria
- Sequencing and Bioinformatics Service, Foundation for the Promotion of Health and Biomedical Research of Valencia Region, FISABIO, 46020 Valencia, Spain; CIBER in Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Ruben Vazquez-Uribe
- Technical University of Denmark, The Novo Nordisk Foundation Center for Biosustainability, 2800 Kongens Lyngby, Denmark
| | - Troels Holger Vaaben
- Technical University of Denmark, The Novo Nordisk Foundation Center for Biosustainability, 2800 Kongens Lyngby, Denmark
| | - Mareike Bongers
- Technical University of Denmark, The Novo Nordisk Foundation Center for Biosustainability, 2800 Kongens Lyngby, Denmark
| | - Santtu Mikkonen
- University Department of Technical Physics, University of Eastern Finland, 70211 Kuopio, Finland
| | - Maija Vaittinen
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland
| | - Ida Tikkanen
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland
| | - Mikko Kettunen
- Biomedical Imaging Unit, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Anton Klåvus
- Afekta Technologies Ltd., Microkatu 1, 70210 Kuopio, Finland
| | - Ratika Sehgal
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland
| | - Dorota Kaminska
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland; Department of Medicine, Division of Cardiology, University of California, Los Angeles, CA 90095, USA
| | - Jussi Pihlajamaki
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland; Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Kati Hanhineva
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland; Afekta Technologies Ltd., Microkatu 1, 70210 Kuopio, Finland; Department of Life Technologies, Food Sciences Unit, University of Turku, 20014 Turku, Finland
| | - Hani El-Nezami
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland; University of Hong Kong, Hong Kong SAR, Molecular and Cell Biology Research Area, School of Biological Sciences, Hong Kong, Hong Kong, China
| | - Morten Otto Alexander Sommer
- Technical University of Denmark, The Novo Nordisk Foundation Center for Biosustainability, 2800 Kongens Lyngby, Denmark.
| | - Marjukka Kolehmainen
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200 Kuopio, Finland
| |
Collapse
|
8
|
Pascut D, Giraudi PJ, Banfi C, Ghilardi S, Tiribelli C, Bondesan A, Caroli D, Minocci A, Grugni G, Sartorio A. Proteome profiling identifies circulating biomarkers associated with hepatic steatosis in subjects with Prader-Willi syndrome. Front Endocrinol (Lausanne) 2023; 14:1254778. [PMID: 38034016 PMCID: PMC10684934 DOI: 10.3389/fendo.2023.1254778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/13/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction Prader-Willi syndrome (PWS) is a rare genetic disorder characterized by loss of expression of paternal chromosome 15q11.2-q13 genes. Individuals with PWS exhibit unique physical, endocrine, and metabolic traits associated with severe obesity. Identifying liver steatosis in PWS is challenging, despite its lower prevalence compared to non-syndromic obesity. Reliable biomarkers are crucial for the early detection and management of this condition associated with the complex metabolic profile and cardiovascular risks in PWS. Methods Circulating proteome profiling was conducted in 29 individuals with PWS (15 with steatosis, 14 without) using the Olink Target 96 metabolism and cardiometabolic panels. Correlation analysis was performed to identify the association between protein biomarkes and clinical variables, while the gene enrichment analysis was conducted to identify pathways linked to deregulated proteins. Receiver operating characteristic (ROC) curves assessed the discriminatory power of circulating protein while a logistic regression model evaluated the potential of a combination of protein biomarkers. Results CDH2, CTSO, QDPR, CANT1, ALDH1A1, TYMP, ADGRE, KYAT1, MCFD, SEMA3F, THOP1, TXND5, SSC4D, FBP1, and CES1 exhibited a significant differential expression in liver steatosis, with a progressive increase from grade 1 to grade 3. FBP1, CES1, and QDPR showed predominant liver expression. The logistic regression model, -34.19 + 0.85 * QDPR*QDPR + 0.75 * CANT1*TYMP - 0.46 * THOP1*ALDH1A, achieved an AUC of 0.93 (95% CI: 0.63-0.99), with a sensitivity of 93% and specificity of 80% for detecting steatosis in individuals with PWS. These biomarkers showed strong correlations among themselves and were involved in an interconnected network of 62 nodes, related to seven metabolic pathways. They were also significantly associated with cholesterol, LDL, triglycerides, transaminases, HbA1c, FLI, APRI, and HOMA, and showed a negative correlation with HDL levels. Conclusion The biomarkers identified in this study offer the potential for improved patient stratification and personalized therapeutic protocols.
Collapse
Affiliation(s)
- Devis Pascut
- Liver Cancer Unit, Fondazione Italiana Fegato - ONLUS, Trieste, Italy
| | - Pablo J. Giraudi
- Metabolic Liver Disease Unit, Fondazione Italiana Fegato - ONLUS, Trieste, Italy
| | - Cristina Banfi
- Unit of Functional Proteomics, Metabolomics, and Network analysis, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Stefania Ghilardi
- Unit of Functional Proteomics, Metabolomics, and Network analysis, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Claudio Tiribelli
- Liver Cancer Unit, Fondazione Italiana Fegato - ONLUS, Trieste, Italy
- Metabolic Liver Disease Unit, Fondazione Italiana Fegato - ONLUS, Trieste, Italy
| | - Adele Bondesan
- Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Experimental Laboratory for Auxo-endocrinological Research, Piancavallo-Verbania, Italy
| | - Diana Caroli
- Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Experimental Laboratory for Auxo-endocrinological Research, Piancavallo-Verbania, Italy
| | - Alessandro Minocci
- Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Division of Metabolic Diseases, Piancavallo-Verbania, Italy
| | - Graziano Grugni
- Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Experimental Laboratory for Auxo-endocrinological Research, Piancavallo-Verbania, Italy
- Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Division of Auxology, Piancavallo-Verbania, Italy
| | - Alessandro Sartorio
- Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Experimental Laboratory for Auxo-endocrinological Research, Piancavallo-Verbania, Italy
| |
Collapse
|
9
|
Rosa LF, Haasis E, Knauss A, Guseva D, Bischoff SC. Serotonin reuptake transporter deficiency promotes liver steatosis and impairs intestinal barrier function in obese mice fed a Western-style diet. Neurogastroenterol Motil 2023; 35:e14611. [PMID: 37246491 DOI: 10.1111/nmo.14611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/23/2023] [Accepted: 05/01/2023] [Indexed: 05/30/2023]
Abstract
BACKGROUND Intestinal barrier dysfunctions have been associated with liver steatosis and metabolic diseases. Besides nutritional factors, like a Western-style diet (WSD), serotonin has been linked with leaky gut. Therefore, we aimed to evaluate the role of serotonin in the pathogenesis of intestinal barrier dysfunctions and liver steatosis in mice fed high-fat and high-sugar diets. METHODS 6-8 weeks old male serotonin reuptake transporter knockout mice (SERT-/- ) and wild-type controls (SERT+/+ ) were fed either a WSD or a control diet (CD) ad libitum with or without fructose 30% (F) added to the drinking water for 12 weeks. Markers of liver steatosis and intestinal barrier function were assessed. KEY RESULTS SERT-/- mice showed increased weight gain compared with SERT+/+ mice when fed a WSD ± F for 12 weeks (p < 0.05), whereby SERT-/- mice exhibited reduced energy (-21%) intake. Furthermore, SERT knockout resulted in a more pronounced liver steatosis (p < 0.05), enhanced levels of endotoxin in portal vein plasma (p < 0.05), and increased liver expression of Tnf and Myd88 (p < 0.05), when mice were fed a WSD ± F. Finally, SERT-/- mice, when compared with SERT+/+ mice, had a decreased mRNA expression of Muc2 (p < 0.01), Ocln (p < 0.05), Cldn5 (p = 0.054) and 7 (p < 0.01), Defa5 (p < 0.05) and other antimicrobial peptides in the ileum. On the protein level, ZO-1 (p < 0.01) and DEFA5 protein (p < 0.0001) were decreased. CONCLUSION AND INFERENCES Our data demonstrate that SERT knockout causes weight gain, liver steatosis, and leaky gut, especially in mice fed a WSD. Therefore, SERT induction could be a novel therapeutic approach to improve metabolic diseases associated with intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Louisa Filipe Rosa
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Eva Haasis
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Annkathrin Knauss
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Daria Guseva
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
10
|
Qiu F, Zeng R, Li D, Ye T, Xu W, Wang X, Yan X, Li H, Hu X. Establishment and bioinformatics evaluation of the ethanol combined with palmitic acid-induced mouse hepatocyte AFLD model (the Hu-Qiu Model). Heliyon 2023; 9:e19359. [PMID: 37681138 PMCID: PMC10481297 DOI: 10.1016/j.heliyon.2023.e19359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 08/15/2023] [Accepted: 08/20/2023] [Indexed: 09/09/2023] Open
Abstract
Chronic alcoholic liver disease has brought great harm to human health. Alcoholic fatty liver disease is the first stage in the progression of all chronic alcoholic liver diseases. At present, there is no cell model that fully matches the etiology (high-fat diet + alcohol) of human alcoholic fatty liver disease. We used 100 mM ethanol +6.25 μM PA to establish the ethanol combined with PA-induced mouse hepatocyte AFLD model (EP-AFLD hepatocyte model) and performed the RNA-seq transcriptome sequencing. Through bioinformatics analysis and comparison, we discovered that the EP-AFLD hepatocyte model was more suitable for studying the pathological mechanism of AFLD than the mouse AFLD hepatocyte model induced by ethanol alone. And through bioinformatics analysis, we further discovered that 77 genes from the differential expression gene set of EP-AFLD hepatocyte model were engaged in the pathological process of mouse AFLD and 40 genes were involved in the pathogenesis of both mouse AFLD and human AFLD. In this study, a novel mouse hepatocyte AFLD model was successfully established by combining ethanol and PA, which can be used to study the molecular mechanism of the pathogenesis of AFLD in mice or humans. This study will provide a brand-new in vitro experimental platform for the in-depth study of AFLD pathogenesis and the screening of AFLD therapeutic drugs.
Collapse
Affiliation(s)
| | | | - Du Li
- Department of Biology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tingjie Ye
- Department of Biology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wei Xu
- Department of Biology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaoling Wang
- Department of Biology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaofeng Yan
- Department of Biology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hua Li
- Department of Biology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xudong Hu
- Department of Biology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
11
|
Meyer RK, Duca FA. RISING STARS: Endocrine regulation of metabolic homeostasis via the intestine and gut microbiome. J Endocrinol 2023; 258:e230019. [PMID: 37171833 PMCID: PMC10524498 DOI: 10.1530/joe-23-0019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 05/12/2023] [Indexed: 05/13/2023]
Abstract
The gastrointestinal system is now considered the largest endocrine organ, highlighting the importance of gut-derived peptides and metabolites in metabolic homeostasis. Gut peptides are secreted from intestinal enteroendocrine cells in response to nutrients, microbial metabolites, and neural and hormonal factors, and they regulate systemic metabolism via multiple mechanisms. While extensive research is focused on the neuroendocrine effects of gut peptides, evidence suggests that several of these hormones act as endocrine signaling molecules with direct effects on the target organ, especially in a therapeutic setting. Additionally, the gut microbiota metabolizes ingested nutrients and fiber to produce compounds that impact host metabolism indirectly, through gut peptide secretion, and directly, acting as endocrine factors. This review will provide an overview of the role of endogenous gut peptides in metabolic homeostasis and disease, as well as the potential endocrine impact of microbial metabolites on host metabolic tissue function.
Collapse
Affiliation(s)
- Rachel K Meyer
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, Arizona, USA
| | - Frank A Duca
- School of Animal and Comparative Biomedical Sciences, College of Agricultural and Life Sciences, University of Arizona, Tucson, Arizona, USA
- BIO5 Institute, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
12
|
The development of the Metabolic-associated Fatty Liver Disease during pharmacotherapy of mental disorders - a review. CURRENT PROBLEMS OF PSYCHIATRY 2022. [DOI: 10.2478/cpp-2022-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Introduction: Metabolic-associated Fatty Liver Disease (MAFLD) is a term for Non-alcoholic Fatty Liver Disease (NAFLD) that highlights its association with components of the Metabolic Syndrome (MetS). MAFLD is becoming a clinically significant problem due to its increasing role in the pathogenesis of cryptogenic cirrhosis of the liver.
Material and methods: The resulting work is a review of the most important information on the risk of MAFLD development in the context of the use of particular groups of psychotropic drugs. The study presents the epidemiology, with particular emphasis on the population of psychiatric patients, pathophysiology and scientific reports analyzing the effect of the psychotropic medications on MAFLD development.
Results: The drugs that can have the greatest impact on the development of MAFLD are atypical antipsychotics, especially olanzapine, and mood stabilizers (MS) - valproic acid (VPA). Their effect is indirect, mainly through dysregulation of organism’s carbohydrate and lipid metabolism.
Conclusions: The population of psychiatric patients is particularly vulnerable to the development of MAFLD. At the root of this disorder lies the specificity of mental disorders, improper dietary habits, low level of physical activity and tendency to addictions. Also, the negative impact of the psychotropic drugs on the systemic metabolism indirectly contributes to the development of MAFLD. In order to prevent fatty liver disease, it is necessary to monitor metabolic and liver parameters regularly, and patients should be screened by ultrasound examination of the liver. There are also important preventive actions from the medical professionals, including education of patients and sensitizing to healthy lifestyle.
Collapse
|
13
|
Obeng KA, Mochizuki S, Koike S, Toyoshima Y, Sato Y, Yoshizawa F. Analysis of the Stimulative Effect of Tryptophan on Hepatic Protein Synthesis in Rats. J Nutr Sci Vitaminol (Tokyo) 2022; 68:312-319. [PMID: 36047103 DOI: 10.3177/jnsv.68.312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Tryptophan is an essential amino acid important as a protein building block, but it also serves as substrate for the generation of several bioactive compounds with important physiological roles. Furthermore, tryptophan has been reported to have a unique role as a nutritional signaling molecule that regulates protein synthesis in mouse and rat liver. In the present study, the acute effects of tryptophan on protein synthesis were confirmed and compared with those of leucine in rats. Eighteen hours fasted rats were orally administered of tryptophan or leucine at a dose of 135 mg/100 g body weight by gavage and then sacrificed 1 h after administration. The effects of tryptophan and leucine on the rate of protein synthesis were evaluated by the surface sensing of translation (SUnSET) method. We also examined the ability of tryptophan to induce activation of the mTOR pathway by measuring phosphorylation of 4E-BP1 and S6K1. Oral administration of tryptophan led to a stimulation of the rate of protein synthesis concomitant with activation of mTOR pathway in the liver, but not in skeletal muscle. We also investigated the sensitivity of liver protein synthesis to tryptophan administration. The half-maximal effective doses (ED50) of tryptophan in stimulating 4E-BP1 and S6K1 phosphorylation were both about 60% of daily intake. The effect of tryptophan on hepatic protein synthesis was similar to that of leucine on muscle protein synthesis, and the sensitivity of liver protein synthesis to tryptophan administration appeared to be almost the same or slightly lower than that of muscle protein synthesis to leucine administration.
Collapse
Affiliation(s)
- Kodwo Amuzuah Obeng
- Department of Biological Production Science, The United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology
| | - Shinji Mochizuki
- Department of Agrobiology and Bioresources, School of Agriculture, Utsunomiya University
| | - Shinichiro Koike
- Department of Biological Production Science, The United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology
| | - Yuka Toyoshima
- Department of Agrobiology and Bioresources, School of Agriculture, Utsunomiya University
| | - Yusuke Sato
- Department of Biological Production Science, The United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology.,Department of Agrobiology and Bioresources, School of Agriculture, Utsunomiya University
| | - Fumiaki Yoshizawa
- Department of Biological Production Science, The United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology.,Department of Agrobiology and Bioresources, School of Agriculture, Utsunomiya University
| |
Collapse
|
14
|
Carbidopa, an activator of aryl hydrocarbon receptor, suppresses IDO1 expression in pancreatic cancer and decreases tumor growth. Biochem J 2022; 479:1807-1824. [PMID: 35997090 PMCID: PMC9472820 DOI: 10.1042/bcj20210851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 08/08/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022]
Abstract
IDO1 is an immunomodulatory enzyme responsible for tryptophan catabolism. Its expression in immune cells, especially the DCs, has attracted attention because it leads to tryptophan depletion at the immunological synapse, thereby causing T-cell anergy and immune evasion by the tumor cells. Cancer cells also overexpress IDO1. Immunotherapy targeting IDO1 has been one of the focus areas in cancer biology, but lately studies have identified non-immune related functions of IDO1 leading to a paradigm shift with regard to IDO1 function in the context of tumor cells. In this study, we show that PDAC tissues and PDAC cells overexpress IDO1. The expression level is reciprocally related to overall patient survival. We further show that carbidopa, an FDA-approved drug for Parkinson's disease as well as an AhR agonist, inhibits IDO1 expression in PDAC cells. Using athymic nude mice, we demonstrate that carbidopa-mediated suppression of IDO1 expression attenuates tumor growth. Mechanistically, we show that AhR is responsible for carbidopa-mediated suppression of IDO1, directly as a transcription factor and indirectly by interfering with the JAK/STAT pathway. Overall, targeting IDO1 not only in immune cells but also in cancer cells could be a beneficial therapeutic strategy for PDAC and potentially for other cancers as well and that carbidopa could be repurposed to treat cancers that overexpress IDO1.
Collapse
|
15
|
Barra NG, Kwon YH, Morrison KM, Steinberg GR, Wade MG, Khan WI, Vijayan MM, Schertzer JD, Holloway AC. Increased gut serotonin production in response to bisphenol A structural analogs may contribute to their obesogenic effects. Am J Physiol Endocrinol Metab 2022; 323:E80-E091. [PMID: 35575233 DOI: 10.1152/ajpendo.00049.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Obesogens are synthetic, environmental chemicals that can disrupt endocrine control of metabolism and contribute to the risk of obesity and metabolic disease. Bisphenol A (BPA) is one of the most studied obesogens. There is considerable evidence that BPA exposure is associated with weight gain, increased adiposity, poor blood glucose control, and nonalcoholic fatty liver disease in animal models and human populations. Increased usage of structural analogs of BPA has occurred in response to legislation banning their use in some commercial products. However, BPA analogs may also cause some of the same metabolic impairments because of common mechanisms of action. One key effector that is altered by BPA and its analogs is serotonin, however, it is unknown if BPA-induced changes in peripheral serotonin pathways underlie metabolic perturbations seen with BPA exposure. Upon ingestion, BPA and its analogs act as endocrine-disrupting chemicals in the gastrointestinal tract to influence serotonin production by the gut, where over 95% of serotonin is produced. The purpose of this review is to evaluate how BPA and its analogs alter gut serotonin regulation and then discuss how disruption of serotonergic networks influences host metabolism. We also provide evidence that BPA and its analogs enhance serotonin production in gut enterochromaffin cells. Taken together, we propose that BPA and many BPA analogs represent endocrine-disrupting chemicals that can influence host metabolism through the endogenous production of gut-derived factors, such as serotonin.
Collapse
Affiliation(s)
- Nicole G Barra
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Yun Han Kwon
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Katherine M Morrison
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Gregory R Steinberg
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Michael G Wade
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Waliul I Khan
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Alison C Holloway
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
16
|
Banskota S, Khan WI. Gut-derived serotonin and its emerging roles in immune function, inflammation, metabolism and the gut-brain axis. Curr Opin Endocrinol Diabetes Obes 2022; 29:177-182. [PMID: 35197425 DOI: 10.1097/med.0000000000000713] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW To shed light on the recently uncovered diverse role of serotonin (5-hydroxytryptamine; 5-HT) in the regulation of immune functions, inflammation, metabolism, and gut-brain axis. RECENT FINDINGS Peripheral 5-HT which accounts for approximately 95% of the total is largely synthesized in the gut by enterochromaffin cells. Enterochromaffin cells release 5-HT in response to various stimuli including microbial products. Released 5-HT influences secretomotor, sensory and immune functions as well as inflammatory processes in the gut. 5-HT released from enterochromaffin cells enters circulation and is taken up and concentrated in platelets. 5-HT released from the activated platelets interacts with different organs to alter their metabolic activity. 5-HT also serves as a link in the gut-brain axis. SUMMARY Emerging evidence regarding the role of peripheral 5-HT in the regulation of various physiological and pathophysiological conditions opens up new targets for researchers to explore and for clinicians to treat and manage different diseases associated with the altered 5-HT signalling.
Collapse
Affiliation(s)
- Suhrid Banskota
- Department of Pathology and Molecular Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | | |
Collapse
|
17
|
Li XY, Tan ZJ. Modern biological connotation of diarrhea with kidney-Yang deficiency syndrome. Shijie Huaren Xiaohua Zazhi 2022; 30:119-127. [DOI: 10.11569/wcjd.v30.i3.119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Kidney-Yang deficiency syndrome, a common traditional Chinese medicine syndrome of diarrhea, has a complex pathogenesis. This paper explores the mechanisms of the development of diarrhea with kidney-Yang deficiency syndrome from three aspects: Gut flora, signaling pathway, and molecules related to the "kidney-gut axis", and tries to identify biomarkers for diarrhea with kidney-Yang deficiency syndrome. It is of great significance to reveal the modern biological connotation of diarrhea with kidney-Yang deficiency syndrome, which can promote the subsequent clinical targeted therapy.
Collapse
Affiliation(s)
- Xiao-Ya Li
- Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| | - Zhou-Jin Tan
- Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| |
Collapse
|
18
|
Chen J, Vitetta L, Henson JD, Hall S. Intestinal Dysbiosis, the Tryptophan Pathway and Nonalcoholic Steatohepatitis. Int J Tryptophan Res 2022; 15:11786469211070533. [PMID: 35153490 PMCID: PMC8829707 DOI: 10.1177/11786469211070533] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/07/2021] [Indexed: 12/15/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) progresses from simple steatosis to steatohepatitis (NASH), which may then progress to the development of cirrhosis and hepatocarcinoma. NASH is characterized by both steatosis and inflammation. Control of inflammation in NASH is a key step for the prevention of disease progression to severe sequalae. Intestinal dysbiosis has been recognized to be an important causal factor in the pathogenesis of NASH, involving both the accumulation of lipids and aggravation of inflammation. The effects of gut dysbiosis are mediated by adverse shifts of various intestinal commensal bacterial genera and their associated metabolites such as butyrate, tryptophan, and bile acids. In this review, we focus on the roles of tryptophan and its metabolites in NASH in association with intestinal dysbiosis and discuss possible therapeutic implications.
Collapse
Affiliation(s)
- Jiezhong Chen
- Research Department, Medlab Clinical, Sydney, NSW, Australia
| | - Luis Vitetta
- Research Department, Medlab Clinical, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Jeremy D Henson
- Research Department, Medlab Clinical, Sydney, NSW, Australia
- Faculty of Medicine, Prince of Wales Clinical School, The University of New South Wales, Sydney, NSW, Australia
| | - Sean Hall
- Research Department, Medlab Clinical, Sydney, NSW, Australia
| |
Collapse
|
19
|
Exploring the potential mechanism of Rhodomyrtus tomentosa (Ait.) Hassk fruit phenolic rich extract on ameliorating nonalcoholic fatty liver disease by integration of transcriptomics and metabolomics profiling. Food Res Int 2022; 151:110824. [PMID: 34980375 DOI: 10.1016/j.foodres.2021.110824] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 11/23/2021] [Indexed: 12/16/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD), as the commonest form of chronic liver disease, is accompanied by liver oxidative stress and inflammatory responses. Rhodomyrtus tomentosa (Ait.) Hassk fruit phenolic rich extract (RTE) possesses multiple pharmacological effects in management of chronic diseases. In this study, the liver-protective effect of RTE on mice with high-fat-diet (HFD)-induced NAFLD was investigated for the first time, and the underlying molecular mechanism was explored via integration of transcriptomics and metabolomics. The results showed that RTE mitigated liver damage, which was evidenced by declined inflammatory cell infiltration in liver, decreased liver function markers, oxidative stress indexes, lipid profile levels and inflammatory cytokines levels. The differential metabolites by metabonomics illustrated supplementation of RTE affected metabolomics pathways including tryptophan metabolism, alanine, aspartate and glutamate metabolism, D-glutamine and D-glutamate metabolism, cysteine and methionine metabolism, arginine and proline metabolism, which are all involved in oxidative stress and inflammation. Furthermore, the five differential expression genes (DEGs) through liver transcriptomics were screened and recognized, namely Tnfrsf21, Ifit1, Inhbb, Mapk15 and Gadd45g, which revealed that HFD induced Cytokine-cytokine receptor interaction pathway, NF-κB signaling pathway NOD-like receptor pathway, TNF signaling pathway. Integrated analysis of transcriptomics and metabolomics confirmed the supplementation of RTE had significantly regulatory effects on the metabolic pathways involved in inflammatory responses. Additionally, RT-PCR and western blot authenticated RTE intervention regulated the mRNA levels of liver genes involved in inflammation response and inhibited the liver endotoxin-TLR4-NF-κB pathway triggered by HFD, thus alleviating NAFLD. Our findings strongly support the possibility that RTE can be regarded as a potential therapeutic method for obesity-associated NAFLD.
Collapse
|
20
|
Yin H, Shi A, Wu J. Platelet-Activating Factor Promotes the Development of Non-Alcoholic Fatty Liver Disease. Diabetes Metab Syndr Obes 2022; 15:2003-2030. [PMID: 35837578 PMCID: PMC9275506 DOI: 10.2147/dmso.s367483] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/28/2022] [Indexed: 11/23/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifaceted clinicopathological syndrome characterised by excessive hepatic lipid accumulation that causes steatosis, excluding alcoholic factors. Platelet-activating factor (PAF), a biologically active lipid transmitter, induces platelet activation upon binding to the PAF receptor. Recent studies have found that PAF is associated with gamma-glutamyl transferase, which is an indicator of liver disease. Moreover, PAF can stimulate hepatic lipid synthesis and cause hypertriglyceridaemia. Furthermore, the knockdown of the PAF receptor gene in the animal models of NAFLD helped reduce the inflammatory response, improve glucose homeostasis and delay the development of NAFLD. These findings suggest that PAF is associated with NAFLD development. According to reports, patients with NAFLD or animal models have marked platelet activation abnormalities, mainly manifested as enhanced platelet adhesion and aggregation and altered blood rheology. Pharmacological interventions were accompanied by remission of abnormal platelet activation and significant improvement in liver function and lipids in the animal model of NAFLD. These confirm that platelet activation may accompany a critical importance in NAFLD development and progression. However, how PAFs are involved in the NAFLD signalling pathway needs further investigation. In this paper, we review the relevant literature in recent years and discuss the role played by PAF in NAFLD development. It is important to elucidate the pathogenesis of NAFLD and to find effective interventions for treatment.
Collapse
Affiliation(s)
- Hang Yin
- Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China
| | - Anhua Shi
- Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China
| | - Junzi Wu
- Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China
- Correspondence: Junzi Wu; Anhua Shi, Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China, Tel/Fax +86 187 8855 7524; +86 138 8885 0813, Email ;
| |
Collapse
|
21
|
Yue Q, Cai M, Xiao B, Zhan Q, Zeng C. A High-Tryptophan Diet Reduces Seizure-Induced Respiratory Arrest and Alters the Gut Microbiota in DBA/1 Mice. Front Neurol 2021; 12:762323. [PMID: 34887831 PMCID: PMC8650499 DOI: 10.3389/fneur.2021.762323] [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: 08/21/2021] [Accepted: 10/26/2021] [Indexed: 12/20/2022] Open
Abstract
Background and Aims: Central 5-hydroxytryptamine (5-HT) defects are responsible for the occurrence of sudden unexpected death in epilepsy (SUDEP). The DBA/1 mouse is an animal model of SUDEP since the mouse exhibits audiogenic seizure-induced respiratory arrest (S-IRA). The synthesis of central 5-HT is closely related to the gut microbiota. Moreover, emerging studies suggest a possible role for the microbiota in mitigating seizure likelihood. Based on this, we aimed to explore the effect of a high-tryptophan diet (HTD) on SUDEP as well as the synthesis and metabolism of central 5-HT. Furthermore, we investigated the involvement of the gut microbiota in this process. Methods: All DBA/1 mice were subjected to acoustic stimulation to induce seizures. Only those mice that exhibited S-IRA were randomly assigned to the normal diet (ND) group (n = 39) or HTD group (n = 53). After 1 month of dietary intervention, (1) S-IRA rates were evaluated, (2) the concentrations of 5-HT and its metabolite 5-hydroxyindoleacetic acid (5-HIAA) in the plasma and brain were determined by ultra-high-pressure liquid chromatography, and (3) the fecal flora biodiversity and species composition were analyzed by 16S rDNA microbiota profiling. Results: The S-IRA rate in DBA/1 mice was significantly reduced in the HTD group compared with that in the control group. HTD increased the levels of 5-HT and 5-HIAA in both the telencephalon and midbrain. HTD significantly elevated the species richness and diversity of the gut microbiota. Moreover, there was a significant difference in the gut microbiota composition between the two groups, and the intestinal flora was dominated by Proteobacteria and Actinobacteria after HTD. Conclusions: HTD is efficient in lowering S-IRA rates and elevating the central 5-HT level in DBA/1 mice. The gut microbiota was altered after HTD intervention. The significant increase in Proteobacteria and Actinobacteria may be related to the SUDEP-protective effect of HTD. Our findings shed light on a candidate choice of dietary prevention for SUDEP.
Collapse
Affiliation(s)
- Qiang Yue
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Mingfei Cai
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Qiong Zhan
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chang Zeng
- Health Management Center, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
22
|
Yang Z, Li Z, Guo Z, Ren Y, Zhou T, Xiao Z, Duan J, Han C, Cheng Y, Xu F. Antitumor Effect of Fluoxetine on Chronic Stress-Promoted Lung Cancer Growth via Suppressing Kynurenine Pathway and Enhancing Cellular Immunity. Front Pharmacol 2021; 12:685898. [PMID: 34413774 PMCID: PMC8369900 DOI: 10.3389/fphar.2021.685898] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/15/2021] [Indexed: 01/22/2023] Open
Abstract
Background: Chronic stress promotes cancer growth. Antidepressant fluoxetine (FLX) is usually prescribed for cancer patients with comorbid depression. FLX displays inhibition on cancer cell proliferation, however, the in vivo activity has not been investigated. Methods: We explored the antitumor effect of FLX in subcutaneous transplanted lung cancer cells in a tumor-bearing mouse model. Fifty-six C57BL/6 mice were randomly divided into group A (blank control), group B (tumor-bearing control), group C (tumor-bearing + FLX), group D (CUMS control), group E (CUMS + FLX), group F (tumor-bearing + CUMS), and group G (tumor-bearing + CUMS + FLX). 5-HT, tryptophane (Trp), kynurenine, IFN-γ, TNF-α, IL-1α, IL-1β, IL-2, IL-4, IL-6, IL-10, IL-17A levels were measured by ELISA. T helper (Th), cytotoxic T (Tc) and regulatory T cells (Tregs) subtype were measured by flow cytometry. The antitumor effects of FLX were evaluated by tumor weight. The expression of kynurenine pathway related genes TDO, IDO1, IDO2, and apoptosis-related genes caspase1, 3, 4, 5, 7, 12 in tumor tissues were measured by western blotting and qRT-PCR. A549 cells were exposed with FLX (15 μmol/L) and its effect on cell proliferation, migration, and clonal formation were detected. Kynurenine pathway and apoptosis related gene expression were also measured. Results:In vivo, chronic stress promoted tumor growth in C57BL/6 mice. FLX administration not only significantly reversed chronic unpredictable mild stress (CUMS)-induced reduction of 5-HT and Trp, increment of kynurenine, but increased CD4+ Th and CD8+ Tc cells, and reduced CD25+ FOXP3+ Tregs. FLX promoted Th to differentiate into Th1 cells and increased IL-2 and IFN-γ, meanwhile inhibited Th differentiate into Th2 and Th17 cells and decreased the concentrations of IL-4, IL-6, IL-10, and IL-17A. Chronic stress obviously up-regulated IDO1 and IDO2 expression, down-regulated caspase 4, 7, and 12 expression, meanwhile FLX administration reversed this regulation. However, there was no significant change in TDO, caspase 1, 3, 5. Similarly, in vitro, FLX administration significantly inhibited the proliferation, migration, and clonal formation of A549 cells and induced cell apoptosis. FLX administration down-regulated the expression of IDO1, IDO2, and up-regulated caspase 4, 5, and 7. Conclusion: Fluoxetine administration could inhibit tumor growth. The inhibition might be via suppressing kynurenine pathway and enhancing cellular immunity.
Collapse
Affiliation(s)
- Zhen Yang
- Department of Central Laboratory, Fengxian Hospital, Southern Medical University, Shanghai, China
| | - Zhuman Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,Department of Pharmacy, Fengxian Hospital, Southern Medical University, Shanghai, China
| | - Zhijun Guo
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,Department of Pharmacy, Fengxian Hospital, Southern Medical University, Shanghai, China
| | - Yu Ren
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,Department of Pharmacy, Fengxian Hospital, Southern Medical University, Shanghai, China
| | - Ting Zhou
- Department of Pharmacy, Fengxian Hospital, Southern Medical University, Shanghai, China
| | - Zhijun Xiao
- Department of Pharmacy, Fengxian Hospital, Southern Medical University, Shanghai, China
| | - Jingjing Duan
- Department of Pharmacy, Fengxian Hospital, Southern Medical University, Shanghai, China
| | - Chuangchuang Han
- Department of Neurosurgery, Sixth People's Hospital South Campus, Shanghai Jiaotong University, Shanghai, China
| | - Yuanchi Cheng
- Department of Neurosurgery, Sixth People's Hospital South Campus, Shanghai Jiaotong University, Shanghai, China
| | - Feng Xu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,Department of Pharmacy, Fengxian Hospital, Southern Medical University, Shanghai, China
| |
Collapse
|
23
|
Rebelos E, Iozzo P, Guzzardi MA, Brunetto MR, Bonino F. Brain-gut-liver interactions across the spectrum of insulin resistance in metabolic fatty liver disease. World J Gastroenterol 2021; 27:4999-5018. [PMID: 34497431 PMCID: PMC8384743 DOI: 10.3748/wjg.v27.i30.4999] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/29/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
Metabolic associated fatty liver disease (MAFLD), formerly named "nonalcoholic fatty liver disease" occurs in about one-third of the general population of developed countries worldwide and behaves as a major morbidity and mortality risk factor for major causes of death, such as cardiovascular, digestive, metabolic, neoplastic and neuro-degenerative diseases. However, progression of MAFLD and its associated systemic complications occur almost invariably in patients who experience the additional burden of intrahepatic and/or systemic inflammation, which acts as disease accelerator. Our review is focused on the new knowledge about the brain-gut-liver axis in the context of metabolic dysregulations associated with fatty liver, where insulin resistance has been assumed to play an important role. Special emphasis has been given to digital imaging studies and in particular to positron emission tomography, as it represents a unique opportunity for the noninvasive in vivo study of tissue metabolism. An exhaustive revision of targeted animal models is also provided in order to clarify what the available preclinical evidence suggests for the causal interactions between fatty liver, dysregulated endogenous glucose production and insulin resistance.
Collapse
Affiliation(s)
- Eleni Rebelos
- Turku PET Centre, University of Turku, Turku 20500, Finland
| | - Patricia Iozzo
- Institute of Clinical Physiology, National Research Council, Pisa 56124, Italy
| | | | - Maurizia Rossana Brunetto
- Hepatology Unit and Laboratory of Molecular Genetics and Pathology of Hepatitis, Pisa University Hospital, Pisa 56121, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56121, Italy
- Institute of Biostructure and Bioimaging, National Research Council, Napoli 80145, Italy
| | - Ferruccio Bonino
- Institute of Biostructure and Bioimaging, National Research Council, Napoli 80145, Italy
| |
Collapse
|
24
|
Carli M, Kolachalam S, Longoni B, Pintaudi A, Baldini M, Aringhieri S, Fasciani I, Annibale P, Maggio R, Scarselli M. Atypical Antipsychotics and Metabolic Syndrome: From Molecular Mechanisms to Clinical Differences. Pharmaceuticals (Basel) 2021; 14:238. [PMID: 33800403 PMCID: PMC8001502 DOI: 10.3390/ph14030238] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/15/2022] Open
Abstract
Atypical antipsychotics (AAPs) are commonly prescribed medications to treat schizophrenia, bipolar disorders and other psychotic disorders. However, they might cause metabolic syndrome (MetS) in terms of weight gain, dyslipidemia, type 2 diabetes (T2D), and high blood pressure, which are responsible for reduced life expectancy and poor adherence. Importantly, there is clear evidence that early metabolic disturbances can precede weight gain, even if the latter still remains the hallmark of AAPs use. In fact, AAPs interfere profoundly with glucose and lipid homeostasis acting mostly on hypothalamus, liver, pancreatic β-cells, adipose tissue, and skeletal muscle. Their actions on hypothalamic centers via dopamine, serotonin, acetylcholine, and histamine receptors affect neuropeptides and 5'AMP-activated protein kinase (AMPK) activity, thus producing a supraphysiological sympathetic outflow augmenting levels of glucagon and hepatic glucose production. In addition, altered insulin secretion, dyslipidemia, fat deposition in the liver and adipose tissues, and insulin resistance become aggravating factors for MetS. In clinical practice, among AAPs, olanzapine and clozapine are associated with the highest risk of MetS, whereas quetiapine, risperidone, asenapine and amisulpride cause moderate alterations. The new AAPs such as ziprasidone, lurasidone and the partial agonist aripiprazole seem more tolerable on the metabolic profile. However, these aspects must be considered together with the differences among AAPs in terms of their efficacy, where clozapine still remains the most effective. Intriguingly, there seems to be a correlation between AAP's higher clinical efficacy and increase risk of metabolic alterations. Finally, a multidisciplinary approach combining psychoeducation and therapeutic drug monitoring (TDM) is proposed as a first-line strategy to avoid the MetS. In addition, pharmacological treatments are discussed as well.
Collapse
Affiliation(s)
- Marco Carli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (S.K.); (B.L.); (A.P.); (M.B.); (S.A.)
| | - Shivakumar Kolachalam
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (S.K.); (B.L.); (A.P.); (M.B.); (S.A.)
| | - Biancamaria Longoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (S.K.); (B.L.); (A.P.); (M.B.); (S.A.)
| | - Anna Pintaudi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (S.K.); (B.L.); (A.P.); (M.B.); (S.A.)
| | - Marco Baldini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (S.K.); (B.L.); (A.P.); (M.B.); (S.A.)
| | - Stefano Aringhieri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (S.K.); (B.L.); (A.P.); (M.B.); (S.A.)
| | - Irene Fasciani
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (I.F.); (R.M.)
| | - Paolo Annibale
- Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany;
| | - Roberto Maggio
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (I.F.); (R.M.)
| | - Marco Scarselli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (S.K.); (B.L.); (A.P.); (M.B.); (S.A.)
| |
Collapse
|
25
|
Deregulated Serotonin Pathway in Women with Morbid Obesity and NAFLD. Life (Basel) 2020; 10:life10100245. [PMID: 33081272 PMCID: PMC7603041 DOI: 10.3390/life10100245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/08/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) extends from simple steatosis (SS) to non-alcoholic steatohepatitis (NASH). Peripheral serotonin (5-HT) has become as an important regulator of different metabolic pathways. 5-HT has been related to obesity and lipid accumulation in the liver. The objective of this study was to assess the relationship between the 5-HT signaling pathway and the degree of NAFLD, as well as to investigate whether peripheral 5-HT levels are related to the hepatic and jejunal mRNA abundance of serotonin receptors (HTR) in a cohort of women with morbid obesity (MO) and NAFLD. ELISA was used to quantify the serum 5-HT from normal-weight subjects (n = 26) and patients with MO (n = 58). We used RTq-PCR analysis to evaluate the relative expression of HTR in women with MO with normal liver (n = 22), SS (n = 21), and NASH (n = 15). The 5-HT was diminished in women with MO under a hypocaloric diet, regardless of the presence of NAFLD. Additionally, we report a negative correlation of 5-HT levels with metabolic syndrome criteria, suggesting that serotonin may have a protective role in obesity. Additionally, the hepatic expression of HTR2A and HTR2B were decreased in women with MO and NAFLD, but no significant differences in the HTR jejunal expression according to the presence of NAFLD were found.
Collapse
|
26
|
Wang Z, Zhao P, Zhang Y, Shi S, Chen X. The hepatoprotective effect and mechanism of lotus leaf on liver injury induced by Genkwa Flos. J Pharm Pharmacol 2020; 72:1909-1920. [PMID: 32979237 DOI: 10.1111/jphp.13355] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVES As a traditional Chinese medicine, lotus leaf was reported to have significant hepatoprotective effect. To explore the hepatoprotective mechanism of lotus leaf, a rapid and reliable UPLC-MS/MS method was conducted to simultaneously determine six specific endogenous substances including 5-oxoproline, phenylalanine, tryptophan, C18 -phytosphingosine, lysophosphatidylcholine (16 : 0) and lysophosphatidylcholine (18 : 1). METHODS With the help of HPLC-FT-ICR-MS, the chemical constituents of louts leaf extract were elucidated. By observing histopathological changes and determining hepatotoxicity-related biochemical indicators, rat model of liver injury was developed and the hepatoprotective effect of lotus leaf was verified. With the developed UPLC-MS/MS method, six endogenous metabolites related to hepatotoxicity were monitored to investigate the hepatoprotective mechanism of lotus leaf. KEY FINDINGS In the qualitative analysis, a total of twenty compounds including ten flavonoids, nine alkaloids and one proanthocyanidin were identified. Based on the results of determining six endogenous metabolites related to hepatotoxicity, it was predicted that the hepatoprotective mechanism of lotus leaf might be related to glutathione metabolism, phenylalanine metabolism, tryptophan metabolism, sphingolipid metabolism and phospholipid metabolism. CONCLUSIONS This study could be a meaningful investigation to provide mechanistic insights into the hepatoprotective effect of lotus leaf and further lay a theoretical basis for the clinical application of lotus leaf.
Collapse
Affiliation(s)
- Zhipeng Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China.,Department of Pharmacy, Hebei General Hospital, Shijiazhuang, China
| | - Panpan Zhao
- Department of Endocrinology and Metabolism, Hebei Provincial Chest Hospital, Shijiazhuang, China
| | - Yuanyuan Zhang
- Department of Pharmaceutical Analysis, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Shan Shi
- Department of Pharmacy, the First Hospital of China Medical University, Shenyang, China
| | - Xiaohui Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
27
|
An Increase in Liver Polyamine Concentration Contributes to the Tryptophan-Induced Acute Stimulation of Rat Hepatic Protein Synthesis. Nutrients 2020; 12:nu12092665. [PMID: 32882842 PMCID: PMC7551729 DOI: 10.3390/nu12092665] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 01/02/2023] Open
Abstract
Tryptophan has a unique role as a nutritional signaling molecule that regulates protein synthesis in mouse and rat liver. However, the mechanism underlying the stimulating actions of tryptophan on hepatic protein synthesis remains unclear. Proteomic and metabolomic analyses were performed to identify candidate proteins and metabolites likely to play a role in the stimulation of protein synthesis by tryptophan. Overnight-fasted rats were orally administered L-tryptophan and then sacrificed 1 or 3 h after administration. Four differentially expressed protein spots were detected in rat liver at 3 h after tryptophan administration, of which one was identified as an ornithine aminotransferase (OAT) precursor. OAT is the main catabolic enzyme for ornithine, and its expression was significantly decreased by tryptophan administration. The concentration of ornithine was increased in the liver at 3 h after tryptophan administration. Ornithine is a precursor for polyamine biosynthesis. Significantly increased concentrations of polyamines were found in the liver at 3 h after administration of tryptophan. Additionally, enhanced hepatic protein synthesis was demonstrated by oral administration of putrescine. We speculate that the increase in ornithine level through suppression of OAT expression by tryptophan administration may lead to accelerated polyamine synthesis, thereby promoting protein synthesis in the liver.
Collapse
|
28
|
Feng H, Wu YQ, Xu YS, Wang KX, Qin XM, Lu YF. LC-MS-Based Metabolomic Study of Oleanolic Acid-Induced Hepatotoxicity in Mice. Front Pharmacol 2020; 11:747. [PMID: 32670053 PMCID: PMC7326119 DOI: 10.3389/fphar.2020.00747] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022] Open
Abstract
Oleanolic acid (OA), a natural triterpenoid, which has the development prospects in anti-tumor therapy is a widely used hepatoprotective drug in China. It has been reported that OA can cause liver toxicity after higher doses or longer-term use. Therefore, the study aims to explore the possible hepatotoxicity mechanism based on liver metabolic profiles. Liver metabolic profiles were obtained from untargeted ultrahigh performance liquid chromatography (UHPLC)-Q Exactive Orbitrap mass spectrometry (MS) technique. It was found that altered bile acid, amino acid, and energy metabolism might be at least partly responsible for OA-induced hepatotoxicity. Bile acid metabolism, as the most important pathway, was verified by using UHPLC-TSQ-MS, indicating that conjugated bile acids were the main contributors to OA-induced liver toxicity. Our findings confirmed that increased bile acids were the key element of OA hepatotoxicity, which may open new insights for OA hepatotoxicity in-depth investigations, as well as provide a reference basis for more hepatotoxic drug mechanism research.
Collapse
Affiliation(s)
- Hong Feng
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Ying-Qiu Wu
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Ya-Sha Xu
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Ke-Xin Wang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China
| | - Yuan-Fu Lu
- Key Laboratory of Basic Pharmacology of the Ministry of Education and Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
29
|
Kim M, Hwang I, Pagire HS, Pagire SH, Choi W, Choi WG, Yoon J, Lee WM, Song JS, Yoo EK, Lee SM, Kim MJ, Bae MA, Kim D, Lee H, Lee EY, Jeon JH, Lee IK, Kim H, Ahn JH. Design, Synthesis, and Biological Evaluation of New Peripheral 5HT2A Antagonists for Nonalcoholic Fatty Liver Disease. J Med Chem 2020; 63:4171-4182. [DOI: 10.1021/acs.jmedchem.0c00002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Minhee Kim
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Inseon Hwang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Haushabhau S. Pagire
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Suvarna H. Pagire
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Wonsuk Choi
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Won Gun Choi
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jihyeon Yoon
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Won Mi Lee
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jin Sook Song
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Eun Kyung Yoo
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41010, Republic of Korea
| | - Seung Mi Lee
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41010, Republic of Korea
| | - Mi-jin Kim
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41010, Republic of Korea
| | - Myung Ae Bae
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Dooseop Kim
- R&D center, JD Bioscience, 123 Cheomdan-dwagiro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Heejong Lee
- R&D center, JD Bioscience, 123 Cheomdan-dwagiro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Eun-Young Lee
- R&D center, JD Bioscience, 123 Cheomdan-dwagiro, Buk-gu, Gwangju 61005, Republic of Korea
| | - Jae-Han Jeon
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41010, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - In-Kyu Lee
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41010, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jin Hee Ahn
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
- R&D center, JD Bioscience, 123 Cheomdan-dwagiro, Buk-gu, Gwangju 61005, Republic of Korea
| |
Collapse
|
30
|
Nouwen LV, Everts B. Pathogens MenTORing Macrophages and Dendritic Cells: Manipulation of mTOR and Cellular Metabolism to Promote Immune Escape. Cells 2020; 9:cells9010161. [PMID: 31936570 PMCID: PMC7017145 DOI: 10.3390/cells9010161] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/01/2020] [Accepted: 01/07/2020] [Indexed: 02/06/2023] Open
Abstract
Myeloid cells, including macrophages and dendritic cells, represent an important first line of defense against infections. Upon recognition of pathogens, these cells undergo a metabolic reprogramming that supports their activation and ability to respond to the invading pathogens. An important metabolic regulator of these cells is mammalian target of rapamycin (mTOR). During infection, pathogens use host metabolic pathways to scavenge host nutrients, as well as target metabolic pathways for subversion of the host immune response that together facilitate pathogen survival. Given the pivotal role of mTOR in controlling metabolism and DC and macrophage function, pathogens have evolved strategies to target this pathway to manipulate these cells. This review seeks to discuss the most recent insights into how pathogens target DC and macrophage metabolism to subvert potential deleterious immune responses against them, by focusing on the metabolic pathways that are known to regulate and to be regulated by mTOR signaling including amino acid, lipid and carbohydrate metabolism, and autophagy.
Collapse
|
31
|
Lei X, Zhang C. Predicting metabolite-disease associations based on KATZ model. BioData Min 2019; 12:19. [PMID: 31673292 PMCID: PMC6815005 DOI: 10.1186/s13040-019-0206-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 09/12/2019] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Increasing numbers of evidences have illuminated that metabolites can respond to pathological changes. However, identifying the diseases-related metabolites is a magnificent challenge in the field of biology and medicine. Traditional medical equipment not only has the limitation of its accuracy but also is expensive and time-consuming. Therefore, it's necessary to take advantage of computational methods for predicting potential associations between metabolites and diseases. RESULTS In this study, we develop a computational method based on KATZ algorithm to predict metabolite-disease associations (KATZMDA). Firstly, we extract data about metabolite-disease pairs from the latest version of HMDB database for the materials of prediction. Then we take advantage of disease semantic similarity and the improved disease Gaussian Interaction Profile (GIP) kernel similarity to obtain more reliable disease similarity and enhance the predictive performance of our proposed computational method. Simultaneously, KATZ algorithm is applied in the domains of metabolomics for the first time. CONCLUSIONS According to three kinds of cross validations and case studies of three common diseases, KATZMDA is worth serving as an impactful measuring tool for predicting the potential associations between metabolites and diseases.
Collapse
Affiliation(s)
- Xiujuan Lei
- School of Computer Science, Shaanxi Normal University, Xi’an, 710119 Shaanxi China
| | - Cheng Zhang
- School of Computer Science, Shaanxi Normal University, Xi’an, 710119 Shaanxi China
| |
Collapse
|
32
|
Challa TD, Wueest S, Lucchini FC, Dedual M, Modica S, Borsigova M, Wolfrum C, Blüher M, Konrad D. Liver ASK1 protects from non-alcoholic fatty liver disease and fibrosis. EMBO Mol Med 2019; 11:e10124. [PMID: 31595673 PMCID: PMC6783644 DOI: 10.15252/emmm.201810124] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/15/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is strongly associated with obesity and may progress to non-alcoholic steatohepatitis (NASH) and liver fibrosis. The deficit of pharmacological therapies for the latter mainly results from an incomplete understanding of involved pathological mechanisms. Herein, we identify apoptosis signal-regulating kinase 1 (ASK1) as a suppressor of NASH and fibrosis formation. High-fat diet-fed and aged chow-fed liver-specific ASK1-knockout mice develop a higher degree of hepatic steatosis, inflammation, and fibrosis compared to controls. In addition, pharmacological inhibition of ASK1 increased hepatic lipid accumulation in wild-type mice. In line, liver-specific ASK1 overexpression protected mice from the development of high-fat diet-induced hepatic steatosis and carbon tetrachloride-induced fibrosis. Mechanistically, ASK1 depletion blunts autophagy, thereby enhancing lipid droplet accumulation and liver fibrosis. In human livers of lean and obese subjects, ASK1 expression correlated negatively with liver fat content and NASH scores, but positively with markers for autophagy. Taken together, ASK1 may be a novel therapeutic target to tackle NAFLD and liver fibrosis.
Collapse
Affiliation(s)
- Tenagne D Challa
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
| | - Stephan Wueest
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
| | - Fabrizio C Lucchini
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
- Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
| | - Mara Dedual
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
- Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
| | - Salvatore Modica
- Institute of Food, Nutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | - Marcela Borsigova
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
| | - Christian Wolfrum
- Institute of Food, Nutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | | | - Daniel Konrad
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
- Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
| |
Collapse
|
33
|
Yabut JM, Crane JD, Green AE, Keating DJ, Khan WI, Steinberg GR. Emerging Roles for Serotonin in Regulating Metabolism: New Implications for an Ancient Molecule. Endocr Rev 2019; 40:1092-1107. [PMID: 30901029 PMCID: PMC6624793 DOI: 10.1210/er.2018-00283] [Citation(s) in RCA: 206] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 03/18/2019] [Indexed: 12/12/2022]
Abstract
Serotonin is a phylogenetically ancient biogenic amine that has played an integral role in maintaining energy homeostasis for billions of years. In mammals, serotonin produced within the central nervous system regulates behavior, suppresses appetite, and promotes energy expenditure by increasing sympathetic drive to brown adipose tissue. In addition to these central circuits, emerging evidence also suggests an important role for peripheral serotonin as a factor that enhances nutrient absorption and storage. Specifically, glucose and fatty acids stimulate the release of serotonin from the duodenum, promoting gut peristalsis and nutrient absorption. Serotonin also enters the bloodstream and interacts with multiple organs, priming the body for energy storage by promoting insulin secretion and de novo lipogenesis in the liver and white adipose tissue, while reducing lipolysis and the metabolic activity of brown and beige adipose tissue. Collectively, peripheral serotonin acts as an endocrine factor to promote the efficient storage of energy by upregulating lipid anabolism. Pharmacological inhibition of serotonin synthesis or signaling in key metabolic tissues are potential drug targets for obesity, type 2 diabetes, and nonalcoholic fatty liver disease (NAFLD).
Collapse
Affiliation(s)
- Julian M Yabut
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Justin D Crane
- Department of Biology, Northeastern University, Boston, Massachusetts
| | - Alexander E Green
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Damien J Keating
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - Waliul I Khan
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gregory R Steinberg
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
34
|
Hirose M, Schilf P, Zarse K, Busch H, Fuellen G, Jöhren O, Köhling R, König IR, Richer B, Rupp J, Schwaninger M, Seeger K, Sina C, Ristow M, Ibrahim SM. Maternally Inherited Differences within Mitochondrial Complex I Control Murine Healthspan. Genes (Basel) 2019; 10:genes10070532. [PMID: 31337008 PMCID: PMC6678443 DOI: 10.3390/genes10070532] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/02/2019] [Accepted: 07/10/2019] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial complex I-the largest enzyme complex of the mitochondrial oxidative phosphorylation machinery-has been proposed to contribute to a variety of age-related pathological alterations as well as longevity. The enzyme complex-consisting proteins are encoded by both nuclear (nDNA) and mitochondrial DNA (mtDNA). While some association studies of mtDNA encoded complex I genes and lifespan in humans have been reported, experimental evidence and the functional consequence of such variants is limited to studies using invertebrate models. Here, we present experimental evidence that a homoplasmic mutation in the mitochondrially encoded complex I gene mt-Nd2 modulates lifespan by altering cellular tryptophan levels and, consequently, ageing-related pathways in mice. A conplastic mouse strain carrying a mutation at m.4738C > A in mt-Nd2 lived slightly, but significantly, shorter than the controls did. The same mutation led to a higher susceptibility to glucose intolerance induced by high-fat diet feeding. These phenotypes were not observed in mice carrying a mutation in another mtDNA encoded complex I gene, mt-Nd5, suggesting the functional relevance of particular mutations in complex I to ageing and age-related diseases.
Collapse
Affiliation(s)
- Misa Hirose
- Luebeck Institute of Experimental Dermatology, University of Luebeck, 23562 Luebeck, Germany
| | - Paul Schilf
- Luebeck Institute of Experimental Dermatology, University of Luebeck, 23562 Luebeck, Germany
| | - Kim Zarse
- Energy Metabolism Laboratory, Institute of Translational Medicine, Swiss Federal Institute of Technology (ETH) Zurich, 8603 Schwerzenbach, Switzerland
| | - Hauke Busch
- Group of Systems Biology, Institute of Cardiogenetics and Luebeck Institute of Experimental Dermatology, University of Luebeck, 23562 Luebeck, Germany
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, 18057 Rostock, Germany
| | - Olaf Jöhren
- Center of Brain, Behavior & Metabolism, University of Luebeck, 23562 Luebeck, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Inke R König
- Institute of Medical Biometry and Statistics, University of Luebeck, 23562 Luebeck, Germany
| | - Barbara Richer
- Institute of Chemistry and Metabolomics, University of Luebeck, 23562 Luebeck, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Luebeck, 23562 Luebeck, Germany
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Luebeck, 23562 Luebeck, Germany
| | - Karsten Seeger
- Institute of Chemistry and Metabolomics, University of Luebeck, 23562 Luebeck, Germany
| | - Christian Sina
- Institute for Nutritional Medicine, University of Luebeck, 23562 Luebeck, Germany
| | - Michael Ristow
- Energy Metabolism Laboratory, Institute of Translational Medicine, Swiss Federal Institute of Technology (ETH) Zurich, 8603 Schwerzenbach, Switzerland
| | - Saleh M Ibrahim
- Luebeck Institute of Experimental Dermatology, University of Luebeck, 23562 Luebeck, Germany.
| |
Collapse
|
35
|
Hendrikx T, Schnabl B. Indoles: metabolites produced by intestinal bacteria capable of controlling liver disease manifestation. J Intern Med 2019; 286:32-40. [PMID: 30873652 DOI: 10.1111/joim.12892] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alterations in the bacteria that reside in our gastrointestinal tract play a role in the pathogenesis and progression of many disorders including liver and gastrointestinal diseases. Both qualitative (composition) and quantitative (amount) changes in gut microbes are associated with increased susceptibility to liver disease. Importantly, the intestinal microbiota is involved in the regulation of many host signalling pathways via the generation of different metabolites. Hence, dysbiosis influences disease development and progression by directly affecting the host-bacteria metabolic interaction. Microbe-derived harmful metabolites can translocate to distant organs due to increased intestinal permeability as observed during dysbiosis. Contrary, certain bacterial metabolites such as tryptophan metabolites contribute to intestinal and systemic homeostasis. Here, we provide an overview of current evidence describing to what extent microbial metabolites modulate the development of chronic liver diseases such as alcoholic steatohepatitis and nonalcoholic fatty liver disease with a special emphasis on indoles.
Collapse
Affiliation(s)
- T Hendrikx
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - B Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.,Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
36
|
Regulation of obesity-associated metabolic disturbance by the antipsychotic drug olanzapine: Role of the autophagy-lysosome pathway. Biochem Pharmacol 2018; 158:114-125. [DOI: 10.1016/j.bcp.2018.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 10/01/2018] [Indexed: 11/22/2022]
|
37
|
Choi W, Namkung J, Hwang I, Kim H, Lim A, Park HJ, Lee HW, Han KH, Park S, Jeong JS, Bang G, Kim YH, Yadav VK, Karsenty G, Ju YS, Choi C, Suh JM, Park JY, Park S, Kim H. Serotonin signals through a gut-liver axis to regulate hepatic steatosis. Nat Commun 2018; 9:4824. [PMID: 30446669 PMCID: PMC6240035 DOI: 10.1038/s41467-018-07287-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 10/19/2018] [Indexed: 12/13/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is increasing in worldwide prevalence, closely tracking the obesity epidemic, but specific pharmaceutical treatments for NAFLD are lacking. Defining the key molecular pathways underlying the pathogenesis of NAFLD is essential for developing new drugs. Here we demonstrate that inhibition of gut-derived serotonin synthesis ameliorates hepatic steatosis through a reduction in liver serotonin receptor 2A (HTR2A) signaling. Local serotonin concentrations in the portal blood, which can directly travel to and affect the liver, are selectively increased by high-fat diet (HFD) feeding in mice. Both gut-specific Tph1 knockout mice and liver-specific Htr2a knockout mice are resistant to HFD-induced hepatic steatosis, without affecting systemic energy homeostasis. Moreover, selective HTR2A antagonist treatment prevents HFD-induced hepatic steatosis. Thus, the gut TPH1-liver HTR2A axis shows promise as a drug target to ameliorate NAFLD with minimal systemic metabolic effects. No effective pharmacological treatments exist for nonalcoholic fatty liver disease (NAFLD). Here, the authors show that serotonin concentration in the portal blood is increased in nine human subjects and in mice fed a high-fat diet, and that local serotonin signaling ablation, either genetically or with an antagonist, prevents hepatic steatosis in mice.
Collapse
Affiliation(s)
- Wonsuk Choi
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Jun Namkung
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea.,Department of Biochemistry, Yonsei University Wonju College of Medicine, Wonju, 26426, Republic of Korea
| | - Inseon Hwang
- Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon, 34141, Republic of Korea
| | - Hyeongseok Kim
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Ajin Lim
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Hye Jung Park
- Department of Internal Medicine, Yonsei University College of Medicine, Yonsei Liver Center, Severance Hospital, Seoul, 03722, Republic of Korea
| | - Hye Won Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Yonsei Liver Center, Severance Hospital, Seoul, 03722, Republic of Korea
| | - Kwang-Hyub Han
- Department of Internal Medicine, Yonsei University College of Medicine, Yonsei Liver Center, Severance Hospital, Seoul, 03722, Republic of Korea
| | - Seongyeol Park
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Ji-Seon Jeong
- Center for Bioanalysis, Division of Metrology for Quality of Life, Korea Research Institute of Standards and Science, Daejeon, 34113, Republic of Korea
| | - Geul Bang
- Biomedical Omics Group, Korea Basic Science Institute, Chungbuk, 28119, Republic of Korea
| | - Young Hwan Kim
- Biomedical Omics Group, Korea Basic Science Institute, Chungbuk, 28119, Republic of Korea
| | - Vijay K Yadav
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, 10032, USA
| | - Gerard Karsenty
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, 10032, USA
| | - Young Seok Ju
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Chan Choi
- Department of Pathology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Jae Myoung Suh
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea.,Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon, 34141, Republic of Korea
| | - Jun Yong Park
- Department of Internal Medicine, Yonsei University College of Medicine, Yonsei Liver Center, Severance Hospital, Seoul, 03722, Republic of Korea.
| | - Sangkyu Park
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea. .,Department of Biochemistry, College of Medicine, Catholic Kwandong University, Gangneung, 25601, Republic of Korea.
| | - Hail Kim
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, 34141, Republic of Korea. .,Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon, 34141, Republic of Korea. .,KAIST Institute for the BioCentury, KAIST, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
38
|
Niture S, Gyamfi MA, Kedir H, Arthur E, Ressom H, Deep G, Kumar D. Serotonin induced hepatic steatosis is associated with modulation of autophagy and notch signaling pathway. Cell Commun Signal 2018; 16:78. [PMID: 30409162 PMCID: PMC6225666 DOI: 10.1186/s12964-018-0282-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/09/2018] [Indexed: 01/08/2023] Open
Abstract
Background Besides its neurotransmitter and vasoconstriction functions, serotonin is an important mediator of numerous biological processes in peripheral tissues including cell proliferation, steatosis, and fibrogenesis. Recent reports indicate that serotonin may promote tumor growth in liver cancer, however, the molecular mechanisms remain elusive. n this study, we investigated the role and molecular signaling mechanisms mediated by serotonin in liver cancer cell survival, drug resistance, and steatosis. Methods Effect of serotonin on modulation of cell survival/proliferation was determined by MTT/WST1 assay. Effect of serotonin on the regulation of autophagy biomarkers and lipid/fatty acid proteins expression, AKT/mTOR and Notch signaling was evaluated by immunoblotting. The role of serotonin in normal human hepatocytes and liver cancer cell steatosis was analyzed by Oil Red O staining. The mRNA expression levels of lipid/fatty acid proteins and serotonin receptors were validated by qRT-PCR. The important roles of autophagy, Notch signaling, serotonin receptors and serotonin re-uptake proteins on serotonin-mediated cell steatosis were investigated by using selective inhibitors or antagonists. The association of peripheral serotonin, autophagy, and hepatic steatosis was also investigated using chronic EtOH fed mouse model. Results Exposure of liver cancer cells to serotonin induced Notch signaling and autophagy, independent of AKT/mTOR pathway. Also, serotonin enhanced cancer cell proliferation/survival and drug resistance. Furthermore, serotonin treatment up-regulated the expression of lipogenic proteins and increased steatosis in liver cancer cells. Inhibition of autophagy or Notch signaling reduced serotonin-mediated cell steatosis. Treatment with serotonin receptor antagonists 5-HTr1B and 5-HTr2B reduced serotonin-mediated cell steatosis; in contrast, treatment with selective serotonin reuptake inhibitors (SSRIs) increased steatosis. In addition, mice fed with chronic EtOH resulted in increased serum serotonin levels which were associated with the induction of hepatic steatosis and autophagy. Conclusions Serotonin regulates liver cancer cell steatosis, cells survival, and may promote liver carcinogenesis by activation of Notch signaling and autophagy. Electronic supplementary material The online version of this article (10.1186/s12964-018-0282-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Suryakant Niture
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 1801 Fayetteville St, Durham, NC, 27707, USA
| | - Maxwell A Gyamfi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 1801 Fayetteville St, Durham, NC, 27707, USA
| | - Habib Kedir
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 1801 Fayetteville St, Durham, NC, 27707, USA
| | - Elena Arthur
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 1801 Fayetteville St, Durham, NC, 27707, USA
| | - Habtom Ressom
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20008, USA
| | - Gagan Deep
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, 27109, USA
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 1801 Fayetteville St, Durham, NC, 27707, USA. .,Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC, 27707, USA. .,Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20008, USA.
| |
Collapse
|
39
|
Beaumont M, Neyrinck AM, Olivares M, Rodriguez J, de Rocca Serra A, Roumain M, Bindels LB, Cani PD, Evenepoel P, Muccioli GG, Demoulin JB, Delzenne NM. The gut microbiota metabolite indole alleviates liver inflammation in mice. FASEB J 2018; 32:fj201800544. [PMID: 29906245 PMCID: PMC6219839 DOI: 10.1096/fj.201800544] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/29/2018] [Indexed: 12/20/2022]
Abstract
The gut microbiota regulates key hepatic functions, notably through the production of bacterial metabolites that are transported via the portal circulation. We evaluated the effects of metabolites produced by the gut microbiota from aromatic amino acids (phenylacetate, benzoate, p-cresol, and indole) on liver inflammation induced by bacterial endotoxin. Precision-cut liver slices prepared from control mice, Kupffer cell (KC)-depleted mice, and obese mice ( ob/ ob) were treated with or without LPS and bacterial metabolites. We observed beneficial effects of indole that dose-dependently reduced the LPS-induced up-regulation of proinflammatory mediators at both mRNA and protein levels in precision-cut liver slices prepared from control or ob/ ob mice. KC depletion partly prevented the antiinflammatory effects of indole, notably through a reduction of nucleotide-binding domain and leucine-rich repeat containing (NLR) family pyrin domain-containing 3 (NLRP3) pathway activation. In vivo, the oral administration of indole before an LPS injection reduced the expression of key proteins of the NF-κB pathway and downstream proinflammatory gene up-regulation. Indole also prevented LPS-induced alterations of cholesterol metabolism through a transcriptional regulation associated with increased 4β-hydroxycholesterol hepatic levels. In summary, indole appears as a bacterial metabolite produced from tryptophan that is able to counteract the detrimental effects of LPS in the liver. Indole could be a new target to develop innovative strategies to decrease hepatic inflammation.-Beaumont, M., Neyrinck, A. M., Olivares, M., Rodriguez, J., de Rocca Serra, A., Roumain, M., Bindels, L. B., Cani, P. D., Evenepoel, P., Muccioli, G. G., Demoulin, J.-B., Delzenne, N. M. The gut microbiota metabolite indole alleviates liver inflammation in mice.
Collapse
Affiliation(s)
- Martin Beaumont
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Audrey M. Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Marta Olivares
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Audrey de Rocca Serra
- Pole of Experimental Medicine, De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Martin Roumain
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Laure B. Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Patrice D. Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Pieter Evenepoel
- Department of Immunology and Microbiology, Laboratory of Nephrology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Giulio G. Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Jean-Baptiste Demoulin
- Pole of Experimental Medicine, De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
40
|
Namkung J, Shong KE, Kim H, Oh CM, Park S, Kim H. Inhibition of Serotonin Synthesis Induces Negative Hepatic Lipid Balance. Diabetes Metab J 2018; 42:233-243. [PMID: 29885107 PMCID: PMC6015967 DOI: 10.4093/dmj.2017.0084] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/14/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Hepatic steatosis is caused by metabolic stress associated with a positive lipid balance, such as insulin resistance and obesity. Previously we have shown the anti-obesity effects of inhibiting serotonin synthesis, which eventually improved insulin sensitivity and hepatic steatosis. However, it is not clear whether serotonin has direct effect on hepatic lipid accumulation. Here, we showed the possibility of direct action of serotonin on hepatic steatosis. METHODS Mice were treated with para-chlorophenylalanine (PCPA) or LP-533401 to inhibit serotonin synthesis and fed with high fat diet (HFD) or high carbohydrate diet (HCD) to induce hepatic steatosis. Hepatic triglyceride content and gene expression profiles were analyzed. RESULTS Pharmacological and genetic inhibition of serotonin synthesis reduced HFD-induced hepatic lipid accumulation. Furthermore, short-term PCPA treatment prevented HCD-induced hepatic steatosis without affecting glucose tolerance and browning of subcutaneous adipose tissue. Gene expression analysis revealed that the expressions of genes involved in de novo lipogenesis and triacylglycerol synthesis were downregulated by short-term PCPA treatment as well as long-term PCPA treatment. CONCLUSION Short-term inhibition of serotonin synthesis prevented hepatic lipid accumulation without affecting systemic insulin sensitivity and energy expenditure, suggesting the direct steatogenic effect of serotonin in liver.
Collapse
Affiliation(s)
- Jun Namkung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
- Department of Biochemistry, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Ko Eun Shong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Hyeongseok Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Chang Myung Oh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Sangkyu Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
- Department of Biochemistry, Catholic Kwandong University College of Medicine, Gangneung, Korea.
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon, Korea.
| |
Collapse
|
41
|
Reifsnyder PC, Ryzhov S, Flurkey K, Anunciado-Koza RP, Mills I, Harrison DE, Koza RA. Cardioprotective effects of dietary rapamycin on adult female C57BLKS/J-Lepr db mice. Ann N Y Acad Sci 2018; 1418:106-117. [PMID: 29377150 PMCID: PMC5934313 DOI: 10.1111/nyas.13557] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/24/2017] [Accepted: 10/30/2017] [Indexed: 02/06/2023]
Abstract
Rapamycin (RAPA), an inhibitor of mTORC signaling, has been shown to extend life span in mice and other organisms. Recently, animal and human studies have suggested that inhibition of mTORC signaling can alleviate or prevent the development of cardiomyopathy. In view of this, we used a murine model of type 2 diabetes (T2D), BKS-Leprdb , to determine whether RAPA treatment can mitigate the development of T2D-induced cardiomyopathy in adult mice. Female BKS-Leprdb mice fed diet supplemented with RAPA from 11 to 27 weeks of age showed reduced weight gain and significant reductions of fat and lean mass compared with untreated mice. No differences in plasma glucose or insulin levels were observed between groups; however, RAPA-treated mice were more insulin sensitive (P < 0.01) than untreated mice. Urine albumin/creatinine ratio was lower in RAPA-treated mice, suggesting reduced diabetic nephropathy and improved kidney function. Echocardiography showed significantly reduced left ventricular wall thickness in mice treated with RAPA compared with untreated mice (P = 0.02) that was consistent with reduced heart weight/tibia length ratios, reduced myocyte size and cardiac fibrosis measured by histomorphology, and reduced mRNA expression of Col1a1, a marker for cardiomyopathy. Our results suggest that inhibition of mTORC signaling is a plausible strategy for ameliorating complications of obesity and T2D, including cardiomyopathy.
Collapse
Affiliation(s)
| | - Sergey Ryzhov
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine
| | | | - Rea P Anunciado-Koza
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine
| | - Ian Mills
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine
| | | | - Robert A Koza
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine
| |
Collapse
|
42
|
Chang H, Meng HY, Liu SM, Wang Y, Yang XX, Lu F, Wang HY. Identification of key metabolic changes during liver fibrosis progression in rats using a urine and serum metabolomics approach. Sci Rep 2017; 7:11433. [PMID: 28900168 PMCID: PMC5595818 DOI: 10.1038/s41598-017-11759-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 08/29/2017] [Indexed: 12/21/2022] Open
Abstract
Reversibility of hepatic fibrosis is an intrinsic response to chronic injury, and with on-going damage, fibrosis can progress to its end-stage consequence, cirrhosis. Non-invasive and reliable biomarkers for early detection of liver fibrosis are needed. Based on the CCl4-induced liver fibrosis rat model, urinary and serum metabolic profiling performed by LC-QTOF-MS associated with histological progression were utilized to identify liver fibrosis-specific potential biomarkers for early prediction and to reveal significant fibrotic pathways and their dynamic changes in different stages of liver fibrosis. Finally, nine differential metabolites in urine and ten in serum were selected and identified involving the most relevant metabolic pathways. Perturbations of tryptophan, valine, leucine, isoleucine, and citrate (TCA) cycle metabolites, along with sphingolipid and glycerophospholipid metabolites, occurred from the onset of liver fibrosis. Furthermore, dysregulation of valine and bile acid biosynthesis metabolites occurred in the intermediate and advanced stages. More importantly, among these metabolites, urinary kynurenic acid, 5-hydroxyindoleacetyl glycine, 4-(2-amino-3-hydroxyphenyl)-2,4-dioxobutanoic acid and serum sphinganine, sphingomyelin, L-leucine, L-tryptophan, and LysoPC(17:0) changed at all time points and may serve as potential early biomarkers for the diagnosis of hepatic fibrosis and as therapeutic targets. Overall, this work evaluates the potential of these metabolites for the early detection of liver fibrosis.
Collapse
Affiliation(s)
- Hong Chang
- Chinese Medicine Toxicological Laboratory, Heilongjiang University of Chinese Medicine, Harbin, P.R. China
- School of Pharmacy, Baotou Medical College, Inner Mongolia, Baotou, P.R. China
| | - Hong-Yu Meng
- Chinese Medicine Toxicological Laboratory, Heilongjiang University of Chinese Medicine, Harbin, P.R. China
| | - Shu-Min Liu
- Chinese Medicine Toxicological Laboratory, Heilongjiang University of Chinese Medicine, Harbin, P.R. China.
- Drug Safety Evaluation Center, Heilongjiang University of Chinese Medicine, Harbin, P.R. China.
| | - Yu Wang
- Chinese Medicine Toxicological Laboratory, Heilongjiang University of Chinese Medicine, Harbin, P.R. China
| | - Xiao-Xu Yang
- Chinese Medicine Toxicological Laboratory, Heilongjiang University of Chinese Medicine, Harbin, P.R. China
| | - Fang Lu
- Chinese Medicine Toxicological Laboratory, Heilongjiang University of Chinese Medicine, Harbin, P.R. China
| | - Hong-Yu Wang
- Chinese Medicine Toxicological Laboratory, Heilongjiang University of Chinese Medicine, Harbin, P.R. China
| |
Collapse
|
43
|
Xu K, Liu H, Bai M, Gao J, Wu X, Yin Y. Redox Properties of Tryptophan Metabolism and the Concept of Tryptophan Use in Pregnancy. Int J Mol Sci 2017; 18:E1595. [PMID: 28737706 PMCID: PMC5536082 DOI: 10.3390/ijms18071595] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/11/2017] [Accepted: 07/19/2017] [Indexed: 12/30/2022] Open
Abstract
During pregnancy, tryptophan (Trp) is required for several purposes, and Trp metabolism varies over time in the mother and fetus. Increased oxidative stress (OS) with high metabolic, energy and oxygen demands during normal pregnancy or in pregnancy-associated disorders has been reported. Taking the antioxidant properties of Trp and its metabolites into consideration, we made four hypotheses. First, the use of Trp and its metabolites is optional based on their antioxidant properties during pregnancy. Second, dynamic Trp metabolism is an accommodation mechanism in response to OS. Third, regulation of Trp metabolism could be used to control/attenuate OS according to variations in Trp metabolism during pregnancy. Fourth, OS-mediated injury could be alleviated by regulation of Trp metabolism in pregnancy-associated disorders. Future studies in normal/abnormal pregnancies and in associated disorders should include measurements of free Trp, total Trp, Trp metabolites, and activities of Trp-degrading enzymes in plasma. Abnormal pregnancies and some associated disorders may be associated with disordered Trp metabolism related to OS. Mounting evidence suggests that the investigation of the use of Trp and its metabolites in pregnancy will be meanful.
Collapse
Affiliation(s)
- Kang Xu
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| | - Hongnan Liu
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| | - Miaomiao Bai
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| | - Jing Gao
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| | - Xin Wu
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| | - Yulong Yin
- Chinese Academy of Sciences, Institute of Subtropical Agriculture, Key Laboratory of Agroecological Processes in Subtropical Region, Changsha 410125, China.
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha 410125, China.
- Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
- Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South Central, Ministry of Agriculture, Changsha 410125, China.
| |
Collapse
|
44
|
Kubo H, Hoshi M, Matsumoto T, Irie M, Oura S, Tsutsumi H, Hata Y, Yamamoto Y, Saito K. Sake lees extract improves hepatic lipid accumulation in high fat diet-fed mice. Lipids Health Dis 2017; 16:106. [PMID: 28578672 PMCID: PMC5457550 DOI: 10.1186/s12944-017-0501-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 05/25/2017] [Indexed: 02/06/2023] Open
Abstract
Background Nonalcoholic fatty liver disease (NAFLD) is increasing worldwide as one of the leading causes of chronic liver disease. Sake lees (SL) are secondary products of sake manufacturing and are considered to have beneficial effects on human health. To investigate these effects, we used high fat diet (HFD)-fed mice treated with or without the SL extract. Method Mice were the HFD ad libitum for 8 weeks and were administered 500 μL of distilled water with or without the SL extract (350 mg/mL) by a feeding needle daily for the last 4 weeks. Food intake, body weight, and liver weight were measured. Triacylglycerol content and the mRNA and protein expression levels of various lipid and glucose metabolism-related genes were determined in liver tissues. The levels of triglyceride, free fatty acids, glucose, insulin, and liver cell damage markers were determined in serum. Fatty acid-induced lipid accumulation in HepG2 cells was assessed in the presence or absence of the SL extract. Results Mice fed a HFD and treated with the SL extract demonstrated a significant reduction in hepatic lipid accumulation and mRNA and protein levels of peroxidome proliferator-activated receptor γ (PPARγ), PPARα, CD36, and phosphoenolpyruvate carboxykinase 1 in the liver, while the SL extract did not affect body weight and food intake. Moreover, insulin resistance and hepatic inflammation in HFD-fed mice improved after administration of the SL extract. In HepG2 cells, the SL extract suppressed fatty acid-induced intracellular lipid accumulation. Conclusions These findings suggest that treatment with the SL extract could potentially reduce the risk of NAFLD development, and that the SL extract may be clinically useful for the treatment of NAFLD.
Collapse
Affiliation(s)
- Hisako Kubo
- Human Health Sciences, Graduate School of Medicine and Faculty of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masato Hoshi
- Department of Biochemical and Analytical Sciences, Fujita Health University Graduate School of Health Sciences, 1-98 Dengakugakubo, Kutsukakecho, Toyoake, Aichi, 470-1192, Japan
| | - Takuya Matsumoto
- Human Health Sciences, Graduate School of Medicine and Faculty of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Motoko Irie
- Research Institute, Gekkeikan Sake Co. Ltd., 247 Minamihamcho, Fushimi, Kyoto, 612-8385, Japan
| | - Shin Oura
- Research Institute, Gekkeikan Sake Co. Ltd., 247 Minamihamcho, Fushimi, Kyoto, 612-8385, Japan
| | - Hiroko Tsutsumi
- Research Institute, Gekkeikan Sake Co. Ltd., 247 Minamihamcho, Fushimi, Kyoto, 612-8385, Japan
| | - Yoji Hata
- Research Institute, Gekkeikan Sake Co. Ltd., 247 Minamihamcho, Fushimi, Kyoto, 612-8385, Japan
| | - Yasuko Yamamoto
- Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Sciences, 1-98 Dengakugakubo, Kutsukakecho, Toyoake, Aichi, 470-1192, Japan
| | - Kuniaki Saito
- Human Health Sciences, Graduate School of Medicine and Faculty of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan. .,Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Sciences, 1-98 Dengakugakubo, Kutsukakecho, Toyoake, Aichi, 470-1192, Japan.
| |
Collapse
|
45
|
Liu W, Mi S, Ruan Z, Li J, Shu X, Yao K, Jiang M, Deng Z. Dietary Tryptophan Enhanced the Expression of Tight Junction Protein ZO-1 in Intestine. J Food Sci 2017; 82:562-567. [PMID: 28125771 DOI: 10.1111/1750-3841.13603] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 11/06/2016] [Accepted: 12/01/2016] [Indexed: 02/06/2023]
Abstract
Dietary amino acids provide various beneficial effects for our health. The aim of the present study was to assess the effects of tryptophan (Trp) supplementation on barrier function. Ninety-six healthy finishing pigs (initial body weight 51.49 ± 1.12 kg) were randomly allocated into 2 treatment groups, control group, and 0.2% Trp group. The control group was fed the basal diet, and 0.2% Trp group was fed basal diet plus 0.2% Trp. The trial period is 60 d. Compared with control group, the mRNA abundance of claudin-3 and zonula occluden-1 (ZO-1) in the jejunum in 0.2% Trp group (P < 0.05) was increased. According to immunohistochemistry and immunoblotting test, the expression of ZO-1 in jejunum in 0.2% Trp group was also significantly increased compared with the control group (P < 0.05). These results revealed that Trp enhanced the expression of tight junction protein ZO-1 in the intestine of pig model. Trp may be potential and beneficial dietary functional factor for regulating the intestinal development and inhibiting intestinal aging.
Collapse
Affiliation(s)
- Wenhui Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang Univ., Nanchang, 330047, China
| | - Shumei Mi
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang Univ., Nanchang, 330047, China
| | - Zheng Ruan
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang Univ., Nanchang, 330047, China
| | - Jing Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang Univ., Nanchang, 330047, China
| | - Xugang Shu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang Univ., Nanchang, 330047, China
| | - Kang Yao
- Inst. of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Min Jiang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang Univ., Nanchang, 330047, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Nanchang Univ., Nanchang, 330047, China
| |
Collapse
|
46
|
Ma S, Li T, Guo K, Li X, An S, Hou S, Chen R, Yang B, Liu S, Fu J. Effective treatment with combination of peripheral 5-hydroxytryptamine synthetic inhibitor and 5-hydroxytryptamine 2 receptor antagonist on glucocorticoid-induced whole-body insulin resistance with hyperglycemia. J Diabetes Investig 2016; 7:833-844. [PMID: 27177506 PMCID: PMC5089945 DOI: 10.1111/jdi.12526] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/24/2016] [Accepted: 03/16/2016] [Indexed: 12/12/2022] Open
Abstract
AIMS/INTRODUCTION Our previous study found that dexamethasone-induced insulin resistance (IR) was involved in 5-hydroxytryptamine (5-HT) synthesis and 5-hydroxytryptamine 2 receptor (5-HT2 R) in the periphery. The present study examined the effects of inhibitions of both peripheral 5-HT synthesis and 5-HT2 R on dexamethasone-induced IR. MATERIALS AND METHODS Male rats were exposed to dexamethasone for 10 days, then treated with or without a 5-HT2 R antagonist, sarpogrelate, a 5-HT synthetic inhibitor, carbidopa, alone or in combination for 20 days. RESULTS Dexamethasone-induced whole-body IR, with glucose intolerance, decreased insulin sensitivity, hyperglycemia, hyperinsulinemia and dyslipidemia, could be effectively abolished by sarpogrelate or/and carbidopa, whereas IR-related actions of dexamethasone in tissues were accompanied by increased 5-HT synthesis in the liver and visceral adipose, and upregulated 5-HT2 R (5-HT2A R and 5-HT2B R) expression in these two tissues as well as in skeletal muscle. Sarpogrelate or/and carbidopa treatment significantly abolished dexamethasone-caused tissue-specific IR. In the liver, increased gluconeogenesis, triglycerides and very low-density lipoprotein syntheses with steatosis, and downregulated expression of plasmalemmal glucose transporter-2 were markedly reversed. In the visceral adipose and skeletal muscle, downregulated expression of plasmalemmal glucose transporter-4 was significantly reversed, and increased lipolysis was also reversed in the visceral adipose. Dexamethasone-induced activations of hepatic mammalian target of rapamycin serine2448 , and S6K threonine389/412 phosphorylation were also abolished markedly by sarpogrelate or/and carbidopa. Co-treatment with sarpogrelate and carbidopa showed a synergistic effect on suppressing dexamethasone actions. CONCLUSION Inhibitions of both peripheral 5-HT synthesis and 5-HT2 R are expected to be a dependable target for treatment of steroid-induced diabetes.
Collapse
Affiliation(s)
- Shaoxin Ma
- Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Tao Li
- Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Keke Guo
- Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Xin Li
- Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Shanshan An
- Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Shanshan Hou
- Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Ru Chen
- Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Bo Yang
- Undergraduates of China Pharmaceutical University, Nanjing, China
| | - Siyu Liu
- Undergraduates of China Pharmaceutical University, Nanjing, China
| | - Jihua Fu
- Department of Physiology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
47
|
Li X, Guo K, Li T, Ma S, An S, Wang S, Di J, He S, Fu J. 5-HT 2 receptor mediates high-fat diet-induced hepatic steatosis and very low density lipoprotein overproduction in rats. Obes Res Clin Pract 2016; 12:16-28. [PMID: 27133527 DOI: 10.1016/j.orcp.2016.03.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/27/2016] [Accepted: 03/31/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND 5-HT has been shown to mediate abnormality of hepatic lipid metabolism through activation of mammalian target of rapamycin (mTOR). However, it is unclear whether 5-HT is directly involved in high-fat diet (HFD)-induced hepatic steatosis. MATERIALS AND METHODS Male rats were allocated into seven groups with control, either HFD feeding, 5-HT treatment, or HFD feeding and 5-HT treatment with or without sarpogrelate treatment, all of which were executed for 4 weeks. HepG2 cells were exposed to 5-HT or palmitic acid (PA) with or without rapamycin or Sar treatment. RESULTS Rats fed with HFD or exposed to 5-HT led to abnormalities with activated hepatic mTOR-S6K pathway, overproduction of hepatic triglycerides and VLDL with steatosis, and hyperlipidemia, which were exacerbated by a combination of HFD and 5-HT. Sarpogrelate significantly inhibited above abnormalities induced by HFD and 5-HT, alone or in a combination. Additionally, HFD caused up-regulation of 5-HT2 receptors (5-HT2R), including 5-HT2AR and 5-HT2BR, and 5-HT synthesis in the liver, without obvious influence on other 5-HT receptors gene expression. In HepG2 cells, both PA and 5-HT induced overproduction of triglycerides and VLDL with lipid droplets, and PA up-regulated 5-HT2AR and 5-HT2BR expression and 5-HT synthesis as well. Rapamycin fully abolished PA or 5-HT-induced mTOR activation, which was more effective than sarpogrelate. However, the inhibitory effects of rapamycin on PA or 5-HT-induced overproduction of triglycerides and VLDL were less than sarpogrelate. CONCLUSIONS Up-regulation of hepatic 5-HT2R and 5-HT synthesis by HFD is crucial for HFD-induced overproduction of hepatic triglycerides and VLDL with hyperlipidemia.
Collapse
Affiliation(s)
- Xin Li
- Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Keke Guo
- Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Tao Li
- Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Shaoxin Ma
- Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Shanshan An
- Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Shanshan Wang
- Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Jiao Di
- Undergraduates of China Pharmaceutical University, Nanjing, China
| | - Siyu He
- Undergraduates of China Pharmaceutical University, Nanjing, China
| | - Jihua Fu
- Department of Physiology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
48
|
Wang B, Min Z, Yuan J. Apparent ileal digestible tryptophan requirements of 22- to 42-day-old broiler chicks. J APPL POULTRY RES 2016. [DOI: 10.3382/japr/pfv061] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
49
|
Ma X, Chi YH, Niu M, Zhu Y, Zhao YL, Chen Z, Wang JB, Zhang CE, Li JY, Wang LF, Gong M, Wei SZ, Chen C, Zhang L, Wu MQ, Xiao XH. Metabolomics Coupled with Multivariate Data and Pathway Analysis on Potential Biomarkers in Cholestasis and Intervention Effect of Paeonia lactiflora Pall. Front Pharmacol 2016; 7:14. [PMID: 26869930 PMCID: PMC4740759 DOI: 10.3389/fphar.2016.00014] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/13/2016] [Indexed: 12/15/2022] Open
Abstract
Background: The dried root of Paeonia lactiflora Pall. (PLP) is a classical Chinese herbal medicine that has been used to treat hepatic disease for 1000s of years. Our previous work suggested that PLP can be used to treat hepatitis with severe cholestasis. This study explored the mechanism by which PLP affects ANIT-induced cholestasis in rats using a metabolomics approach. Methods: The effects of PLP on serum indices (TBIL, DBIL, AST, ALT, ALP, and TBA) and the histopathology of the liver were analyzed. Moreover, UHPLC-Q-TOF was performed to identify the possible effect of PLP on metabolites. The pathway analysis was conducted to illustrate the pathways and network by which PLP treats cholestasis. Result: High-dose PLP remarkably down-regulated the serum indices and alleviated histological damage to the liver. Metabolomics analyses showed that the therapeutic effect of high-dose PLP is mainly associated with the regulation of several metabolites, such as glycocholic acid, taurocholic acid, glycochenodeoxycholic acid, L(D)-arginine, and L-tryptophan. A pathway analysis showed that the metabolites were related to bile acid secretion and amino acid metabolism. In addition, the significant changes in bile acid transporters also indicated that bile acid metabolism might be involved in the therapeutic effect of PLP on cholestasis. Moreover, a principal component analysis indicated that the metabolites in the high-dose PLP group were closer to those of the control, whereas those of the moderate dose or low-dose PLP group were closer to those of the ANIT group. This finding indicated that metabolites may be responsible for the differences between the effects of low-dose and moderate-dose PLP. Conclusion: The therapeutic effect of high-dose PLP on cholestasis is possibly related to regulation of bile acid secretion and amino acid metabolism. Moreover, these findings may help better understand the mechanisms of disease and provide a potential therapy for cholestasis.
Collapse
Affiliation(s)
- Xiao Ma
- Department of Pharmacy, 302 Military Hospital of ChinaBeijing, China; Pharmacy College, Chengdu University of Traditional Chinese MedicineChengdu, China
| | - Yong-Hui Chi
- Cardiology Department, Beijing Chao-Yang Hospital, Capital Medical University Beijing, China
| | - Ming Niu
- China Military Institute of Chinese Medicine, 302 Military Hospital of China Beijing, China
| | - Yun Zhu
- Department of Integrative Medical Center, 302 Military Hospital of China Beijing, China
| | - Yan-Ling Zhao
- Department of Pharmacy, 302 Military Hospital of China Beijing, China
| | - Zhe Chen
- Department of Pharmacy, 302 Military Hospital of ChinaBeijing, China; Pharmacy College, Chengdu University of Traditional Chinese MedicineChengdu, China
| | - Jia-Bo Wang
- China Military Institute of Chinese Medicine, 302 Military Hospital of China Beijing, China
| | - Cong-En Zhang
- Pharmacy College, Chengdu University of Traditional Chinese MedicineChengdu, China; China Military Institute of Chinese Medicine, 302 Military Hospital of ChinaBeijing, China
| | - Jian-Yu Li
- Department of Integrative Medical Center, 302 Military Hospital of China Beijing, China
| | - Li-Fu Wang
- Department of Integrative Medical Center, 302 Military Hospital of China Beijing, China
| | - Man Gong
- Department of Integrative Medical Center, 302 Military Hospital of China Beijing, China
| | - Shi-Zhang Wei
- Department of Pharmacy, 302 Military Hospital of ChinaBeijing, China; Pharmacy College, Chengdu University of Traditional Chinese MedicineChengdu, China
| | - Chang Chen
- Department of Pharmacy, 302 Military Hospital of ChinaBeijing, China; Pharmacy College, Chengdu University of Traditional Chinese MedicineChengdu, China
| | - Lu Zhang
- Department of Pharmacy, 302 Military Hospital of ChinaBeijing, China; Pharmacy College, Chengdu University of Traditional Chinese MedicineChengdu, China
| | - Ming-Quan Wu
- Department of Pharmacy, 302 Military Hospital of ChinaBeijing, China; Pharmacy College, Chengdu University of Traditional Chinese MedicineChengdu, China
| | - Xiao-He Xiao
- China Military Institute of Chinese Medicine, 302 Military Hospital of China Beijing, China
| |
Collapse
|
50
|
Fu J, Ma S, Li X, An S, Li T, Guo K, Lin M, Qu W, Wang S, Dong X, Han X, Fu T, Huang X, Wang T, He S. Long-term Stress with Hyperglucocorticoidemia-induced Hepatic Steatosis with VLDL Overproduction Is Dependent on both 5-HT2 Receptor and 5-HT Synthesis in Liver. Int J Biol Sci 2016; 12:219-34. [PMID: 26884719 PMCID: PMC4737678 DOI: 10.7150/ijbs.13062] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 10/23/2015] [Indexed: 01/09/2023] Open
Abstract
Hepatic triglycerides production and adipose lipolysis are pivotal for long-term stress (LTS) or hyperglucocorticoidemia-induced insulin resistance. 5-hydroxytryptamine (5-HT) has been demonstrated to induce hepatic lipid metabolic abnormality by activating mammalian target of rapamycin (mTOR). In present study, we explored whether 5-HT is involved in LTS effects in liver using restraint stress-exposed rats and cultured primary rat hepatocytes and HepG2 cells. LTS with hyperglucocorticoidemia induced hepatic 5-HT synthetic increase with tryptophan hydroxylase 1 (Tph1) up-regulation, and 5-HT2 receptor (5-HT2R, including 5-HT2A, 2B receptor) up-regulation in liver and visceral adipose, as well as hepatic mTOR activation with triglycerides and VLDL overproduction with steatosis, and visceral adipose lipolytic increase with high blood free fatty acids (FFAs) level. 5-HT exposure exhibited LTS-like effects in both tissues, and both LTS and 5-HT effects could be abolished significantly by blocking 5-HT2R. In HepG2 cells dexamethasone or palmitate-induced mTOR activation with triglycerides and VLDL overproduction were accompanied by up-regulations of 5-HT synthesis and 5-HT2R, which were significantly abolished by gene silencing Tph1 or 5-HT2R and were almost fully abolished by co-silencing of both, especially on VLDL overproduction. Chemical inhibition of Tph1 or/and 5-HT2R in both hepatocytes exhibited similar abolishment with genetic inhibition on dexamethason-induced effects. 5-HT-stimulated effects in both hepatocytes were fully abolished by blocking 5-HT2R, while 5-HT itself also up-regulated 5-HT2R. In conclusion, up-regulated hepatic 5-HT synthesis and 5-HT2R induced by both glucocorticoid and FFAs are crucial for LTS-induced hepatic steatosis with VLDL overproduction, while 5-HT by acting on 5-HT2R mediates mTOR activation in liver.
Collapse
Affiliation(s)
- Jihua Fu
- 1. Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Shaoxin Ma
- 1. Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Xin Li
- 1. Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Shanshan An
- 1. Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Tao Li
- 1. Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Keke Guo
- 1. Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Min Lin
- 1. Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Wei Qu
- 1. Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Shanshan Wang
- 1. Postgraduates of China Pharmaceutical University, Nanjing, China
| | - Xinyue Dong
- 2. Undergraduates of China Pharmaceutical University, Nanjing, China
| | - Xiaoyu Han
- 2. Undergraduates of China Pharmaceutical University, Nanjing, China
| | - Ting Fu
- 2. Undergraduates of China Pharmaceutical University, Nanjing, China
| | - Xinping Huang
- 2. Undergraduates of China Pharmaceutical University, Nanjing, China
| | - Tianying Wang
- 2. Undergraduates of China Pharmaceutical University, Nanjing, China
| | - Siyu He
- 2. Undergraduates of China Pharmaceutical University, Nanjing, China
| |
Collapse
|