1
|
Angendohr C, Koppe C, Herebian D, Schneider AT, Keysberg L, Singer MT, Gilljam J, Dille MA, Bode JG, Doll S, Conrad M, Vucur M, Luedde T. The ferroptosis mediator ACSL4 fails to prevent disease progression in mouse models of MASLD. Hepatol Commun 2025; 9:e0684. [PMID: 40377498 PMCID: PMC12088639 DOI: 10.1097/hc9.0000000000000684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/30/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is an increasingly prevalent condition and a major risk factor for chronic liver damage, potentially leading to steatohepatitis and HCC. It is already known that patients with MASLD show increased systemic and hepatic iron concentrations as well as perturbed lipid metabolism, suggesting the involvement of ferroptosis in the development and progression of MASLD. Consequently, inhibition of ferroptosis represents a potential therapeutic option for patients with MASLD. METHODS We investigated whether liver parenchymal cell-specific deletion (LPC-KO) of the pro-ferroptotic gene acyl-CoA synthetase long-chain family member 4 (ACSL4LPC-KO) reduces MASLD onset and progression in mice. ACSL4LPC-KO and wild-type littermates were fed a choline-deficient high-fat diet (CD-HFD) or a Western diet for 20 weeks (CD-HFD and Western diet) or 40 weeks (CD-HFD only) to monitor MASLD progression and metabolic syndrome development. RESULTS In contrast to the recently published studies by Duan et al, our results show no significant differences between ACSL4LPC-KO and wild-type mice with regard to the development of MASLD or the progression of metabolic syndrome. Furthermore, no differences were observed in metabolic parameters (ie, weight gain, glucose tolerance test, hepatic steatosis) or MASLD-associated inflammatory response. CONCLUSIONS Our analyses, therefore, suggest that loss of ACSL4 has no effect on the progression of MASLD induced by CD-HFD or the Western diet. The discrepancy between our and previously published results could be due to differences in the diets or the influence of a distinct microbiome, so the results obtained with hepatocyte-specific ACSL4LPC-KO should be taken with caution.
Collapse
Affiliation(s)
- Carolin Angendohr
- Department of Gastroenterology, Hepatology and Infectious Diseases, Faculty of Medicine & Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Christiane Koppe
- Department of Gastroenterology, Hepatology and Infectious Diseases, Faculty of Medicine & Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Diran Herebian
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital, Heinrich Heine University, Düsseldorf, Germany
| | - Anne T. Schneider
- Department of Gastroenterology, Hepatology and Infectious Diseases, Faculty of Medicine & Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Leonie Keysberg
- Department of Gastroenterology, Hepatology and Infectious Diseases, Faculty of Medicine & Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Michael T. Singer
- Department of Gastroenterology, Hepatology and Infectious Diseases, Faculty of Medicine & Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Julian Gilljam
- Department of Gastroenterology, Hepatology and Infectious Diseases, Faculty of Medicine & Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Matthias A. Dille
- Department of Gastroenterology, Hepatology and Infectious Diseases, Faculty of Medicine & Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Johannes G. Bode
- Department of Gastroenterology, Hepatology and Infectious Diseases, Faculty of Medicine & Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sebastian Doll
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Mihael Vucur
- Department of Gastroenterology, Hepatology and Infectious Diseases, Faculty of Medicine & Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, Faculty of Medicine & Düsseldorf University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
2
|
Moradzad M, Ghaderi D, Abdi M, Sheikh Esmaili F, Rahmani K, Vahabzadeh Z. Gut microbiota dysbiosis contributes to choline unavailability and NAFLD development. J Diabetes Metab Disord 2025; 24:37. [PMID: 39801684 PMCID: PMC11711859 DOI: 10.1007/s40200-024-01511-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025]
Abstract
OBJECTIVES Non-alcoholic fatty Liver Disease (NAFLD) poses a growing global health concern, yet its complex aetiology remains incompletely understood. Emerging evidence implicates the gut microbiome and choline metabolism in NAFLD pathogenesis. This study aims to elucidate the association of choline-consuming bacteria in gut microbiome with choline level. METHODS A population comprising 85 NAFLD patients and 30 healthy controls was selected. DNA extraction from stool samples was conducted using the FavorPrep™ Stool DNA Isolation Mini Kit, followed by polymerase chain reaction (PCR) detection of choline-consuming bacterial strains and quantitative PCR (qPCR) for Cut C gene expression. Choline content measurement was performed using fluorescence high-performance liquid chromatography (FL-HPLC). RESULTS Our findings revealed a significant reduction in choline levels among NAFLD patients compared to healthy controls. ROC curve analysis demonstrated choline levels and Cut C expression as a promising diagnostic tool for NAFLD, with high sensitivity and specificity. The microbial analysis identified specific choline-consuming bacteria enriched in NAFLD patients, notably Anarococcus Hydrogenalis and Clostridium asparagiforme. This was consistent with higher Cut C gene expression in patients compared to healthy individuals, which is responsible for encoding an enzyme to consume choline by these bacteria. CONCLUSION The current study gives a possible association between gut microbiota and the development of NAFLD, possibly due to an alteration in choline bioavailability. Further research is required to determine whether gut bacteria alter in the context of NAFLD or a change in their composition might lead to NAFLD progression, possibly via alternation in choline bioavailability. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s40200-024-01511-6.
Collapse
Affiliation(s)
- Mohammad Moradzad
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Dana Ghaderi
- Department of Clinical Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohammad Abdi
- Department of Clinical Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Farshad Sheikh Esmaili
- Liver & Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Khaled Rahmani
- Liver & Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Zakaria Vahabzadeh
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
3
|
Buchynskyi M, Kamyshna I, Halabitska I, Petakh P, Kunduzova O, Oksenych V, Kamyshnyi O. Unlocking the gut-liver axis: microbial contributions to the pathogenesis of metabolic-associated fatty liver disease. Front Microbiol 2025; 16:1577724. [PMID: 40351307 PMCID: PMC12061941 DOI: 10.3389/fmicb.2025.1577724] [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: 02/16/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a complex metabolic disorder characterized by hepatic lipid accumulation and subsequent inflammation. This condition is closely linked to metabolic syndrome and obesity, with its prevalence rising due to sedentary lifestyles and high-calorie diets. The pathogenesis of MAFLD involves multiple factors, including insulin resistance, lipotoxicity, oxidative stress, and inflammatory responses. The gut microbiota plays a crucial role in MAFLD development, with dysbiosis contributing to liver inflammation through various mechanisms, such as enhanced intestinal permeability and the translocation of bacterial products like lipopolysaccharide (LPS). Microbial metabolites, including short-chain fatty acids (SCFAs) and bile acids, influence hepatic function and immune responses, with potential implications for disease progression. Specific gut microbiome signatures have been identified in MAFLD patients, offering potential diagnostic and therapeutic targets. Moreover, gut-derived toxins, such as endotoxins, lipopolysaccharides, trimethylamine-N-oxide and bacterial metabolites, significantly influence liver damage and inflammation, highlighting the complex interplay between the gut microbiome and hepatic health. This review comprehensively examines the complex interplay between the gut microbiota and MAFLD, focusing on underlying pathogenic mechanisms, potential biomarkers, and emerging microbiome-targeted therapeutic strategies for disease management.
Collapse
Affiliation(s)
- Mykhailo Buchynskyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Halabitska
- Department of Therapy and Family Medicine, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Pavlo Petakh
- Department of Biochemistry and Pharmacology, Uzhhorod National University, Uzhhorod, Ukraine
| | - Oksana Kunduzova
- Institute of Metabolic and Cardiovascular Diseases (I2MC), National Institute of Health and Medical Research (INSERM) 1297, Toulouse III University, Toulouse, France
| | - Valentyn Oksenych
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| |
Collapse
|
4
|
Basha EH, Hegab II, Ismail R, Atef MM, El-Deeb OS, Ibrahim RR, Ghanem HB, Eissa R, Taha MS, Mwafy SE, Rizk FH, Salem OM, Ghafar MTA, Hafez YM, Mashal S, Tabaa MME, El-Harty YM. Protective effects of Kaempferol on hepatic apoptosis via miR-26a-5p enhancement and regulation of TLR4/NF-κB and PKCδ in a rat model of nonalcoholic fatty liver. J Nutr Biochem 2025; 137:109833. [PMID: 39701472 DOI: 10.1016/j.jnutbio.2024.109833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 12/21/2024]
Abstract
This study aimed to evaluate kaempferol's, a dietary flavonoid widely present in plants, potential impact on nonalcoholic fatty liver disease (NAFLD) and its underlying mechanisms. In this study, 60 adult male rats were used and divided into a control group receiving a standard pellet diet, a kaempferol-treated group receiving kaempferol (250 mg/kg), a high-fat diet (HFD) group receiving HFD, and a kaempferol-treated HFD group. At the end of the experiment, the total lipid profile and liver enzymes were assayed in the serum. Additionally, oxidative stress (malondialdehyde and superoxide dismutase), inflammatory (tumor necrosis factor-alpha), apoptotic (caspase 3) markers, and nuclear factor-κB (NF-κB) and Toll-like receptor 4 (TLR4) concentrations were assayed in the liver tissues. Furthermore, miR-26a and PKCδ gene expression and beclin 1 immunohistochemical expression were determined in liver tissues. Our findings revealed that kaempferol significantly protects against the development of NAFLD in rats as well as inflammatory, oxidative, and apoptotic changes in their liver tissues by inhibiting PKCδ and the TLR-4/NF-κB signaling pathway while enhancing autophagy (Beclin 1 expression) via upregulating miR-26a expression. Accordingly, kaempferol holds promise as a complementary medication for the prevention of NAFLD. Nonetheless, more research is needed to fully understand its additional effects on liver tissue and to develop novel medications that activate miR-26a. A link between lipid metabolic abnormalities and miRNAs was demonstrated as upregulating miR-26a-5p by kaempferol mitigates the inflammation, apoptosis, and disrupted autophagy via regulating TLR4/NF-κB pathway and PKC in NAFLD.
Collapse
Affiliation(s)
- Eman H Basha
- Departments of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt; Department of Basic Medical Sciences, Physiology, Faculty of Medicine, Ibn Sina University for Medical Sciences, Amman 16197, Jordan
| | - Islam Ibrahim Hegab
- Departments of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt; Department of Physiology, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia
| | - Radwa Ismail
- Departments of Anatomy, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Marwa Mohamed Atef
- Departments of Medical biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Omnia Safwat El-Deeb
- Departments of Medical biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt.
| | - Rowida Rafaat Ibrahim
- Departments of Medical biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Heba Bassiony Ghanem
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia; Departments of Medical biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Radwa Eissa
- Department of Medical Microbiology & Immunology, Faculty of Medicine, Tanta University Tanta, Egypt
| | - Marwa S Taha
- Department of Medical Microbiology & Immunology, Faculty of Medicine, Tanta University Tanta, Egypt; Departments of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Shorouk E Mwafy
- Departemnt of Pathology, Faculty of Medicine, Tanta University Tanta, Egypt
| | - Fatma H Rizk
- Departments of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Ola M Salem
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | - Yasser Mostafa Hafez
- Department of Internal Medicine, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Shimaa Mashal
- Department of Internal Medicine, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Manar Mohammed El Tabaa
- Pharmacology & Environmental Toxicology, Environmental Studies & Research Institute (ESRI), University of Sadat City, Sadat, Menoufia, Egypt
| | - Yasmeen M El-Harty
- Departments of Physiology, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
5
|
Yao M, Xiao Y, Sun Y, Zhang B, Ding Y, Ma Q, Liang F, Yang Z, Ge W, Liu S, Xin L, Yin J, Zhu X. Association of maternal gut microbial metabolites with gestational diabetes mellitus: evidence from an original case-control study, meta-analysis, and Mendelian randomization. Eur J Clin Nutr 2025; 79:33-41. [PMID: 39223299 DOI: 10.1038/s41430-024-01502-z] [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: 12/05/2023] [Revised: 07/06/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The associations of gut microbial metabolites, such as trimethylamine N-oxide (TMAO), its precursors, and phenylacetylglutamine (PAGln), with the risk of gestational diabetes mellitus (GDM) remain unclear. METHODS Serum samples of 201 women with GDM and 201 matched controls were collected and then targeted metabolomics was performed to examine the metabolites of interest. Multivariable conditional logistic regression was applied to investigate the relationship between metabolites and GDM. Meta-analysis was performed to combine our results and four similar articles searched from online databases, and Mendelian randomization (MR) analysis was eventually conducted to explore the causalities. RESULTS In the case-control study, after dichotomization and comparing the higher versus the lower group, the adjusted odds ratio and 95% confidence interval of choline and L-carnitine with GDM were 2.124 (1.186-3.803) and 0.293 (0.134-0.638), respectively; but neutral relationships between TMAO, betaine, and PAGln with GDM were observed. The following meta-analysis consistently revealed that L-carnitine was negatively associated with GDM. However, MR analyses showed no evidence of causalities. CONCLUSIONS Maternal levels of L-carnitine were related to the risk of GDM in both the original case-control study and meta-analysis. However, we did not observe any genetic evidence to establish a causal relationship between this metabolite and GDM.
Collapse
Affiliation(s)
- Mengxin Yao
- Suzhou Center for Disease Prevention and Control, Suzhou, China
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China
| | - Yue Xiao
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China
| | - Yanqun Sun
- Nanjing Municipal Center for Disease Control and Prevention, Nanjing, China
| | - Bing Zhang
- Department of Geriatrics, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Yaling Ding
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China
| | - Qiuping Ma
- Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, 58 Changsheng Road, Suzhou, China
| | - Fei Liang
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China
| | - Zhuoqiao Yang
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China
| | - Wenxin Ge
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China
| | - Songliang Liu
- Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, 58 Changsheng Road, Suzhou, China
| | - Lili Xin
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, Medical College of Soochow University, Suzhou, China
| | - Jieyun Yin
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China.
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, Medical College of Soochow University, Suzhou, China.
| | - Xiaoyan Zhu
- Suzhou Center for Disease Prevention and Control, Suzhou, China.
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
6
|
Feng L, He B, Xia J, Wang Z. Untargeted and Targeted Lipidomics Unveil Dynamic Lipid Metabolism Alterations in Type 2 Diabetes. Metabolites 2024; 14:610. [PMID: 39590846 PMCID: PMC11596168 DOI: 10.3390/metabo14110610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a complex metabolic disorder with a growing body of evidence suggesting the central role of lipid metabolism in its pathogenesis. However, the dynamic changes in lipid metabolism across different stages of T2DM remain understudied. OBJECTIVE This study aimed to elucidate the temporal alterations in lipid metabolism in T2DM using an integrated lipidomics approach. METHOD Serum samples from 155 subjects were analyzed using LC-MS-based lipidomics, including untargeted and targeted approaches. RESULTS We identified significant alterations in 44 lipid metabolites in newly diagnosed T2DM patients and 29 in high-risk individuals, compared with healthy controls. Key metabolic pathways such as sphingomyelin, phosphatidylcholine, and sterol ester metabolism were disrupted, highlighting the involvement of insulin resistance and oxidative stress in T2DM progression. Moreover, 13 lipid metabolites exhibited diagnostic potential for T2DN, showing consistent trends of increase or decrease as the disease progressed. CONCLUSION Our findings underscore the importance of lipid metabolism in T2D development and identify potential lipid biomarkers for early diagnosis and monitoring of disease progression, which contribute to paving the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Li Feng
- School of Agroforestry and Medicine, The Open University of China, Beijing 100039, China;
| | - Bingshu He
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China;
| | - Jianzhen Xia
- School Hospital, Minzu University of China, Beijing 100081, China;
| | - Zhonghua Wang
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China;
| |
Collapse
|
7
|
Xia J, Yin S, Yu J, Wang J, Jin X, Wang Y, Liu H, Sun G. Improvement in Glycolipid Metabolism Parameters After Supplementing Fish Oil-Derived Omega-3 Fatty Acids Is Associated with Gut Microbiota and Lipid Metabolites in Type 2 Diabetes Mellitus. Nutrients 2024; 16:3755. [PMID: 39519588 PMCID: PMC11547733 DOI: 10.3390/nu16213755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES This study aimed to investigate the effects of fish oil-derived omega-3 polyunsaturated fatty acids (omega-3 PUFAs) on gut microbiota and serum lipid metabolites in T2DM. METHODS In a three-month, randomized, double-blind, placebo-controlled study, 110 T2DM patients received either fish oil (n = 55) or corn oil (n = 55) capsules daily. Serum lipids, glycemic parameters, gut microbiota diversity, and lipidomics were assessed. RESULTS This study found that fish oil-derived omega-3 PUFAs intervention did not significantly lower the fasting plasma glucose levels when compared with the baseline level (p > 0.05). However, serum fasting blood glucose (p = 0.039), glycosylated hemoglobin levels (p = 0.048), HOMA-IR (p = 0.022), total cholesterol (p < 0.001), triglyceride (p = 0.034), LDL cholesterol (p = 0.048), and non-HDL levels (p = 0.046) were significantly lower in the fish oil group compared with the corn oil group after three months of intervention. Also, it altered glycerophospholipid metabolism and gut microbiota. After three months, the fish oil group showed a significantly lower abundance of Desulfobacterota compared with the corn oil control group (p = 0.003), with reduced levels of Colidextribacter (p = 0.002), Ralstonia (p = 0.021), and Klebsiella (p = 0.013). Conversely, the abundance of Limosilactobacillus (p = 0.017), Lactobacillus (p = 0.011), and Haemophilus (p = 0.018) increased significantly. In addition, relevant glycolipid metabolism indicators showed significant correlations with the altered profiles of serum lipid metabolites, intestinal bacteria, and fungi. CONCLUSIONS This study highlights the impact of fish oil-derived omega-3 PUFAs on intestinal microbiota structure and function in patients with type 2 diabetes. The observed decrease in pathogenic bacterial species and the enhancement of beneficial species may have significant implications for gut health and systemic inflammation, both of which are pivotal in managing diabetes. Further research is warranted to comprehensively elucidate the long-term benefits and underlying mechanisms of these microbiota alterations.
Collapse
Affiliation(s)
- Jiayue Xia
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Shiyu Yin
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Junhui Yu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Jiongnan Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Xingyi Jin
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Yuanyuan Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Hechun Liu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Guiju Sun
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; (J.X.); (S.Y.); (J.Y.); (J.W.); (X.J.); (Y.W.)
- Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| |
Collapse
|
8
|
Yuan K, Lai K, Miao G, Zhang J, Zhao X, Tan G, Wang X, Wang X. Cholinized-Polymer Functionalized Lipid-Based Drug Carriers Facilitate Liver Fibrosis Therapy via Ultrafast Liver-Targeting Delivery. Biomacromolecules 2024; 25:6526-6538. [PMID: 39213520 DOI: 10.1021/acs.biomac.4c00691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Here, we report novel cholinized-polymer functionalized lipid-based nanoparticles (CP-LNPs) for rapid and highly effective delivery of drugs to the liver, achieving targeting within 10 min and nearly 100% efficiency. In this study, CP-LNPs loaded with a promising antifibrotic agent curcumin (CP-LNPs/Cur) significantly improved the stability of curcumin under physiological conditions and its distribution in the liver. In vitro experiments demonstrated that CP-LNPs/Cur effectively suppressed the proliferation and migration of activated hepatic stellate cells (aHSCs), as evidenced by the decreased expression of α-SMA. Moreover, CP-LNPs/Cur attenuated oxidative stress levels in hepatocytes while improving mitochondrial physiological activity. In vivo antifibrosis studies have shown that CP-LNPs/Cur only require a low dose to significantly alleviate liver injury and collagen deposition, thereby preventing the progression of liver fibrosis. These findings indicated that CP-LNPs exhibit great potential in liver fibrosis therapy benefiting from the novel targeting strategy.
Collapse
Affiliation(s)
- Kun Yuan
- Guangdong Provincial Key Laboratory of Constructionand Detection in Tissue Engineering, Biomaterials Research Center, School ofBiomedical Engineering, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Keren Lai
- Guangdong Provincial Key Laboratory of Constructionand Detection in Tissue Engineering, Biomaterials Research Center, School ofBiomedical Engineering, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Guifeng Miao
- Guangdong Provincial Key Laboratory of Constructionand Detection in Tissue Engineering, Biomaterials Research Center, School ofBiomedical Engineering, Southern Medical University, Guangzhou, Guangdong Province 510515, China
- Department of Cardiovascular Surgery, ZhujiangHospital, Southern Medical University, Guangzhou, Guangdong Province 510280, China
| | - Jibin Zhang
- Guangdong Provincial Key Laboratory of Constructionand Detection in Tissue Engineering, Biomaterials Research Center, School ofBiomedical Engineering, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Xiaoxi Zhao
- Guangdong Provincial Key Laboratory of Constructionand Detection in Tissue Engineering, Biomaterials Research Center, School ofBiomedical Engineering, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Guozhu Tan
- Guangdong Provincial Key Laboratory of Constructionand Detection in Tissue Engineering, Biomaterials Research Center, School ofBiomedical Engineering, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Xiaowu Wang
- Department of Cardiovascular Surgery, ZhujiangHospital, Southern Medical University, Guangzhou, Guangdong Province 510280, China
| | - Xiaorui Wang
- Guangdong Provincial Key Laboratory of Constructionand Detection in Tissue Engineering, Biomaterials Research Center, School ofBiomedical Engineering, Southern Medical University, Guangzhou, Guangdong Province 510515, China
- Department of Cardiovascular Surgery, ZhujiangHospital, Southern Medical University, Guangzhou, Guangdong Province 510280, China
| |
Collapse
|
9
|
Wang P, Zhang C, Pan D, Xia H, Wang Y, Sun J, Jiang T, Sun G, Huang J. The effects of alfalfa powder combined with health education on patients with dyslipidemia: A randomized controlled trial. J Funct Foods 2024; 121:106445. [DOI: 10.1016/j.jff.2024.106445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
|
10
|
Ou X, Chen J, Li B, Yang Y, Liu X, Xu Z, Xiang X, Wang Q. Multiomics reveals the ameliorating effect and underlying mechanism of aqueous extracts of polygonatum sibiricum rhizome on obesity and liver fat accumulation in high-fat diet-fed mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155843. [PMID: 38971026 DOI: 10.1016/j.phymed.2024.155843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 05/21/2024] [Accepted: 06/24/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND Polygonatum sibiricum polysaccharides protect against obesity and NAFLD. However, the potential effects of PS rhizome aqueous extracts (PSRwe) on adiposity and hepatic lipid accumulation remains unexplored. PURPOSE Elucidating the impact and underlying mechanism of PSRwe on HFD-induced obesity and liver fat depostition. STUDY DESIGN 56 male mice, aged eight weeks, were divided into seven groups: Positive, four doses of PSRwe, Model, and Control. HFD was fed for eight weeks, followed by alternate-day gavage of orlistat and PSRwe for an additional eight-week period. Integrative analysis encompassing multiomics, physiological and histopathological, and biochemical indexes was employed. METHODS Body weight (BW); liver, fat and Lee's indexes; TC, TG, LDL-C, HDL-C, AST, ALT, FFA, leptin, and adiponectin in the liver and blood; TNFα, IL-6, and LPS in the colon, plasma, and liver; H&E, PAS and oil red O staining on adipose and liver samples were examined. OGTT and ITT were conducted The gut microbiome, microbial metabolome, colonic and liver transcriptome, plasma and liver metabolites were investigated. RESULTS PSRwe at the dosage of 7.5 mg/kg demonstrated significant and consistent reduction in BW and hepatic fat deposition than orlistat. PSRwe significantly decreased TC, TG, LDL-C, LEP, FFA levels in blood and liver. PSRwe significantly enhanced the relative abundance of probiotics including Akkermansia muciniphila, Bifidobacterium pseudolongum, Lactobacillus reuteri, and metabolic pathways including glycolysis and fatty acids β-oxidation. The 70 up-regulated microbial metabolites in PSRwe-treated mice mainly involved in nucleotides and amino acids metabolism, while 40 decreased metabolites primarily associated with lipid metabolism. The up-regulated colonic differentially expressed genes (DEGs) participate in JAK-STAT/PI3K-Akt/FoxO signaling pathway, serotonergic/cholinergic/glutamatergic synapses, while the down-regulated DEGs predominantly focused on fat absorption and transport. The up-regulated liver DEGs mainly concentrated on fatty acid oxidation and metabolism. Liver metabolisms revealed 131 differential metabolites, among which carnitine and oxidized lipids significantly increased in PSRwe-treated mice. In plasma, the 58 up-regulated metabolites mainly participate in co-factors/vitamins metabolism while 154 down-regulated ones in fatty acids biosynthesis. Comprehensive multiomics association analysis revealed significant associations between gut microbiota and colonic/liver gene expression, and suggested exogenous and endogenous betaine may be active compound in alleviating HFD-induced symptoms. CONCLUSION PSRwe effectively mitigate HFD-induced obesity and hepatic steatosis by increasing beneficial bacteria, reducing colonic fat digestion/absorption, increasing hepatic lipid metabolism, and elevating betaine levels.
Collapse
Affiliation(s)
- Xiaobin Ou
- Gansu Key Laboratory of Protection and Utlization for Biological Resources and Ecological Restoration in Longdong, Longdong University, Qingyang 745000, Gansu, China; College of Life Sciences and Technology, Longdong University, Qingyang 745000, Gansu, China.
| | - Juanjuan Chen
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
| | - Boping Li
- Gansu Key Laboratory of Protection and Utlization for Biological Resources and Ecological Restoration in Longdong, Longdong University, Qingyang 745000, Gansu, China; College of Medicine, Longdong University, Qingyang 745000, Gansu, China
| | - Yan Yang
- Department of Endocrinology and Metabolism, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
| | - Xiuli Liu
- Gansu Key Laboratory of Protection and Utlization for Biological Resources and Ecological Restoration in Longdong, Longdong University, Qingyang 745000, Gansu, China; College of Life Sciences and Technology, Longdong University, Qingyang 745000, Gansu, China
| | - Zaoxu Xu
- Gansu Key Laboratory of Protection and Utlization for Biological Resources and Ecological Restoration in Longdong, Longdong University, Qingyang 745000, Gansu, China; College of Life Sciences and Technology, Longdong University, Qingyang 745000, Gansu, China
| | - Xuesong Xiang
- National Institute of Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Qi Wang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China.
| |
Collapse
|
11
|
Kang W, Xu X, Yang X, Wu Q, Li S, Gao K, Zeng R, Sun L, Lin X. Associations of Plasma Lipidomic Profiles with Uric Acid and Hyperuricemia Risk in Middle-Aged and Elderly Chinese. PHENOMICS (CHAM, SWITZERLAND) 2024; 4:352-364. [PMID: 39583309 PMCID: PMC11584823 DOI: 10.1007/s43657-024-00157-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/11/2024] [Accepted: 01/21/2024] [Indexed: 11/26/2024]
Abstract
Little is known about the links of disturbed lipid metabolism with hyperuricemia (HUA). We aimed to investigate the associations of lipidomic profiles with uric acid (UA)/HUA and their modifying factors in middle-aged and elderly Chinese. A total of 350 lipids were quantified in 2247 community-based Chinese aged 50-70 years by high-coverage targeted lipidomics. HUA was defined by plasma UA > 420 μmol/L in men or > 360 μmol/L in women. The prevalence of HUA in this population was 10.4%. After multivariable adjustment including BMI and lifestyle, 123 lipids were significantly associated with UA, predominantly glycerolipids (GLs) and glycerophospholipids (GPs). Specifically, diacylglycerol [DAG (16:0/22:5), DAG (16:0/22:6), DAG (18:1/20:5), DAG (18:1/22:6)], phosphatidylcholine [PC (16:0/20:5)), and triacylglycerol (TAG (53:0)] were the most significant lipid signatures positively associated with HUA risk, while lysophosphatidylcholine (LPC (20:2)) was inversely associated with HUA risk (p < 0.05). Network analysis also showed a positive association between TAGs/PCs/DAGs contained module and HUA risk (p < 0.01). Notably, HUA-related lipids were associated with de novo lipogenesis fatty acids, especially 16:1n-7 (Spearman correlation coefficients = 0.32-0.41, p < 0.001). Reduced rank regression showed that increased aquatic products intake was correlated to elevated HUA risk and HUA-associated lipids; while high dairy consumption was correlated with low level of HUA-associated lipids (|factor loadings| ≥ 0.2). Moreover, mediation analyses suggested that the lipid-HUA associations were partially mediated by retinol-binding protein 4 (RBP4, mediation proportion 5-14%), an adipokine linked with dyslipidemia and insulin resistance. In conclusion, disturbed specific metabolisms of GLs and GPs were associated with high prevalent HUA, partially mediated by RBP4 and/or influenced by certain dietary factors. Supplementary Information The online version contains supplementary material available at 10.1007/s43657-024-00157-x.
Collapse
Affiliation(s)
- Wanhui Kang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Xiangshanzhi Ln., Hangzhou, 310024 China
| | - Xinming Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Institute of Nutrition, Fudan University, 130 Dongan Rd., Shanghai, 200032 China
| | - Xiaowei Yang
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yue-Yang Rd., Shanghai, 200031 China
| | - Qingqing Wu
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 320 Yue‑Yang Rd., Shanghai, 200031 China
| | - Shuning Li
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Xiangshanzhi Ln., Hangzhou, 310024 China
| | - Keran Gao
- Schulich School of Medicine and Dentistry, Western University, 1465 Richmond St, London, ON N6G 2M1 Canada
| | - Rong Zeng
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Xiangshanzhi Ln., Hangzhou, 310024 China
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 320 Yue‑Yang Rd., Shanghai, 200031 China
| | - Liang Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Institute of Nutrition, Fudan University, 130 Dongan Rd., Shanghai, 200032 China
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yue-Yang Rd., Shanghai, 200031 China
| | - Xu Lin
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, 1 Xiangshanzhi Ln., Hangzhou, 310024 China
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yue-Yang Rd., Shanghai, 200031 China
| |
Collapse
|
12
|
Hense JD, Garcia DN, Zanini BM, Barreto MM, Perreira GC, Isola JVV, de Brito C, Fornalik M, Mondal SA, Ávila BM, Oliveira TL, Rice HC, Lacy CI, Vaucher RA, Mason JB, Masternak MM, Stout MB, Schneider A. MASLD does not affect fertility and senolytics fail to prevent MASLD progression in male mice. Sci Rep 2024; 14:17332. [PMID: 39068167 PMCID: PMC11283523 DOI: 10.1038/s41598-024-67697-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024] Open
Abstract
Senescent cells have been linked to the pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD). However, the effectiveness of senolytic drugs in reducing liver damage in mice with MASLD is not clear. Additionally, MASLD has been reported to adversely affect male reproductive function. Therefore, this study aimed to evaluate the protective effect of senolytic drugs on liver damage and fertility in male mice with MASLD. Three-month-old male mice were fed a standard diet (SD) or a choline-deficient western diet (WD) until 9 months of age. At 6 months of age mice were randomized within dietary treatment groups into senolytic (dasatinib + quercetin [D + Q]; fisetin [FIS]) or vehicle control treatment groups. We found that mice fed choline-deficient WD had liver damage characteristic of MASLD, with increased liver size, triglycerides accumulation, fibrosis, along increased liver cellular senescence and liver and systemic inflammation. Senolytics were not able to reduce liver damage, senescence and systemic inflammation, suggesting limited efficacy in controlling WD-induced liver damage. Sperm quality and fertility remained unchanged in mice developing MASLD or receiving senolytics. Our data suggest that liver damage and senescence in mice developing MASLD is not reversible by the use of senolytics. Additionally, neither MASLD nor senolytics affected fertility in male mice.
Collapse
Affiliation(s)
- Jessica D Hense
- Nutrition College, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 Sala 228, Pelotas, RS, CEP 9601-610, Brazil
| | - Driele N Garcia
- Nutrition College, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 Sala 228, Pelotas, RS, CEP 9601-610, Brazil
| | - Bianka M Zanini
- Nutrition College, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 Sala 228, Pelotas, RS, CEP 9601-610, Brazil
| | - Mariana M Barreto
- Nutrition College, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 Sala 228, Pelotas, RS, CEP 9601-610, Brazil
| | - Giulia C Perreira
- Nutrition College, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 Sala 228, Pelotas, RS, CEP 9601-610, Brazil
| | - José V V Isola
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
| | - Camila de Brito
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
| | - Michal Fornalik
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
| | - Samim A Mondal
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
- Department of Endocrinology, JIPMER, Puducherry, 605006, India
| | - Bianca M Ávila
- Nutrition College, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 Sala 228, Pelotas, RS, CEP 9601-610, Brazil
| | - Thais L Oliveira
- Biotechnology Center, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Heather C Rice
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
| | - Charles I Lacy
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
| | - Rodrigo A Vaucher
- Center for Chemical, Pharmaceutical and Food Sciences, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Jeffrey B Mason
- Department of Veterinary Clinical and Life Sciences, Center for Integrated BioSystems, College of Veterinary Medicine, Utah State University, Logan, UT, USA
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Michael B Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma, OK, USA
| | - Augusto Schneider
- Nutrition College, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1 Sala 228, Pelotas, RS, CEP 9601-610, Brazil.
| |
Collapse
|
13
|
Chen X, Zheng Z, Xie D, Xia L, Chen Y, Dong H, Feng Y. Serum lipid metabolism characteristics and potential biomarkers in patients with unilateral sudden sensorineural hearing loss. Lipids Health Dis 2024; 23:205. [PMID: 38951804 PMCID: PMC11218322 DOI: 10.1186/s12944-024-02189-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 06/16/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Glycerophospholipids (GPLs) are essential for cell membrane structure and function. Sphingomyelin and its metabolites regulate cell growth, apoptosis, and stress responses. This study aimed to investigate lipid metabolism in patients experiencing sudden sensorineural hearing loss across all frequencies (AF-SSNHL). METHODS The study included 60 patients diagnosed with unilateral AF-SSNHL, among whom 30 patients had a level of hearing improvement ≥ 15 dB after 6 months of follow-up. A propensity score-matched (2:1) control group was used. Liquid chromatography‒mass spectrometry based untargeted lipidomics analysis combined with multivariate statistics was performed to investigate the lipids change. The "lipidome" R package and weighted gene co-expression network analysis (WGCNA) were utilised to assess the lipids' structural features and the association between lipids and hearing. RESULTS Lipidomics successfully differentiated the AF-SSNHL group from the control group, identifying 17 risk factors, mainly including phosphatidylcholine (PC), phosphatidylethanolamine (PE), and related metabolites. The ratios of lysophosphatidylcholine/PC, lysophosphatidylethanolamine/PE, and lysodimethylphosphatidylethanolamine/PE were upregulated, while some glycerophospholipid (GPL)-plasmalogens were downregulated in the AF-SSNHL group, indicating abnormal metabolism of GPLs. Trihexosylceramide (d34:1), PE (18:1e_22:5), and sphingomyelin (d40:3) were significantly different between responders and nonresponders, and positively correlated with hearing improvement. Additionally, the results of the WGCNA also suggested that partial GPL-plasmalogens were positively associated with hearing improvement. CONCLUSION AF-SSNHL patients exhibited abnormally high blood lipids and pronounced GPLs metabolic abnormalities. Sphingolipids and GPL-plasmalogens had an association with the level of hearing improvement. By understanding the lipid changes, clinicians may be able to predict the prognosis of hearing recovery and personalize treatment approaches.
Collapse
Affiliation(s)
- Xiaoyan Chen
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Road, Xuhui District, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Zhong Zheng
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Road, Xuhui District, Shanghai, China
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Daoyu Xie
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | - Liang Xia
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Road, Xuhui District, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Yi Chen
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Road, Xuhui District, Shanghai, China
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Hongjun Dong
- Department of Otolaryngology-Head and Neck Surgery, Zhangjiagang TCM Hospital, Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu Province, China.
| | - Yanmei Feng
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, 600 Yishan Road, Xuhui District, Shanghai, China.
- Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China.
| |
Collapse
|
14
|
Min K, Yenilmez B, Kelly M, Echeverria D, Elleby M, Lifshitz LM, Raymond N, Tsagkaraki E, Harney SM, DiMarzio C, Wang H, McHugh N, Bramato B, Morrison B, Rothstein JD, Khvorova A, Czech MP. Lactate transporter MCT1 in hepatic stellate cells promotes fibrotic collagen expression in nonalcoholic steatohepatitis. eLife 2024; 12:RP89136. [PMID: 38564479 PMCID: PMC10987092 DOI: 10.7554/elife.89136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024] Open
Abstract
Circulating lactate is a fuel source for liver metabolism but may exacerbate metabolic diseases such as nonalcoholic steatohepatitis (NASH). Indeed, haploinsufficiency of lactate transporter monocarboxylate transporter 1 (MCT1) in mice reportedly promotes resistance to hepatic steatosis and inflammation. Here, we used adeno-associated virus (AAV) vectors to deliver thyroxin binding globulin (TBG)-Cre or lecithin-retinol acyltransferase (Lrat)-Cre to MCT1fl/fl mice on a choline-deficient, high-fat NASH diet to deplete hepatocyte or stellate cell MCT1, respectively. Stellate cell MCT1KO (AAV-Lrat-Cre) attenuated liver type 1 collagen protein expression and caused a downward trend in trichrome staining. MCT1 depletion in cultured human LX2 stellate cells also diminished collagen 1 protein expression. Tetra-ethylenglycol-cholesterol (Chol)-conjugated siRNAs, which enter all hepatic cell types, and hepatocyte-selective tri-N-acetyl galactosamine (GN)-conjugated siRNAs were then used to evaluate MCT1 function in a genetically obese NASH mouse model. MCT1 silencing by Chol-siRNA decreased liver collagen 1 levels, while hepatocyte-selective MCT1 depletion by AAV-TBG-Cre or by GN-siRNA unexpectedly increased collagen 1 and total fibrosis without effect on triglyceride accumulation. These findings demonstrate that stellate cell lactate transporter MCT1 significantly contributes to liver fibrosis through increased collagen 1 protein expression in vitro and in vivo, while hepatocyte MCT1 appears not to be an attractive therapeutic target for NASH.
Collapse
Affiliation(s)
- Kyounghee Min
- Program in Molecular Medicine, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Batuhan Yenilmez
- Program in Molecular Medicine, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Mark Kelly
- Program in Molecular Medicine, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Dimas Echeverria
- RNA Therapeutics Institute, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Michael Elleby
- Program in Molecular Medicine, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Lawrence M Lifshitz
- Program in Molecular Medicine, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Naideline Raymond
- Program in Molecular Medicine, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Emmanouela Tsagkaraki
- Program in Molecular Medicine, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Shauna M Harney
- Program in Molecular Medicine, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Chloe DiMarzio
- Program in Molecular Medicine, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Hui Wang
- Program in Molecular Medicine, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Nicholas McHugh
- RNA Therapeutics Institute, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Brianna Bramato
- RNA Therapeutics Institute, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Brett Morrison
- Department of Neurology, Johns Hopkins School of MedicineBaltimoreUnited States
| | - Jeffery D Rothstein
- Department of Neurology, Johns Hopkins School of MedicineBaltimoreUnited States
| | - Anastasia Khvorova
- RNA Therapeutics Institute, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Chan Medical SchoolWorcesterUnited States
| |
Collapse
|
15
|
Li Y, Wang X, Zhang Z, Shi L, Cheng L, Zhang X. Effect of the gut microbiome, plasma metabolome, peripheral cells, and inflammatory cytokines on obesity: a bidirectional two-sample Mendelian randomization study and mediation analysis. Front Immunol 2024; 15:1348347. [PMID: 38558794 PMCID: PMC10981273 DOI: 10.3389/fimmu.2024.1348347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Background Obesity is a metabolic and chronic inflammatory disease involving genetic and environmental factors. This study aimed to investigate the causal relationship among gut microbiota abundance, plasma metabolomics, peripheral cell (blood and immune cell) counts, inflammatory cytokines, and obesity. Methods Summary statistics of 191 gut microbiota traits (N = 18,340), 1,400 plasma metabolite traits (N = 8,299), 128 peripheral cell counts (blood cells, N = 408,112; immune cells, N = 3,757), 41 inflammatory cytokine traits (N = 8,293), and 6 obesity traits were obtained from publicly available genome-wide association studies. Two-sample Mendelian randomization (MR) analysis was applied to infer the causal links using inverse variance-weighted, maximum likelihood, MR-Egger, weighted median, weighted mode, and Wald ratio methods. Several sensitivity analyses were also utilized to ensure reliable MR results. Finally, we used mediation analysis to identify the pathway from gut microbiota to obesity mediated by plasma metabolites, peripheral cells, and inflammatory cytokines. Results MR revealed a causal effect of 44 gut microbiota taxa, 281 plasma metabolites, 27 peripheral cells, and 8 inflammatory cytokines on obesity. Among them, five shared causal gut microbiota taxa belonged to the phylum Actinobacteria, order Bifidobacteriales, family Bifidobacteriaceae, genus Lachnospiraceae UCG008, and species Eubacterium nodatum group. Furthermore, we screened 42 shared causal metabolites, 7 shared causal peripheral cells, and 1 shared causal inflammatory cytokine. Based on known causal metabolites, we observed that the metabolic pathways of D-arginine, D-ornithine, linoleic acid, and glycerophospholipid metabolism were closely related to obesity. Finally, mediation analysis revealed 20 mediation relationships, including the causal pathway from gut microbiota to obesity, mediated by 17 metabolites, 2 peripheral cells, and 1 inflammatory cytokine. Sensitivity analysis represented no heterogeneity or pleiotropy in this study. Conclusion Our findings support a causal relationship among gut microbiota, plasma metabolites, peripheral cells, inflammatory cytokines, and obesity. These biomarkers provide new insights into the mechanisms underlying obesity and contribute to its prevention, diagnosis, and treatment.
Collapse
Affiliation(s)
- Ying Li
- Human Molecular Genetics Group, National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
- Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Xin Wang
- Human Molecular Genetics Group, National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Zitong Zhang
- Human Molecular Genetics Group, National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- Department of Medical Genetics, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Lei Shi
- Human Molecular Genetics Group, National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- Department of Medical Genetics, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Liang Cheng
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xue Zhang
- Human Molecular Genetics Group, National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
- Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- Department of Medical Genetics, College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| |
Collapse
|
16
|
Zhou Z, Yao Y, Sun Y, Wang X, Huang S, Hou J, Wang L, Wei F. Serum betaine and dimethylglycine in mid-pregnancy and the risk of gestational diabetes mellitus: a case-control study. Endocrine 2024:10.1007/s12020-024-03732-4. [PMID: 38448678 DOI: 10.1007/s12020-024-03732-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 02/04/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE To investigate the associations of choline, betaine, dimethylglycine (DMG), L-carnitine, and Trimethylamine-N-oxide (TMAO) with the risk of Gestational diabetes mellitus (GDM) as well as the markers of glucose homeostasis. METHODS We performed a case-control study including 200 diagnosed GDM cases and 200 controls matched by maternal age (±2 years) and gestational age (±2 weeks). Concentrations of serum metabolites were measured by the high-performance liquid chromatography - tandem mass spectrometry (HPLC-MS/MS). RESULTS Compared to the control group, GDM group had significantly lower serum betaine concentration and betaine/choline ratio, and higher DMG concentration. Furthermore, decreased betaine concentration and betaine/choline ratio, increased DMG concentration showed significant association with the risk of GDM. In addition, serum betaine concentrations were negatively associated with blood glucose levels at 1-h post-glucose load (OGTT-1h), and both betaine and L-carnitine concentrations were positively associated with 1,5-anhydroglucitol levels. Betaine/choline ratio was negatively associated with OGTT-1h and blood glucose levels at 2-h post-glucose load (OGTT-2h) and serum choline concentrations were negatively associated with fasting blood glucose and positively associated with OGTT-2h. CONCLUSION Decreased serum betaine concentrations and betaine/choline ratio, and elevated DMG concentrations could be significant risk factors for GDM. Furthermore, betaine may be associated with blood glucose regulation and short-term glycemic fluctuations.
Collapse
Affiliation(s)
- Ziqing Zhou
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, China
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
| | - Yao Yao
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
| | - Yanan Sun
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
- Medical Insurance Office of Shenzhen Longgang Central Hospital, Shenzhen, Guangdong Province, China
| | - Xin Wang
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
- Jiamusi University, Jiamusi, Heilongjiang Province, China
| | - Shang Huang
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
- Shenzhen Children's Hospital of China Medical University, Shenzhen, Guangdong Province, China
| | - Jianli Hou
- Department of Gynecology and Obstetrics, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
| | - Lijun Wang
- Department of Nutrition, School of Medicine, Jinan University, Guangzhou, Guangdong Province, China.
| | - Fengxiang Wei
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, China.
- The Genetics Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China.
| |
Collapse
|
17
|
Min K, Yenilmez B, Kelly M, Echeverria D, Elleby M, Lifshitz LM, Raymond N, Tsagkaraki E, Harney SM, DiMarzio C, Wang H, McHugh N, Bramato B, Morrision B, Rothstein JD, Khvorova A, Czech MP. Lactate transporter MCT1 in hepatic stellate cells promotes fibrotic collagen expression in nonalcoholic steatohepatitis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.03.539244. [PMID: 37205462 PMCID: PMC10187148 DOI: 10.1101/2023.05.03.539244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Circulating lactate is a fuel source for liver metabolism but may exacerbate metabolic diseases such as nonalcoholic steatohepatitis (NASH). Indeed, haploinsufficiency of lactate transporter monocarboxylate transporter 1 (MCT1) in mice reportedly promotes resistance to hepatic steatosis and inflammation. Here, we used adeno-associated virus (AAV) vectors to deliver thyroxin binding globulin (TBG)-Cre or lecithin-retinol acyltransferase (Lrat)-Cre to MCT1fl/fl mice on a choline deficient, high fat NASH diet to deplete hepatocyte or stellate cell MCT1, respectively. Stellate cell MCT1KO (AAV-Lrat-Cre) attenuated liver type 1 collagen protein expression and caused a downward trend in trichrome staining. MCT1 depletion in cultured human LX2 stellate cells also diminished collagen 1 protein expression. Tetra-ethylenglycol-cholesterol (Chol)-conjugated siRNAs, which enter all hepatic cell types, and hepatocyte-selective tri-N-acetyl galactosamine (GN)-conjugated siRNAs were then used to evaluate MCT1 function in a genetically obese NASH mouse model. MCT1 silencing by Chol-siRNA decreased liver collagen 1 levels, while hepatocyte-selective MCT1 depletion by AAV-TBG-Cre or by GN-siRNA unexpectedly increased collagen 1 and total fibrosis without effect on triglyceride accumulation. These findings demonstrate that stellate cell lactate transporter MCT1 significantly contributes to liver fibrosis through increased collagen 1 protein expression in vitro and in vivo, while hepatocyte MCT1 appears not to be an attractive therapeutic target for NASH.
Collapse
Affiliation(s)
- Kyounghee Min
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, USA
| | - Batuhan Yenilmez
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, USA
| | - Mark Kelly
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, USA
| | - Dimas Echeverria
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, USA
| | - Michael Elleby
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, USA
| | - Lawrence M Lifshitz
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, USA
| | - Naideline Raymond
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, USA
| | | | - Shauna M Harney
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, USA
| | - Chloe DiMarzio
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, USA
| | - Hui Wang
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, USA
| | - Nicholas McHugh
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, USA
| | - Brianna Bramato
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, USA
| | - Brett Morrision
- Department of Neurology, Johns Hopkins School of Medicine, USA
| | | | - Anastasia Khvorova
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, USA
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, USA
| |
Collapse
|
18
|
Kamioka H, Yogosawa S, Oikawa T, Aizawa D, Ueda K, Saeki C, Haruki K, Shimoda M, Ikegami T, Nishikawa Y, Saruta M, Yoshida K. Dyrk2 gene transfer suppresses hepatocarcinogenesis by promoting the degradation of Myc and Hras. JHEP Rep 2023; 5:100759. [PMID: 37333975 PMCID: PMC10275997 DOI: 10.1016/j.jhepr.2023.100759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 03/06/2023] [Accepted: 03/21/2023] [Indexed: 06/20/2023] Open
Abstract
Background & Aims Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide, and has a poor prognosis. However, the molecular mechanisms underlying hepatocarcinogenesis and progression remain unknown. In vitro gain- and loss-of-function analyses in cell lines and xenografts revealed that dual-specificity tyrosine-regulated kinase 2 (DYRK2) influences tumour growth in HCC. Methods To investigate the role of Dyrk2 during hepatocarcinogenesis, we developed liver-specific Dyrk2 conditional knockout mice and an in vivo gene delivery system with a hydrodynamic tail vein injection and the Sleeping Beauty transposon. The antitumour effects of Dyrk2 gene transfer were investigated in a murine autologous carcinogenesis model. Results Dyrk2 expression was reduced in tumours, and that its downregulation was induced before hepatocarcinogenesis. Dyrk2 gene transfer significantly suppressed carcinogenesis. It also suppresses Myc-induced de-differentiation and metabolic reprogramming, which favours proliferative, and malignant potential by altering gene profiles. Dyrk2 overexpression caused Myc and Hras degradation at the protein level rather than at the mRNA level, and this degradation mechanism was regulated by the proteasome. Immunohistochemical analyses revealed a negative correlation between DYRK2 expression and MYC and longer survival in patients with HCC with high-DYRK2 and low-MYC expressions. Conclusions Dyrk2 protects the liver from carcinogenesis by promoting Myc and Hras degradation. Our findings would pave the way for a novel therapeutic approach using DYRK2 gene transfer. Impact and Implications Hepatocellular carcinoma (HCC) is one of the most common cancers, with a poor prognosis. Hence, identifying molecules that can become promising targets for therapies is essential to improve mortality. No studies have clarified the association between DYRK2 and carcinogenesis, although DYRK2 is involved in tumour growth in various cancer cells. This is the first study to show that Dyrk2 expression decreases during hepatocarcinogenesis and that Dyrk2 gene transfer is an attractive approach with tumour suppressive activity against HCC by suppressing Myc-mediated de-differentiation and metabolic reprogramming that favours proliferative and malignant potential via Myc and Hras degradation.
Collapse
Affiliation(s)
- Hiroshi Kamioka
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo, Japan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Satomi Yogosawa
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo, Japan
| | - Tsunekazu Oikawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Daisuke Aizawa
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kaoru Ueda
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Chisato Saeki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Koichiro Haruki
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Masayuki Shimoda
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Toru Ikegami
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Yuji Nishikawa
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Masayuki Saruta
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kiyotsugu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
19
|
Basha A, May SC, Anderson RM, Samala N, Mirmira RG. Non-Alcoholic Fatty Liver Disease: Translating Disease Mechanisms into Therapeutics Using Animal Models. Int J Mol Sci 2023; 24:9996. [PMID: 37373143 PMCID: PMC10298283 DOI: 10.3390/ijms24129996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a range of pathologies arising from fat accumulation in the liver in the absence of excess alcohol use or other causes of liver disease. Its complications include cirrhosis and liver failure, hepatocellular carcinoma, and eventual death. NAFLD is the most common cause of liver disease globally and is estimated to affect nearly one-third of individuals in the United States. Despite knowledge that the incidence and prevalence of NAFLD are increasing, the pathophysiology of the disease and its progression to cirrhosis remain insufficiently understood. The molecular pathogenesis of NAFLD involves insulin resistance, inflammation, oxidative stress, and endoplasmic reticulum stress. Better insight into these molecular pathways would allow for therapies that target specific stages of NAFLD. Preclinical animal models have aided in defining these mechanisms and have served as platforms for screening and testing of potential therapeutic approaches. In this review, we will discuss the cellular and molecular mechanisms thought to contribute to NAFLD, with a focus on the role of animal models in elucidating these mechanisms and in developing therapies.
Collapse
Affiliation(s)
- Amina Basha
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Sarah C. May
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Ryan M. Anderson
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Niharika Samala
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Raghavendra G. Mirmira
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
20
|
Sheykhsaran E, Abbasi A, Ebrahimzadeh Leylabadlo H, Sadeghi J, Mehri S, Naeimi Mazraeh F, Feizi H, Bannazadeh Baghi H. Gut microbiota and obesity: an overview of microbiota to microbial-based therapies. Postgrad Med J 2023; 99:384-402. [PMID: 35140178 DOI: 10.1136/postgradmedj-2021-141311] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/15/2022] [Indexed: 12/14/2022]
Abstract
The increasing prevalence of obesity and overweight is a significant public concern throughout the world. Obesity is a complex disorder involving an excessive amount of body fat. It is not just a cosmetic concern. It is a medical challenge that increases the risk of other diseases and health circumstances, such as diabetes, heart disease, high blood pressure and certain cancers. Environmental and genetic factors are involved in obesity as a significant metabolic disorder along with diabetes. Gut microbiota (GM) has a high potential for energy harvesting from the diet. In the current review, we aim to consider the role of GM, gut dysbiosis and significant therapies to treat obesity. Dietary modifications, probiotics, prebiotics, synbiotics compounds, using faecal microbiota transplant, and other microbial-based therapies are the strategies to intervene in obesity reducing improvement. Each of these factors serves through various mechanisms including a variety of receptors and compounds to control body weight. Trial and animal investigations have indicated that GM can affect both sides of the energy-balancing equation; first, as an influencing factor for energy utilisation from the diet and also as an influencing factor that regulates the host genes and energy storage and expenditure. All the investigated articles declare the clear and inevitable role of GM in obesity. Overall, obesity and obesity-relevant metabolic disorders are characterised by specific modifications in the human microbiota's composition and functions. The emerging therapeutic methods display positive and promising effects; however, further research must be done to update and complete existing knowledge.
Collapse
Affiliation(s)
- Elham Sheykhsaran
- Immunology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Abbasi
- Student Research Committee, Department of Food Sciences and Technology Research Institute, Faculty of Nutrition Sciences and food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Javid Sadeghi
- Immunology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samaneh Mehri
- Department of Biochemistry and structural Biology, University of Alabama, Birmingham, Alabama, USA
| | - Fariba Naeimi Mazraeh
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Feizi
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Bannazadeh Baghi
- Immunology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
21
|
Abbasi MSP, Tousi AZ, Yazdani Y, Vahdat S, Gharebakhshi F, Nikrad N, Manzouri A, Ardekani AM, Jafarzadeh F. Dietary choline and betaine intake, cardio-metabolic risk factors and prevalence of metabolic syndrome among overweight and obese adults. BMC Endocr Disord 2023; 23:67. [PMID: 36973700 PMCID: PMC10041695 DOI: 10.1186/s12902-023-01323-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Choline is an important metabolite involved in phospholipids synthesis, including serum lipids, and is the immediate precursor of betaine. There are numerous studies with inconsistent results that evaluated the association between dietary choline intakes with cardiovascular risk factors. In addition, the association between dietary betaine and choline intakes with cardio-metabolic risk factors is not well studied. In the current study, our aim was to evaluate dietary choline and betaine intakes in the usual diet of obese individuals and to assess its association with serum lipids, blood pressure and glycemic markers among obese individuals. METHODS We recruited a total number of 359 obese people aged between 20 and 50 years in the present study. A semi-quantitative food frequency questionnaire (FFQ) was used for dietary assessment; dietary choline and betaine intakes were calculated using the United States Department of Agriculture (USDA) database. National cholesterol education program adult treatment panel (NCEP-ATP)-III criteria was used metabolic syndrome (MetS) definition. Enzymatic methods were used to assess biochemical variables. Body composition was measured with the bioelectrical impedance analysis (BIA) method. RESULTS Higher body mass index (BMI), waist to hip ratio (WHR), fat-free mass (FFM) and basal metabolic rate (BMR) were observed in higher tertiles of dietary choline intake (P < 0.01). There was no significant difference in terms of biochemical parameters among different tertiles of dietary choline intake, while systolic blood pressure (SBP) and diastolic blood pressure (DBP) were reduced in higher betaine tertiles (P < 0.05). For total dietary choline and betaine intakes, there was a reduction in DBP and low density lipoprotein (LDL) concentrations (P < 0.05). Also, a non-significant reduction in serum total cholesterol (TC), triglyceride (TG) and MetS prevalence was observed in higher tertiles of dietary choline and betaine intakes. After classification of the study population according to MetS status, there was no significant difference in biochemical variables in subjects with MetS (P > 0.05), while in the non-MetS group, SBP, DBP, TG and insulin levels reduced in higher tertiles of dietary betaine and choline (P > 0.05). CONCLUSION According to our findings, higher dietary intakes of choline and betaine were associated with lower levels of blood pressure and LDL concentrations among obese individuals. Further studies are warranted to confirm the results of the current study.
Collapse
Affiliation(s)
| | - Ayda Zahiri Tousi
- Razavi Cancer Research Center, Razavi Hospital, Imam Reza International University, Mashhad, Iran
| | - Yalda Yazdani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Vahdat
- Isfahan Kidney Disease Research Center, School of Medicine, Khorshid Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farshad Gharebakhshi
- Department of Radiology, School of Medicne, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Negin Nikrad
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Manzouri
- Health Policy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abnoos Mokhtari Ardekani
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Science, & Physiology Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Faria Jafarzadeh
- Department of Internal Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnourd, Iran
| |
Collapse
|
22
|
Papanastasatou M, Verykokakis M. Innate-like T lymphocytes in chronic liver disease. Front Immunol 2023; 14:1114605. [PMID: 37006304 PMCID: PMC10050337 DOI: 10.3389/fimmu.2023.1114605] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/28/2023] [Indexed: 03/17/2023] Open
Abstract
In addition to its metabolic activities, it is now clear that the liver hosts a number of diverse immune cell types that control tissue homeostasis. Foremost among these are innate-like T lymphocytes, including natural killer T (NKT) and mucosal-associated innate T (MAIT) cells, which are a population of specialized T cells with innate characteristics that express semi-invariant T cell receptors with non-peptide antigen specificity. As primary liver residents, innate-like T cells have been associated with immune tolerance in the liver, but also with a number of hepatic diseases. Here, we focus on the biology of NKT and MAIT cells and how they operate during the course of chronic inflammatory diseases that eventually lead to hepatocellular carcinoma.
Collapse
|
23
|
Suoangbaji T, Zhang VX, Ng IOL, Ho DWH. Single-Cell Analysis of Primary Liver Cancer in Mouse Models. Cells 2023; 12:cells12030477. [PMID: 36766817 PMCID: PMC9914042 DOI: 10.3390/cells12030477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/17/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Primary liver cancer (PLC), consisting mainly of hepatocellular carcinoma and intrahepatic cholangiocarcinoma, is one of the major causes of cancer-related mortality worldwide. The curative therapy for PLC is surgical resection and liver transplantation, but most PLCs are inoperable at diagnosis. Even after surgery, there is a high rate of tumor recurrence. There is an unmet clinical need to discover more effective treatment options for advanced PLCs. Pre-clinical mouse models in PLC research have played a critical role in identifying key oncogenic drivers and signaling pathways in hepatocarcinogenesis. Furthermore, recent advances in single-cell RNA sequencing (scRNA-seq) have provided an unprecedented degree of resolution in such characterization. In this review, we will summarize the recent studies that utilized pre-clinical mouse models with the combination of scRNA-seq to provide an understanding of different aspects of PLC. We will focus particularly on the potentially actionable targets regarding the cellular and molecular components. We anticipate that the findings in mouse models could complement those in patients. With more defined etiological background, mouse models may provide valuable insights.
Collapse
Affiliation(s)
| | | | - Irene Oi-Lin Ng
- Correspondence: (I.O.-L.N.); (D.W.-H.H.); Fax: +852-28872-5197 (I.O.-L.N.); +852-2819-5375 (D.W.-H.H.)
| | - Daniel Wai-Hung Ho
- Correspondence: (I.O.-L.N.); (D.W.-H.H.); Fax: +852-28872-5197 (I.O.-L.N.); +852-2819-5375 (D.W.-H.H.)
| |
Collapse
|
24
|
Finney AC, Das S, Kumar D, McKinney MP, Cai B, Yurdagul A, Rom O. The interplay between nonalcoholic fatty liver disease and atherosclerotic cardiovascular disease. Front Cardiovasc Med 2023; 10:1116861. [PMID: 37200978 PMCID: PMC10185914 DOI: 10.3389/fcvm.2023.1116861] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/23/2023] [Indexed: 05/20/2023] Open
Abstract
Therapeutic approaches that lower circulating low-density lipoprotein (LDL)-cholesterol significantly reduced the burden of cardiovascular disease over the last decades. However, the persistent rise in the obesity epidemic is beginning to reverse this decline. Alongside obesity, the incidence of nonalcoholic fatty liver disease (NAFLD) has substantially increased in the last three decades. Currently, approximately one third of world population is affected by NAFLD. Notably, the presence of NAFLD and particularly its more severe form, nonalcoholic steatohepatitis (NASH), serves as an independent risk factor for atherosclerotic cardiovascular disease (ASCVD), thus, raising interest in the relationship between these two diseases. Importantly, ASCVD is the major cause of death in patients with NASH independent of traditional risk factors. Nevertheless, the pathophysiology linking NAFLD/NASH with ASCVD remains poorly understood. While dyslipidemia is a common risk factor underlying both diseases, therapies that lower circulating LDL-cholesterol are largely ineffective against NASH. While there are no approved pharmacological therapies for NASH, some of the most advanced drug candidates exacerbate atherogenic dyslipidemia, raising concerns regarding their adverse cardiovascular consequences. In this review, we address current gaps in our understanding of the mechanisms linking NAFLD/NASH and ASCVD, explore strategies to simultaneously model these diseases, evaluate emerging biomarkers that may be useful to diagnose the presence of both diseases, and discuss investigational approaches and ongoing clinical trials that potentially target both diseases.
Collapse
Affiliation(s)
- Alexandra C. Finney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Sandeep Das
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Dhananjay Kumar
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - M. Peyton McKinney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, NY, United States
| | - Arif Yurdagul
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Correspondence: Arif Yurdagul Oren Rom
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA, United States
- Correspondence: Arif Yurdagul Oren Rom
| |
Collapse
|
25
|
Heintz MM, Eccles JA, Olack EM, Maner-Smith KM, Ortlund EA, Baldwin WS. Human CYP2B6 produces oxylipins from polyunsaturated fatty acids and reduces diet-induced obesity. PLoS One 2022; 17:e0277053. [PMID: 36520866 PMCID: PMC9754190 DOI: 10.1371/journal.pone.0277053] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/18/2022] [Indexed: 12/23/2022] Open
Abstract
Multiple factors in addition to over consumption lead to obesity and non-alcoholic fatty liver disease (NAFLD) in the United States and worldwide. CYP2B6 is the only human detoxification CYP whose loss is associated with obesity, and Cyp2b-null mice show greater diet-induced obesity with increased steatosis than wildtype mice. However, a putative mechanism has not been determined. LC-MS/MS revealed that CYP2B6 metabolizes PUFAs, with a preference for metabolism of ALA to 9-HOTrE and to a lesser extent 13-HOTrE with a preference for metabolism of PUFAs at the 9- and 13-positions. To further study the role of CYP2B6 in vivo, humanized-CYP2B6-transgenic (hCYP2B6-Tg) and Cyp2b-null mice were fed a 60% high-fat diet for 16 weeks. Compared to Cyp2b-null mice, hCYP2B6-Tg mice showed reduced weight gain and metabolic disease as measured by glucose tolerance tests, however hCYP2B6-Tg male mice showed increased liver triglycerides. Serum and liver oxylipin metabolite concentrations increased in male hCYP2B6-Tg mice, while only serum oxylipins increased in female hCYP2B6-Tg mice with the greatest increases in LA oxylipins metabolized at the 9 and 13-positions. Several of these oxylipins, specifically 9-HODE, 9-HOTrE, and 13-oxoODE, are PPAR agonists. RNA-seq data also demonstrated sexually dimorphic changes in gene expression related to nuclear receptor signaling, especially CAR > PPAR with qPCR suggesting PPARγ signaling is more likely than PPARα signaling in male mice. Overall, our data indicates that CYP2B6 is an anti-obesity enzyme, but probably to a lesser extent than murine Cyp2b's. Therefore, the inhibition of CYP2B6 by xenobiotics or dietary fats can exacerbate obesity and metabolic disease potentially through disrupted PUFA metabolism and the production of key lipid metabolites.
Collapse
Affiliation(s)
- Melissa M. Heintz
- Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Jazmine A. Eccles
- Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Emily M. Olack
- Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Kristal M. Maner-Smith
- Emory Integrated Metabolomics and Lipodomics Core, Emory University, Atlanta, Georgia, United States of America
| | - Eric A. Ortlund
- Department of Biochemistry, Emory University School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - William S. Baldwin
- Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
26
|
A Study of the Metabolic Pathways Affected by Gestational Diabetes Mellitus: Comparison with Type 2 Diabetes. Diagnostics (Basel) 2022; 12:diagnostics12112881. [PMID: 36428943 PMCID: PMC9689375 DOI: 10.3390/diagnostics12112881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) remains incompletely understood and increases the risk of developing Diabetes mellitus type 2 (DM2). Metabolomics provides insights etiology and pathogenesis of disease and discovery biomarkers for accurate detection. Nuclear magnetic resonance (NMR) spectroscopy is a key platform defining metabolic signatures in intact serum/plasma. In the present study, we used NMR-based analysis of macromolecules free-serum to accurately characterize the altered metabolic pathways of GDM and assessing their similarities to DM2. Our findings could contribute to the understanding of the pathophysiology of GDM and help in the identification of metabolomic markers of the disease. METHODS Sixty-two women with GDM matched with seventy-seven women without GDM (control group). 1H NMR serum spectra were acquired on an 11.7 T Bruker Avance DRX NMR spectrometer. RESULTS We identified 55 metabolites in both groups, 25 of which were significantly altered in the GDM group. GDM group showed elevated levels of ketone bodies, 2-hydroxybutyrate and of some metabolic intermediates of branched-chain amino acids (BCAAs) and significantly lower levels of metabolites of one-carbon metabolism, energy production, purine metabolism, certain amino acids, 3-methyl-2-oxovalerate, ornithine, 2-aminobutyrate, taurine and trimethylamine N-oxide. CONCLUSION Metabolic pathways affected in GDM were beta-oxidation, ketone bodies metabolism, one-carbon metabolism, arginine and ornithine metabolism likewise in DM2, whereas BCAAs catabolism and aromatic amino acids metabolism were affected, but otherwise than in DM2.
Collapse
|
27
|
Vasishta S, Ganesh K, Umakanth S, Joshi MB. Ethnic disparities attributed to the manifestation in and response to type 2 diabetes: insights from metabolomics. Metabolomics 2022; 18:45. [PMID: 35763080 PMCID: PMC9239976 DOI: 10.1007/s11306-022-01905-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 04/13/2022] [Indexed: 11/21/2022]
Abstract
Type 2 diabetes (T2D) associated health disparities among different ethnicities have long been known. Ethnic variations also exist in T2D related comorbidities including insulin resistance, vascular complications and drug response. Genetic heterogeneity, dietary patterns, nutrient metabolism and gut microbiome composition attribute to ethnic disparities in both manifestation and progression of T2D. These factors differentially regulate the rate of metabolism and metabolic health. Metabolomics studies have indicated significant differences in carbohydrate, lipid and amino acid metabolism among ethnicities. Interestingly, genetic variations regulating lipid and amino acid metabolism might also contribute to inter-ethnic differences in T2D. Comprehensive and comparative metabolomics analysis between ethnicities might help to design personalized dietary regimen and newer therapeutic strategies. In the present review, we explore population based metabolomics data to identify inter-ethnic differences in metabolites and discuss how (a) genetic variations, (b) dietary patterns and (c) microbiome composition may attribute for such differences in T2D.
Collapse
Affiliation(s)
- Sampara Vasishta
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, 576104, Manipal, India
| | - Kailash Ganesh
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, 576104, Manipal, India
| | | | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, 576104, Manipal, India.
- Manipal School of Life Sciences, Planetarium Complex Manipal Academy of Higher Education Manipal, 576104, Manipal, India.
| |
Collapse
|
28
|
Satellite cell content and muscle regeneration in a mouse model of NAFLD. Nutrition 2022; 96:111570. [DOI: 10.1016/j.nut.2021.111570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 11/01/2021] [Accepted: 12/05/2021] [Indexed: 12/20/2022]
|
29
|
Chen S, Zong G, Wu Q, Yun H, Niu Z, Zheng H, Zeng R, Sun L, Lin X. Associations of plasma glycerophospholipid profile with modifiable lifestyles and incident diabetes in middle-aged and older Chinese. Diabetologia 2022; 65:315-328. [PMID: 34800146 DOI: 10.1007/s00125-021-05611-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/17/2021] [Indexed: 10/19/2022]
Abstract
AIMS/HYPOTHESIS Glycerophospholipid (GPL) perturbance was linked to the pathogenesis of diabetes in animal studies but prospective studies in humans are rare, particularly in Asians. We aimed to investigate the associations between plasma GPLs and incident diabetes and to explore effects of lifestyle on the associations in a Chinese population. METHODS The study included 1877 community-dwelling Chinese individuals aged 50-70 years (751 men and 1126 women), free of diabetes at baseline and followed for 6 years. A total of 160 GPL species were quantified in plasma at baseline by using high-throughput targeted lipidomics. Log-Poisson regression was used to assess the associations between GPLs and incidence of diabetes. RESULTS Over the 6 years of follow-up, 499 participants (26.6%) developed diabetes. After multivariable adjustment, eight GPLs were positively associated with incident diabetes (RRper SD 1.13-1.25; all false-discovery rate [FDR]-corrected p < 0.05), including five novel GLPs, namely phosphatidylcholines (PCs; 16:0/18:1, 18:0/16:1, 18:1/20:3), lysophosphatidylcholine (LPC; 20:3) and phosphatidylethanolamine (PE; 16:0/16:1), and three reported GPLs (PCs 16:0/16:1, 16:0/20:3 and 18:0/20:3). In network analysis, a PC-containing module was positively associated with incident diabetes (RRper SD 1.16 [95% CI 1.06, 1.26]; FDR-corrected p < 0.05). Notably, three of the diabetes-associated PCs (16:0/16:1, 16:0/18:1 and 18:0/16:1) and PE (16:0/16:1) were associated not only with fatty acids in the de novo lipogenesis (DNL) pathway, especially 16:1n-7 (Spearman correlation coefficients = 0.35-0.62, p < 0.001), but also with an unhealthy dietary pattern high in refined grains and low in fish, dairy and soy products (|factor loadings| ≥0.2). When stratified by physical activity levels, the associations of the eight GPLs and the PC module with incident diabetes were stronger in participants with lower physical activity (RRper SD 1.24-1.49, FDR-corrected p < 0.05) than in those with the median and higher physical activity levels (RRper SD 1.03-1.12, FDR-corrected p ≥ 0.05; FDR-corrected pinteraction < 0.05). CONCLUSIONS/INTERPRETATION Eight GPLs, especially PCs associated with the DNL pathway, were positively associated with incident diabetes in a cohort of Chinese men and women. The associations were most prominent in participants with a low level of physical activity.
Collapse
Affiliation(s)
- Shuangshuang Chen
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Geng Zong
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qingqing Wu
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Huan Yun
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhenhua Niu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - He Zheng
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rong Zeng
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| | - Liang Sun
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Xu Lin
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
30
|
Chang TY, Wu CH, Chang CY, Lee FJ, Wang BW, Doong JY, Lin YS, Kuo CS, Huang RFS. Optimal Dietary Intake Composition of Choline and Betaine Is Associated with Minimized Visceral Obesity-Related Hepatic Steatosis in a Case-Control Study. Nutrients 2022; 14:261. [PMID: 35057441 PMCID: PMC8779168 DOI: 10.3390/nu14020261] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
Few studies on humans have comprehensively evaluated the intake composition of methyl-donor nutrients (MDNs: choline, betaine, and folate) in relation to visceral obesity (VOB)-related hepatic steatosis (HS), the hallmark of non-alcoholic fatty liver diseases. In this case-control study, we recruited 105 patients with HS and 104 without HS (controls). HS was diagnosed through ultrasound examination. VOB was measured using a whole-body analyzer. MDN intake was assessed using a validated quantitative food frequency questionnaire. After adjustment for multiple HS risk factors, total choline intake was the most significant dietary determinant of HS in patients with VOB (Beta: -0.41, p = 0.01). Low intake of choline (<6.9 mg/kg body weight), betaine (<3.1 mg/kg body weight), and folate (<8.8 μg/kg body weight) predicted increased odds ratios (ORs) of VOB-related HS (choline: OR: 22, 95% confidence interval [CI]: 6.5-80; betaine: OR: 14, 95% CI: 4.4-50; and folate: OR: 19, 95% CI: 5.2-74). Combined high intake of choline and betaine, but not folate, was associated with an 81% reduction in VOB-related HS (OR: 0.19, 95% CI: 0.05-0.69). Our data suggest that the optimal intake of choline and betaine can minimize the risk of VOB-related HS in a threshold-dependent manner.
Collapse
Affiliation(s)
- Ting-Yu Chang
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (T.-Y.C.); (J.-Y.D.); (Y.-S.L.)
| | - Chien-Hsien Wu
- Ph.D. Program in Nutrition and Food Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan;
- Department of Gastroenterology and Hepatology, Taipei Hospital, Ministry of Health and Welfare, New Taipei City 242, Taiwan
| | - Chi-Yang Chang
- Department of Gastroenterology and Hepatology, Fu Jen Catholic University Hospital, New Taipei City 243089, Taiwan; (C.-Y.C.); (F.-J.L.)
| | - Fu-Jen Lee
- Department of Gastroenterology and Hepatology, Fu Jen Catholic University Hospital, New Taipei City 243089, Taiwan; (C.-Y.C.); (F.-J.L.)
| | - Bei-Wen Wang
- Department of Nutrition, Fu Jen Catholic University Hospital, New Taipei City 243089, Taiwan;
| | - Jia-Yau Doong
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (T.-Y.C.); (J.-Y.D.); (Y.-S.L.)
| | - Yu-Shun Lin
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (T.-Y.C.); (J.-Y.D.); (Y.-S.L.)
| | - Chang-Sheng Kuo
- Department of Nutrition, Fu Jen Catholic University Hospital, New Taipei City 243089, Taiwan;
| | - Rwei-Fen S. Huang
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (T.-Y.C.); (J.-Y.D.); (Y.-S.L.)
- Ph.D. Program in Nutrition and Food Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan;
| |
Collapse
|
31
|
SEKI M, MIWA A, OHSAKA F, KARATSU Y, TSURUTA T, HINO S, MORITA T, SONOYAMA K. Local free fatty acids trigger the expression of lipopolysaccharide-binding protein in murine white adipose tissue. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2022; 41:54-65. [PMID: 35433160 PMCID: PMC8970656 DOI: 10.12938/bmfh.2021-061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/26/2021] [Indexed: 11/05/2022]
Abstract
Although lipopolysaccharide (LPS)-binding protein (LBP) is an acute-phase protein mainly
produced by hepatocytes, it has also been proposed to be a pro-inflammatory adipokine.
Obesity and the consumption of a high-fat diet (HFD) are reportedly associated with
elevated levels of LPS in plasma and free fatty acids (FFAs) in white adipose tissue
(WAT). We examined whether circulating LPS or local FFAs are responsible for the
HFD-induced increase of LBP in WAT. Male C57BL/6J mice were fed either a normal-fat diet
(NFD) or an HFD. The mRNA levels in the liver and mesenteric WAT (mWAT), total FFA content
in mWAT, and LBP and LPS concentrations in plasma were determined. The
Lbp mRNA level in mWAT was higher in mice fed the HFD than in those fed
the NFD for 3, 7, or 28 days or 14 weeks, whereas the hepatic Lbp mRNA
level did not differ between the groups. The Lbp mRNA level in mWAT was
also increased by the HFD in germ-free mice, which do not have gut microbiota, the source
of LPS. The plasma LPS level did not show a significant correlation with the mWAT
Lbp mRNA level. The total FFA content in mWAT was higher in mice fed
the HFD than in those fed the NFD and positively correlated with the Lbp
mRNA level. Supplementation with palmitic acid increased the Lbp mRNA
level in 3T3-L1 adipocytes. We propose that local FFAs, but not circulating LPS, are the
trigger for increased Lbp expression in mWAT of mice fed the HFD.
Collapse
Affiliation(s)
- Manami SEKI
- Graduate School of Life Science, Hokkaido University, Kita-9, Nishi-9, Kita-ku, Sapporo 060-8589, Japan
| | - Akiho MIWA
- Graduate School of Life Science, Hokkaido University, Kita-9, Nishi-9, Kita-ku, Sapporo 060-8589, Japan
| | - Fumina OHSAKA
- Graduate School of Agriculture, Hokkaido University, Kita-9, Nishi-9, Kita-ku, Sapporo 060-8589, Japan
| | - Yugo KARATSU
- Graduate School of Agriculture, Hokkaido University, Kita-9, Nishi-9, Kita-ku, Sapporo 060-8589, Japan
| | - Takeshi TSURUTA
- Graduate School of Environmental and Life Science, Okayama University, 2-1-1 Tsushima-Naka, Kita-ku, Okayama-shi, Okayama 700-8530, Japan
| | - Shingo HINO
- College of Agriculture, Academic Institute, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka-shi, Shizuoka 422-8529, Japan
| | - Tatsuya MORITA
- College of Agriculture, Academic Institute, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka-shi, Shizuoka 422-8529, Japan
| | - Kei SONOYAMA
- Research Faculty of Agriculture, Hokkaido University, Kita-9, Nishi-9, Kita-ku, Sapporo 060-8589, Japan
| |
Collapse
|
32
|
Zeng Q, Zhao M, Wang F, Li Y, Li H, Zheng J, Chen X, Zhao X, Ji L, Gao X, Liu C, Wang Y, Cheng S, Xu J, Pan B, Sun J, Li Y, Li D, He Y, Zheng L. Integrating Choline and Specific Intestinal Microbiota to Classify Type 2 Diabetes in Adults: A Machine Learning Based Metagenomics Study. Front Endocrinol (Lausanne) 2022; 13:906310. [PMID: 35832425 PMCID: PMC9271784 DOI: 10.3389/fendo.2022.906310] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/13/2022] [Indexed: 12/12/2022] Open
Abstract
Emerging evidence is examining the precise role of intestinal microbiota in the pathogenesis of type 2 diabetes. The aim of this study was to investigate the association of intestinal microbiota and microbiota-generated metabolites with glucose metabolism systematically in a large cross-sectional study in China. 1160 subjects were divided into three groups based on their glucose level: normal glucose group (n=504), prediabetes group (n=394), and diabetes group (n=262). Plasma concentrations of TMAO, choline, betaine, and carnitine were measured. Intestinal microbiota was measured in a subgroup of 161 controls, 144 prediabetes and 56 diabetes by using metagenomics sequencing. We identified that plasma choline [Per SD of log-transformed change: odds ratio 1.36 (95 confidence interval 1.16, 1.58)] was positively, while betaine [0.77 (0.66, 0.89)] was negatively associated with diabetes, independently of TMAO. Individuals with diabetes could be accurately distinguished from controls by integrating data on choline, and certain microbiota species, as well as traditional risk factors (AUC=0.971). KOs associated with the carbohydrate metabolism pathway were enhanced in individuals with high choline level. The functional shift in the carbohydrate metabolism pathway in high choline group was driven by species Ruminococcus lactaris, Coprococcus catus and Prevotella copri. We demonstrated the potential ability for classifying diabetic population by choline and specific species, and provided a novel insight of choline metabolism linking the microbiota to impaired glucose metabolism and diabetes.
Collapse
Affiliation(s)
- Qiang Zeng
- Health Management Institute, the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Mingming Zhao
- China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, China
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides of Ministry of Health, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Fei Wang
- Health Management Institute, the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yanping Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, China
| | - Huimin Li
- National Human Genetic Resources Center, Beijing, China, National Research Institute for Family Planning, Beijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianqiong Zheng
- Department of Obstetrics and Gynecology, The Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated Hospital of Shanghai University, Wenzhou People’s Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, China
| | - Xianyang Chen
- Bao Feng Key Laboratory of Genetics and Metabolism, Zhongyuan Biotechnology Holdings Group, Beijing, China
- Zhong Guan Cun Biological and Medical Big Data Center, Zhong Guan Cun Medical Engineering & Health Industry Base, Beijing, China
| | - Xiaolan Zhao
- Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Liang Ji
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides of Ministry of Health, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Xiangyang Gao
- Health Management Institute, the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Changjie Liu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides of Ministry of Health, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Yu Wang
- Health Management Center, The 910th Hospital of People's Liberation Army, Quanzhou, China
| | - Si Cheng
- China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, China
| | - Jie Xu
- China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, China
| | - Bing Pan
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides of Ministry of Health, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Jing Sun
- Health Management Center, The China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yongli Li
- Department of Health Management, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Dongfang Li
- Department of Microbial Research, WeHealthGene Institute, Shenzhen, Guangdong, China
- Institute of Statistics, NanKai University, Tianjin, China
| | - Yuan He
- National Human Genetic Resources Center, Beijing, China, National Research Institute for Family Planning, Beijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- *Correspondence: Yuan He, ; Lemin Zheng,
| | - Lemin Zheng
- China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, China
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides of Ministry of Health, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
- *Correspondence: Yuan He, ; Lemin Zheng,
| |
Collapse
|
33
|
Recombinant human GLP-1 beinaglutide regulates lipid metabolism of adipose tissues in diet-induced obese mice. iScience 2021; 24:103382. [PMID: 34841227 PMCID: PMC8605346 DOI: 10.1016/j.isci.2021.103382] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/19/2021] [Accepted: 10/27/2021] [Indexed: 01/18/2023] Open
Abstract
GLP-1 analogs are a class of glucose-lowering agents with multiple benefits in diabetes, but its role in adipose tissues remains to be elucidated. The aim of this study was to determine the action of recombinant human GLP-1 (rhGLP-1) Beinaglutide (BN) in the insulin sensitivity and lipid metabolism of adipose tissues. We have shown that, after BN injection, obese mice displayed lower body weight, fat mass, and plasma lipid levels. In addition, BN promoted the insulin sensitivity in the white adipose tissues. Furthermore, we have found that the BN treatment caused significant changes in content and composition of different lipid classes, including glycerolipids, glycerophospholipids, and sphingolipids, as well as expression of genes in lipid metabolic pathways in the adipose tissues. Taken together, our data demonstrate that BN could resist HFD-induced obesity by targeting the composition of major lipid classes and the expression of genes in lipid metabolism of adipose tissues. Recombinant human GLP-1 Beinaglutide (BN) reduces high-fat-diet-induced obesity BN increases insulin sensitivity of adipocytes in vivo and in vitro BN alters lipidomic and transcriptomic profiles in adipose tissues of obese mice BN promotes thermogenic gene expression in adipose tissues
Collapse
|
34
|
Zhou Y, Zhao R, Lyu Y, Shi H, Ye W, Tan Y, Li R, Xu Y. Serum and Amniotic Fluid Metabolic Profile Changes in Response to Gestational Diabetes Mellitus and the Association with Maternal-Fetal Outcomes. Nutrients 2021; 13:3644. [PMID: 34684645 PMCID: PMC8539410 DOI: 10.3390/nu13103644] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/15/2021] [Accepted: 10/15/2021] [Indexed: 11/25/2022] Open
Abstract
This study was designed to identify serum and amniotic fluid (AF) metabolic profile changes in response to gestational diabetes mellitus (GDM) and explore the association with maternal-fetal outcomes. We established the GDM rat models by combining a high-fat diet (HFD) with an injection of low-dose streptozotocin (STZ), detected the fasting plasma glucose (FPG) of pregnant rats in the second and third trimester, and collected AF and fetal rats by cesarean section on gestational day 19 (GD19), as well as measuring the weight and crown-rump length (CRL) of fetal rats. We applied liquid chromatography-tandem mass spectrometry (LC-MS/MS) for the untargeted metabolomics analyses of serum and AF samples and then explored their correlation with maternal-fetal outcomes via the co-occurrence network. The results showed that 91 and 68 metabolites were upregulated and 125 and 78 metabolites were downregulated in serum and AF samples exposed to GDM, respectively. In maternal serum, the obvious alterations emerged in lipids and lipid-like molecules, while there were great changes in carbohydrate and carbohydrate conjugates, followed by amino acids, peptides, and analogs in amniotic fluid. The altered pathways both in serum and AF samples were amino acid, lipid, nucleotide, and vitamin metabolism pathways. In response to GDM, changes in the steroid hormone metabolic pathway occurred in serum, and an altered carbohydrate metabolism pathway was found in AF samples. Among differential metabolites in two kinds of samples, there were 34 common biochemicals shared by serum and AF samples, and a mutual significant association existed. These shared differential metabolites were implicated in several metabolism pathways, including choline, tryptophan, histidine, and nicotinate and nicotinamide metabolism, and among them, N1-methyl-4-pyridone-3-carboxamide, 5'-methylthioadenosine, and kynurenic acid were significantly associated with both maternal FPG and fetal growth. In conclusion, serum and AF metabolic profiles were remarkably altered in response to GDM. N1-Methyl-4-pyridone-3-carboxamide, 5'-methylthioadenosine, and kynurenic acid have the potential to be taken as biomarkers for maternal-fetal health status of GDM. The common and inter-related differential metabolites both in the serum and AF implied the feasibility of predicting fetal health outcomes via detecting the metabolites in maternal serum exposed to GDM.
Collapse
Affiliation(s)
- Yalin Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, NO.38 Xueyuan Road, Beijing 100083, China; (Y.Z.); (R.Z.); (Y.L.); (H.S.); (W.Y.); (Y.T.); (R.L.)
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, NO.38 Xueyuan Road, Beijing 100083, China
| | - Runlong Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, NO.38 Xueyuan Road, Beijing 100083, China; (Y.Z.); (R.Z.); (Y.L.); (H.S.); (W.Y.); (Y.T.); (R.L.)
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, NO.38 Xueyuan Road, Beijing 100083, China
| | - Ying Lyu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, NO.38 Xueyuan Road, Beijing 100083, China; (Y.Z.); (R.Z.); (Y.L.); (H.S.); (W.Y.); (Y.T.); (R.L.)
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, NO.38 Xueyuan Road, Beijing 100083, China
| | - Hanxu Shi
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, NO.38 Xueyuan Road, Beijing 100083, China; (Y.Z.); (R.Z.); (Y.L.); (H.S.); (W.Y.); (Y.T.); (R.L.)
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, NO.38 Xueyuan Road, Beijing 100083, China
| | - Wanyun Ye
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, NO.38 Xueyuan Road, Beijing 100083, China; (Y.Z.); (R.Z.); (Y.L.); (H.S.); (W.Y.); (Y.T.); (R.L.)
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, NO.38 Xueyuan Road, Beijing 100083, China
| | - Yuwei Tan
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, NO.38 Xueyuan Road, Beijing 100083, China; (Y.Z.); (R.Z.); (Y.L.); (H.S.); (W.Y.); (Y.T.); (R.L.)
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, NO.38 Xueyuan Road, Beijing 100083, China
| | - Rui Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, NO.38 Xueyuan Road, Beijing 100083, China; (Y.Z.); (R.Z.); (Y.L.); (H.S.); (W.Y.); (Y.T.); (R.L.)
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, NO.38 Xueyuan Road, Beijing 100083, China
| | - Yajun Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, NO.38 Xueyuan Road, Beijing 100083, China; (Y.Z.); (R.Z.); (Y.L.); (H.S.); (W.Y.); (Y.T.); (R.L.)
- PKUHSC-China Feihe Joint Research Institute of Nutrition and Healthy Lifespan Development, NO.38 Xueyuan Road, Beijing 100083, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, NO.38 Xueyuan Road, Beijing 100083, China
| |
Collapse
|
35
|
Zhong C, Miao M, Che B, Du J, Wang A, Peng H, Bu X, Zhang J, Ju Z, Xu T, He J, Zhang Y. Plasma choline and betaine and risks of cardiovascular events and recurrent stroke after ischemic stroke. Am J Clin Nutr 2021; 114:1351-1359. [PMID: 34159355 DOI: 10.1093/ajcn/nqab199] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/25/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Choline and betaine have been suggested to play a pivotal role in neurotransmitter synthesis, cell membrane integrity, and methyl-group metabolism, exerting neuroprotective effects in patients with various neurological disorders. However, population-based evidence on choline and betaine with subsequent cardiovascular events after stroke is rare. OBJECTIVES We aimed to prospectively investigate the relationships of circulating choline and betaine with cardiovascular events and recurrent stroke in patients with ischemic stroke. METHODS We performed a nested case-control study within the China Antihypertensive Trial in Acute Ischemic Stroke. A total of 323 cardiovascular events (including 264 recurrent strokes) and 323 controls (free of recurrent cardiovascular events) matched for age (±1 y), sex, and treatment group were included. The primary endpoint was a composite of cardiovascular events after ischemic stroke. Plasma choline and betaine were measured at baseline by ultra-high-performance LC-MS/MS. Conditional logistic regression models were applied, and discrimination, reclassification, and calibration of models with choline pathway metabolites were evaluated. RESULTS Plasma choline and betaine were inversely associated with cardiovascular events and recurrent stroke after ischemic stroke. Specifically, in fully adjusted models, each additional SD of choline and betaine was associated with 35% (95% CI: 20%-48%) and 30% (95% CI: 14%-43%) decreased risks of subsequent cardiovascular events, respectively, and 34% (95% CI: 16%-48%) and 29% (95% CI: 12%-43%) decreased risks of recurrent stroke, respectively. In addition, both choline and betaine offered substantial risk discrimination and reclassification improvement for cardiovascular events and recurrent stroke beyond traditional risk factors, as evidenced by an increase in C statistics, the net reclassification index, and integrated discrimination improvement. CONCLUSIONS Plasma choline pathway metabolites, including choline and betaine, were associated with decreased risks of cardiovascular events and recurrent stroke and provided incremental value in risk discrimination and stratification in patients with ischemic stroke. This nested case-control study was based on the China Antihypertensive Trial in Acute Ischemic Stroke, which is registered at clinicaltrials.gov as NCT01840072.
Collapse
Affiliation(s)
- Chongke Zhong
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Mengyuan Miao
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Bizhong Che
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Jigang Du
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Aili Wang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Hao Peng
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Xiaoqing Bu
- Department of Epidemiology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Jintao Zhang
- Department of Neurology, The 88th Hospital of PLA, Shandong, China
| | - Zhong Ju
- Department of Neurology, Kerqin District First People's Hospital of Tongliao City, Tongliao, China
| | - Tan Xu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
36
|
Chen S, Wu Q, Zhu L, Zong G, Li H, Zheng H, Zeng R, Lin X, Sun L. Plasma glycerophospholipid profile, erythrocyte n-3 PUFAs, and metabolic syndrome incidence: a prospective study in Chinese men and women. Am J Clin Nutr 2021; 114:143-153. [PMID: 33829226 DOI: 10.1093/ajcn/nqab050] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 02/09/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Animal studies have highlighted critical roles of glycerophospholipid (GP) metabolism in various metabolic syndrome (MetS)-related features such as dyslipidemia, obesity, and insulin resistance. However, human prospective studies of associations between circulating GPs and risks of MetS are scarce. OBJECTIVES We aimed to investigate whether GPs are associated with incidence of MetS in a well-established cohort. METHODS A total of 1243 community-dwelling Chinese aged 50-70 y without MetS at baseline and followed up for 6 y were included in current analyses. A total of 145 plasma GPs were quantified by high-throughput targeted lipidomics. MetS was defined using the updated National Cholesterol Education Program Adult Treatment Panel III criteria for Asian Americans. RESULTS After 6 y, 429 participants developed MetS. Eleven GPs, especially those with long-chain polyunsaturated fatty acids (LCPUFAs) or very-long-chain polyunsaturated fatty acids (VLCPUFAs) at the sn-2 position, including 1 phosphatidylcholine (PC) [PC(18:0/22:6)], 9 phosphatidylethanolamines (PEs) [PE(16:0/22:6), PE(18:0/14:0), PE(18:0/18:1), PE(18:0/18:2), PE(18:0/20:3), PE(18:0/22:5), PE(18:0/22:6), PE(18:1/22:6), and PE(18:2/22:6)], and 1 phosphatidylserine (PS) [PS(18:0/18:0)], were positively associated with incident MetS (RRs: 1.16-1.30 per SD change; Bonferroni-corrected P < 0.05). In network analysis, the strongest positive association for MetS incidence was evidenced in a module mainly composed of PEs containing C22:6 and PSs [RR: 1.21; 95% CI: 1.12, 1.31 per SD change; Bonferroni-corrected P < 0.05]. This association was more pronounced in participants with lower erythrocyte total n-3 PUFA concentrations [Bonferroni-corrected Pinter(P value for the interaction)< 0.05]. CONCLUSIONS Elevated plasma concentrations of GPs, especially PEs with LCPUFAs or VLCPUFAs at the sn-2 position, are associated with higher risk of incident MetS. Future studies are merited to confirm our findings.
Collapse
Affiliation(s)
- Shuangshuang Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qingqing Wu
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Li Zhu
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Geng Zong
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Huaixing Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - He Zheng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rong Zeng
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.,Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China
| | - Xu Lin
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China.,Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Liang Sun
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
37
|
Hampel D, Shahab-Ferdows S, Nguyen N, Kac G, Allen LH. High-Throughput Analysis of Water-Soluble Forms of Choline and Related Metabolites in Human Milk by UPLC-MS/MS and Its Application. Front Nutr 2021; 7:604570. [PMID: 33614690 PMCID: PMC7892616 DOI: 10.3389/fnut.2020.604570] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/21/2020] [Indexed: 12/20/2022] Open
Abstract
Choline and related metabolites are key factors in many metabolic processes, and insufficient supply can adversely affect reproduction and fetal development. Choline status is mainly regulated by intake, and human milk is the only choline source for exclusively breastfed infants. Further, maternal status, genotype, and phenotype, as well as infant outcomes, have been related to milk choline concentrations. In order to enable the rapid assessment of choline intake for exclusively breastfed infants and to further investigate the associations between milk choline and maternal and infant status and other outcomes, we have developed a simplified method for the simultaneous analysis of human milk choline, glycerophosphocholine, phosphocholine, and the less abundant related metabolites betaine, carnitine, creatinine, dimethylglycine (DMG), methionine, and trimethylamine N-oxide (TMAO) using ultraperformance liquid chromatography–tandem mass spectrometry (UPLC–MS/MS). These analytes have milk concentrations ranging over 3 orders of magnitude. Unlike other recently described LC-based methods, our approach does not require an ion-pairing reagent or high concentrations of solvent modifiers for successful analyte separation and thus avoid signal loss and potential permanent contamination. Milk samples (10 μl) were diluted (1:80) in water : methanol (1:4, v:v) and filtered prior to analysis with an optimized gradient of 0.1% propionic acidaq and acetonitrile, allowing efficient separation and removal of contaminants. Recovery rates ranged from 108.0 to 130.9% (inter-day variation: 3.3–9.6%), and matrix effects (MEs) from 54.1 to 114.3%. MEs were greater for carnitine, creatinine, and TMAO at lower dilution (1:40, p < 0.035 for all), indicating concentration-dependent ion suppression. Milk from Brazilian women (2–8, 28–50, and 88–119 days postpartum, ntotal = 53) revealed increasing concentration throughout lactation for glycerophosphocholine, DMG, and methionine, while carnitine decreased. Choline and phosphocholine were negatively correlated consistently at all three collection time intervals. The method is suitable for rapid analysis of human milk water-soluble forms of choline as well as previously not captured related metabolites with minimal sample volumes and preparation.
Collapse
Affiliation(s)
- Daniela Hampel
- United States Department of Agriculture/Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, United States.,Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Setareh Shahab-Ferdows
- United States Department of Agriculture/Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, United States
| | - Ngoc Nguyen
- United States Department of Agriculture/Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, United States.,Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Gilberto Kac
- Nutrition Institute, Rio de Janeiro Federal University, Rio de Janeiro, Brazil
| | - Lindsay H Allen
- United States Department of Agriculture/Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, United States.,Department of Nutrition, University of California, Davis, Davis, CA, United States
| |
Collapse
|
38
|
Mehmood A, Zhao L, Wang Y, Pan F, Hao S, Zhang H, Iftikhar A, Usman M. Dietary anthocyanins as potential natural modulators for the prevention and treatment of non-alcoholic fatty liver disease: A comprehensive review. Food Res Int 2021; 142:110180. [PMID: 33773656 DOI: 10.1016/j.foodres.2021.110180] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 01/18/2021] [Accepted: 01/24/2021] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) refers to a metabolic syndrome linked with type 2 diabetes mellitus, obesity, and cardiovascular diseases. It is characterized by the accumulation of triglycerides in the hepatocytes in the absence of alcohol consumption. The prevalence of NAFLD has abruptly increased worldwide, with no effective treatment yet available. Anthocyanins (ACNs) belong to the flavonoid subclass of polyphenols, are commonly present in various edible plants, and possess a broad array of health-promoting properties. ACNs have been shown to have strong potential to combat NAFLD. We critically assessed the literature regarding the pharmacological mechanisms and biopharmaceutical features of the action of ACNs on NAFLD in humans and animal models. We found that ACNs ameliorate NAFLD by improving lipid and glucose metabolism, increasing antioxidant and anti-inflammatory activities, and regulating gut microbiota dysbiosis. In conclusion, ACNs have potential to attenuate NAFLD. However, further mechanistic studies are required to confirm these beneficial impacts of ACNs on NAFLD.
Collapse
Affiliation(s)
- Arshad Mehmood
- Beijing Advance Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Lei Zhao
- Beijing Advance Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China.
| | - Yong Wang
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Fei Pan
- Beijing Advance Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Shuai Hao
- Beijing Advance Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Huimin Zhang
- Beijing Advance Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Asra Iftikhar
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, The University of Faisalabad, Faisalabad 38000, Pakistan
| | - Muhammad Usman
- Beijing Advance Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
39
|
Zhang H, Léveillé M, Courty E, Gunes A, N Nguyen B, Estall JL. Differences in metabolic and liver pathobiology induced by two dietary mouse models of nonalcoholic fatty liver disease. Am J Physiol Endocrinol Metab 2020; 319:E863-E876. [PMID: 32924526 DOI: 10.1152/ajpendo.00321.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a growing epidemic linked to metabolic disease. The first stage of NAFLD is characterized by lipid accumulation in hepatocytes, but this can progress into nonalcoholic steatohepatitis (NASH), cirrhosis, and hepatocellular carcinoma (HCC). Western diets, high in fats, sugars, and cholesterol, are linked to NAFLD development. Murine models are often used to study NAFLD; however, there remains debate on which diet-induced model best mimics both human disease progression and pathogenesis. In this study, we performed a side-by-side comparison of two popular diet models of murine NAFLD/NASH and associated HCC, a high-fat diet supplemented with 30% fructose water (HFHF) and a Western diet high in cholesterol (WDHC), and these were compared with a common grain-based chow diet (GBD). Mice on both experimental diets developed liver steatosis, and WDHC-fed mice had greater levels of hepatic inflammation and fibrosis than HFHF-fed mice. In contrast, HFHF-fed mice were more obese and developed more severe metabolic syndrome, with less pronounced liver disease. Despite these differences, WDHC-fed and HFHF-fed mice had similar tumor burdens in a model of diet-potentiated liver cancer. Response to diet and resulting phenotypes were generally similar between sexes, albeit delayed in females. This study shows that modest differences in diet can significantly uncouple glucose homeostasis and liver damage. In conclusion, long-term feeding of either HFHF or WDHC is a reliable method to induce NASH and diet-potentiated liver cancer in mice of both sexes; however, the choice of diet involves a trade-off between severity of metabolic syndrome and liver damage.
Collapse
Affiliation(s)
- Hannah Zhang
- Institut de recherches cliniques de Montréal (IRCM), Montreal, Quebec, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Mélissa Léveillé
- Institut de recherches cliniques de Montréal (IRCM), Montreal, Quebec, Canada
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Emilie Courty
- Institut de recherches cliniques de Montréal (IRCM), Montreal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Aysim Gunes
- Institut de recherches cliniques de Montréal (IRCM), Montreal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Bich N Nguyen
- Department of Pathology and Cell Biology, University of Montreal, Montreal, Quebec, Canada
- University of Montreal Health Network (CHUM), Montreal, Quebec Canada
| | - Jennifer L Estall
- Institut de recherches cliniques de Montréal (IRCM), Montreal, Quebec, Canada
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
40
|
Dibaba DT, Johnson KC, Kucharska-Newton AM, Meyer K, Zeisel SH, Bidulescu A. The Association of Dietary Choline and Betaine With the Risk of Type 2 Diabetes: The Atherosclerosis Risk in Communities (ARIC) Study. Diabetes Care 2020; 43:2840-2846. [PMID: 32900787 PMCID: PMC7576425 DOI: 10.2337/dc20-0733] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/11/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To examine the association between dietary intake of choline and betaine and the risk of type 2 diabetes. RESEARCH DESIGN AND METHODS Among 13,440 Atherosclerosis Risk in Communities (ARIC) study participants, the prospective longitudinal association between dietary choline and betaine intake and the risk of type 2 diabetes was assessed using interval-censored Cox proportional hazards and logistic regression models adjusted for baseline potential confounding variables. RESULTS Among 13,440 participants (55% women, mean age 54 [SD 7.4] years), 1,396 developed incident type 2 diabetes during median follow-up of 9 years from 1987 to 1998. There was no statistically significant association between every 1-SD increase in dietary choline and risk of type 2 diabetes (hazard ratio [HR] 1.01 [95% CI 0.87, 1.16]) nor between dietary betaine intake and the risk of type 2 diabetes (HR 1.01 [0.94, 1.10]). Those in the highest quartile of dietary choline intake did not have a statistically significant higher risk of type 2 diabetes than those in the lowest choline quartile (HR 1.09 [0.84, 1.42]); similarly, dietary betaine intake was not associated with the risk of type 2 diabetes comparing the highest quartile to the lowest (HR 1.06 [0.87, 1.29]). Among women, there was a higher risk of type 2 diabetes, comparing the highest to lowest dietary choline quartile (HR 1.54 [1.06, 2.25]), while in men, the association was null (HR 0.82 [0.57, 1.17]). Nevertheless, there was a nonsignificant interaction between high choline intake and sex on the risk of type 2 diabetes (P = 0.07). The results from logistic regression were similar. CONCLUSIONS Overall and among male participants, dietary choline or betaine intakes were not associated with the risk of type 2 diabetes. Among female participants, there was a trend for a modestly higher risk of type 2 diabetes among those with the highest as compared with the lowest quartile of dietary choline intake. Our study should inform clinical trials on dietary choline and betaine supplementation in relationship with the risk of type 2 diabetes.
Collapse
Affiliation(s)
- Daniel T Dibaba
- Tennessee Clinical and Translational Science Institute, University of Tennessee Health Science Center, Memphis, TN.,Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Karen C Johnson
- Tennessee Clinical and Translational Science Institute, University of Tennessee Health Science Center, Memphis, TN.,Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Anna M Kucharska-Newton
- Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY.,Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Katie Meyer
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Steven H Zeisel
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC
| | - Aurelian Bidulescu
- Department of Epidemiology and Biostatistics, School of Public Health-Bloomington, Indiana University, Bloomington, IN
| |
Collapse
|
41
|
Longo L, Tonin Ferrari J, Rampelotto PH, Hirata Dellavia G, Pasqualotto A, P Oliveira C, Thadeu Schmidt Cerski C, Reverbel da Silveira T, Uribe-Cruz C, Álvares-da-Silva MR. Gut Dysbiosis and Increased Intestinal Permeability Drive microRNAs, NLRP-3 Inflammasome and Liver Fibrosis in a Nutritional Model of Non-Alcoholic Steatohepatitis in Adult Male Sprague Dawley Rats. Clin Exp Gastroenterol 2020; 13:351-368. [PMID: 32982365 PMCID: PMC7509481 DOI: 10.2147/ceg.s262879] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND/AIM The interactions between the gut and liver have been described in the progression of non-alcoholic steatohepatitis (NASH). The aim of this study was to develop an experimental nutritional model of NASH simulating metabolic changes occurring in humans. MATERIALS AND METHODS Adult male Sprague Dawley rats were randomized into two groups: controls (standard diet) and intervention (high-fat and choline-deficient diet) for 16 weeks, each experimental group with 10 animals. Biochemical analysis, hepatic lipid content, microRNAs, inflammatory, gut permeability markers and gut microbiota were measured. RESULTS Animals in the intervention group showed significantly higher delta Lee index (p=0.017), abdominal circumference (p<0.001), abdominal adipose tissue (p<0.001) and fresh liver weight (p<0.001), as well as higher serum levels of alanine aminotransferase (p=0.010), glucose (p=0.013), total cholesterol (p=0.033), LDL cholesterol (p=0.011), and triglycerides (p=0.011), and lower HDL cholesterol (p=0.006) compared to the control group. Higher TLR4 (p=0.041), TLR9 (p=0.033), MyD88 (p=0.001), Casp1 (p<0.001), NLPR3 (p=0.019), liver inflammation index interleukin (IL)-1β/IL10 (p<0.001), IL6/IL10 (p=0.002) and TNFα/IL10 (p=0.001) were observed in the intervention group, and also lower permeability markers Ocln (p=0.003) and F11r (p=0.041). Gene expression of miR-122 increased (p=0.041) and miR-145 (p=0.010) decreased in the intervention group. Liver steatosis, inflammation and fibrosis, along with collagen fiber deposition increment (p<0.001), were seen in the intervention group. Regarding gut microbiota, Bray-Curtis dissimilarity index and number of operational taxonomic units were significantly different (p<0.001) between the groups. Composition of the gut microbiota showed a significant correlation with histopathological score of NAFLD (r=0.694) and index IL-1β/IL-10 (r=0.522). CONCLUSION This experimental model mimicking human NASH demonstrated gut and liver interaction, with gut microbiota and intestinal permeability changes occurring in parallel with systemic and liver inflammation, miRNAs regulation and liver tissue damage.
Collapse
Affiliation(s)
- Larisse Longo
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jéssica Tonin Ferrari
- Experimental Laboratory of Hepatology and Gastroenterology, Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Pabulo Henrique Rampelotto
- Experimental Laboratory of Hepatology and Gastroenterology, Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Graduate Program in Pharmaceutical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Gustavo Hirata Dellavia
- School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Amanda Pasqualotto
- Experimental Laboratory of Hepatology and Gastroenterology, Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Claudia P Oliveira
- Department of Gastroenterology (LIM07), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Carlos Thadeu Schmidt Cerski
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Unit of Surgical Pathology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Themis Reverbel da Silveira
- Experimental Laboratory of Hepatology and Gastroenterology, Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Carolina Uribe-Cruz
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Mário Reis Álvares-da-Silva
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Division of Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
42
|
Sarkar S, Bhattacharya S, Alam MJ, Yadav R, Banerjee SK. Hypoxia aggravates non-alcoholic fatty liver disease in presence of high fat choline deficient diet: A pilot study. Life Sci 2020; 260:118404. [PMID: 32920003 DOI: 10.1016/j.lfs.2020.118404] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/31/2020] [Accepted: 09/06/2020] [Indexed: 12/21/2022]
Abstract
AIM NAFLD is a chronic and progressive disease for which there are no FDA-approved drugs available in the market. Drug discovery is a time-consuming procedure and requires screening of hundreds of small molecules to find new chemical entities (NECs) for a particular disease. Current preclinical NAFLD animal models take a longer time, which enhances the duration and expenses of the screening procedure. Hence to shorten the duration, we have proposed a preclinical animal model for rapid induction of non-alcoholic steatohepatitis (NASH), an advanced stage of NAFLD in rats. METHODOLOGY The animals were divided into three groups; control, high fat choline deficient (HFCD) and high fat choline deficient diet with sodium nitrite (40 mg/kg b.w. i.p. per day) (HFCD + NaNO2) respectively. Four weeks later physical and serum biochemical parameters were assessed, intraperitoneal glucose tolerance test was performed, and histopathology and gene expression were analysed. KEY FINDINGS Hypoxic stress aggravates the lipid accumulation, ballooning, lobular inflammation and fibrosis in hepatic tissue in presence of HFCD diet. SIGNIFICANCE This novel rodent model could be a useful NAFLD model to screen small molecules rapidly for treatment of NASH.
Collapse
Affiliation(s)
- Soumalya Sarkar
- Non-communicable Disease Group, Translational Health Science and Technology Institute (THSTI), Faridabad 121001, Haryana, India
| | - Sankarsan Bhattacharya
- Non-communicable Disease Group, Translational Health Science and Technology Institute (THSTI), Faridabad 121001, Haryana, India
| | - Md Jahangir Alam
- Non-communicable Disease Group, Translational Health Science and Technology Institute (THSTI), Faridabad 121001, Haryana, India
| | - Rajni Yadav
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Sanjay K Banerjee
- Non-communicable Disease Group, Translational Health Science and Technology Institute (THSTI), Faridabad 121001, Haryana, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, Assam, India.
| |
Collapse
|
43
|
Long-Term Ketogenic Diet Induces Metabolic Acidosis, Anemia, and Oxidative Stress in Healthy Wistar Rats. J Nutr Metab 2020; 2020:3642035. [PMID: 32685205 PMCID: PMC7341377 DOI: 10.1155/2020/3642035] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/19/2020] [Accepted: 05/21/2020] [Indexed: 12/27/2022] Open
Abstract
Background Ketogenic diet has been used as supportive therapy in a range of conditions including epilepsy, diabetes mellitus, and cancer. Objective This study aimed to investigate the effects of long-term consumption of ketogenic diet on blood gas, hematological profiles, organ functions, and superoxide dismutase level in a rat model. Materials and Methods Fifteen male Wistar rats were divided into control (n = 8) and ketogenic (n = 7) groups. Controls received standard diet contained 52.20% of carbohydrates, 7.00% fat, and 15.25% protein; meanwhile, the ketogenic group received a high-fat-low-carbohydrate diet which contained 5.66% of carbohydrate, 86.19% fat, and 8.15% protein. All rats were caged individually and received 30g of either standard or high-fat-low-carbohydrate pellets. The experiment was carried out for 60 days before the blood samples were taken and analyzed to obtain blood gas, cell counts, organ biomarkers, and plasma antioxidant superoxide dismutase (SOD) levels. Results The rats subjected to ketogenic diet experienced a marked decrease in body weight, blood sugar, and increased blood ketones (p < 0.05). The average blood pH was 7.36 ± 0.02 and base excess was −5.57 ± 2.39 mOsm/L, which were significantly lower than controls (p < 0.05). Hematological analysis showed significantly lower erythrocyte, hemoglobin, and hematocrit levels. No significant changes were found in alanine aminotransferase, aspartate aminotransferase, urea, and creatinine levels, indicating normal liver and kidney functions. Nevertheless, plasma SOD level significantly reduced with ketogenic diet. Conclusion Long-term ketogenic diet induces metabolic acidosis, anemia, and reduced antioxidant enzyme level in rats following 60 days of consuming high-fat-low-carbohydrate diet.
Collapse
|
44
|
Prabhu GS, K G Rao M, Rai KS. Hippocampal neural cell degeneration and memory deficit in high-fat diet-induced postnatal obese rats- exploring the comparable benefits of choline and DHA or environmental enrichment. Int J Neurosci 2020; 131:1066-1077. [PMID: 32498586 DOI: 10.1080/00207454.2020.1773819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Purpose: Childhood obesity increases risk for neural dysfunctions causing learning and memory deficits. The objective of the study is to identify the effects of high fat diet-induced obesity in postnatal period on serum lipids, memory and neural cell survival in hippocampus and compare the role of choline and DHA or environmental enrichment in attenuating the alterationsMaterials and methods: 21 day postnatal male Sprague Dawley rats were assigned as Normal control [NC] fed normal chow diet, Obesity-induced [OB] fed high fat diet, Obesity-induced fed choline & DHA [OB + CHO + DHA], Obesity-induced environmental enrichment [OB + EE] [n = 8/group]. Memory was assessed using radial arm maze. Subsequently blood was collected for serum lipid analysis and rats were euthanized. 5 µm hippocampal sections were processed for cresyl-violet stain. Surviving neural cells were counted using 100 µm scale.Results: Memory errors were significantly higher [p < 0.001, 0.01] in OB compared to same in NC rats. Mean number of surviving neural cells in hippocampus of OB was significantly lesser [p < 0.01] compared to same in NC. Interventions in OB + CHO + DHA and OB + EE significantly attenuated [p < 0.01] memory errors and number of surviving neural cells in hippocampus [CA1, CA3 and DG] compared to same in OB. Moreover, hippocampal neural cell survival was found to be inversely related to serum lipid profile in OB group and was attenuated in OB + CHO + DHA and OB + EE rats.Conclusions: High fat diet-induced postnatal obesity in rats causes CA1/CA3 hippocampal neuro-degeneration and memory deficits. Supplementation of choline and DHA in obese rats attenuates these deficits.
Collapse
Affiliation(s)
- Gayathri S Prabhu
- Department of Anatomy, Melaka Manipal Medical College [Manipal campus], Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mohandas K G Rao
- Department of Anatomy, Melaka Manipal Medical College [Manipal campus], Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kiranmai S Rai
- Department of Physiology, Melaka Manipal Medical College [Manipal campus], Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
45
|
Radhakrishnan S, Ke JY, Pellizzon MA. Targeted Nutrient Modifications in Purified Diets Differentially Affect Nonalcoholic Fatty Liver Disease and Metabolic Disease Development in Rodent Models. Curr Dev Nutr 2020; 4:nzaa078. [PMID: 32494762 PMCID: PMC7250583 DOI: 10.1093/cdn/nzaa078] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/16/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a complex spectrum of disorders ranging from simple benign steatosis to more aggressive forms of nonalcoholic steatohepatitis (NASH) and fibrosis. Although not every patient with NAFLD/NASH develops liver complications, if left untreated it may eventually lead to cirrhosis and hepatocellular carcinoma. Purified diets formulated with specific nutritional components can drive the entire spectrum of NAFLD in rodent models. Although they may not perfectly replicate the clinical and histological features of human NAFLD, they provide a model to gain further understanding of disease progression in humans. Owing to the growing demand of diets for NAFLD research, and for our further understanding of how manipulation of dietary components can alter disease development, we outlined several commonly used dietary approaches for rodent models, including mice, rats, and hamsters, time frames required for disease development and whether other metabolic diseases commonly associated with NAFLD in humans occur.
Collapse
Affiliation(s)
| | - Jia-Yu Ke
- Research Diets, Inc., New Brunswick, NJ, USA
| | | |
Collapse
|
46
|
Guarino M, Kumar P, Felser A, Terracciano LM, Guixé-Muntet S, Humar B, Foti M, Nuoffer JM, St-Pierre MV, Dufour JF. Exercise Attenuates the Transition from Fatty Liver to Steatohepatitis and Reduces Tumor Formation in Mice. Cancers (Basel) 2020; 12:E1407. [PMID: 32486073 PMCID: PMC7352494 DOI: 10.3390/cancers12061407] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/18/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) leads to steatohepatitis (NASH), fibrosis, and hepatocellular carcinoma. For sedentary patients, lifestyle interventions combining exercise and dietary changes are a cornerstone of treatment. However, the benefit of exercise alone when dietary changes have failed is uncertain. We query whether exercise alone arrests the progression of NASH and tumorigenesis in a choline-deficient, high-fat diet (CD-HFD) murine model. Male C57Bl/6N mice received a control diet or CD-HFD for 12 weeks. CD-HFD mice were randomized further for 8 weeks of sedentariness (SED) or treadmill exercise (EXE). CD-HFD for 12 weeks produced NAFL. After 20 weeks, SED mice developed NASH and hepatic adenomas. Exercise attenuated the progression to NASH. EXE livers showed lower triglycerides and tumor necrosis factor-α expression, less fibrosis, less ballooning, and a lower NAFLD activity score than did SED livers. Plasma transaminases and triglycerides were lower. Exercise activated AMP-activated protein kinase (AMPK) with inhibition of mTORC1 and decreased S6 phosphorylation, reducing hepatocellular adenoma. Exercise activated autophagy with increased LC3-II/LC3-I and mitochondrial recruitment of phosphorylated PTEN-induced kinase. Therefore, exercise attenuates the transition from NAFL to NASH, improves biochemical and histological parameters of NAFLD, and impedes the progression of fibrosis and tumorigenesis associated with enhanced activation of AMPK signaling and favors liver autophagy. Our work supports the benefits of exercise independently of dietary changes.
Collapse
Affiliation(s)
- Maria Guarino
- Hepatology, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland; (M.G.); (P.K.); (S.G.-M.); (M.V.S.-P.)
- Gastroenterology, Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy
| | - Pavitra Kumar
- Hepatology, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland; (M.G.); (P.K.); (S.G.-M.); (M.V.S.-P.)
| | - Andrea Felser
- University Institute of Clinical Chemistry, Bern University Hospital, 3010 Bern, Switzerland; (A.F.); (J.-M.N.)
| | | | - Sergi Guixé-Muntet
- Hepatology, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland; (M.G.); (P.K.); (S.G.-M.); (M.V.S.-P.)
| | - Bostjan Humar
- Laboratory of the Swiss Hepato-Pancreato-Biliary (HPB) and Transplantation Center, Department of Surgery, University Hospital Zürich, 8091 Zürich, Switzerland;
| | - Michelangelo Foti
- Department of Cell Physiology and Metabolism, University of Geneva, 1206 Geneva, Switzerland;
| | - Jean-Marc Nuoffer
- University Institute of Clinical Chemistry, Bern University Hospital, 3010 Bern, Switzerland; (A.F.); (J.-M.N.)
| | - Marie V. St-Pierre
- Hepatology, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland; (M.G.); (P.K.); (S.G.-M.); (M.V.S.-P.)
| | - Jean-François Dufour
- Hepatology, Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland; (M.G.); (P.K.); (S.G.-M.); (M.V.S.-P.)
- University Clinic of Visceral Surgery and Medicine, Inselspital Bern, 3010 Bern, Switzerland
| |
Collapse
|
47
|
A Role for Gut Microbiome Fermentative Pathways in Fatty Liver Disease Progression. J Clin Med 2020; 9:jcm9051369. [PMID: 32392712 PMCID: PMC7291163 DOI: 10.3390/jcm9051369] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/24/2020] [Accepted: 05/03/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifactorial disease in which environmental and genetic factors are involved. Although the molecular mechanisms involved in NAFLD onset and progression are not completely understood, the gut microbiome (GM) is thought to play a key role in the process, influencing multiple physiological functions. GM alterations in diversity and composition directly impact disease states with an inflammatory course, such as non-alcoholic steatohepatitis (NASH). However, how the GM influences liver disease susceptibility is largely unknown. Similarly, the impact of strategies targeting the GM for the treatment of NASH remains to be evaluated. This review provides a broad insight into the role of gut microbiota in NASH pathogenesis, as a diagnostic tool, and as a therapeutic target in this liver disease. We highlight the idea that the balance in metabolic fermentations can be key in maintaining liver homeostasis. We propose that an overabundance of alcohol-fermentation pathways in the GM may outcompete healthier, acid-producing members of the microbiota. In this way, GM ecology may precipitate a self-sustaining vicious cycle, boosting liver disease progression.
Collapse
|
48
|
Gut microbiota and human NAFLD: disentangling microbial signatures from metabolic disorders. Nat Rev Gastroenterol Hepatol 2020; 17:279-297. [PMID: 32152478 DOI: 10.1038/s41575-020-0269-9] [Citation(s) in RCA: 666] [Impact Index Per Article: 133.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/21/2020] [Indexed: 02/07/2023]
Abstract
Gut microbiota dysbiosis has been repeatedly observed in obesity and type 2 diabetes mellitus, two metabolic diseases strongly intertwined with non-alcoholic fatty liver disease (NAFLD). Animal studies have demonstrated a potential causal role of gut microbiota in NAFLD. Human studies have started to describe microbiota alterations in NAFLD and have found a few consistent microbiome signatures discriminating healthy individuals from those with NAFLD, non-alcoholic steatohepatitis or cirrhosis. However, patients with NAFLD often present with obesity and/or insulin resistance and type 2 diabetes mellitus, and these metabolic confounding factors for dysbiosis have not always been considered. Patients with different NAFLD severity stages often present with heterogeneous lesions and variable demographic characteristics (including age, sex and ethnicity), which are known to affect the gut microbiome and have been overlooked in most studies. Finally, multiple gut microbiome sequencing tools and NAFLD diagnostic methods have been used across studies that could account for discrepant microbiome signatures. This Review provides a broad insight into microbiome signatures for human NAFLD and explores issues with disentangling these signatures from underlying metabolic disorders. More advanced metagenomics and multi-omics studies using system biology approaches are needed to improve microbiome biomarkers.
Collapse
|
49
|
Liang Y, Liu D, Zhan J, Luo M, Han J, Wang P, Zhou Z. New insight into the mechanism of POP-induced obesity: Evidence from DDE-altered microbiota. CHEMOSPHERE 2020; 244:125123. [PMID: 32050320 DOI: 10.1016/j.chemosphere.2019.125123] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/27/2019] [Accepted: 10/13/2019] [Indexed: 06/10/2023]
Abstract
Although epidemiological studies demonstrate that persistent organic pollutants (POPs) could lead to metabolic syndrome, the mechanism has remained unclear. The dysbiosis of gut microbiota and the lipid metabolome have been put forward in the pathophysiology of metabolic syndrome. In this study, we used dichlorodiphenyldichloroethylene (DDE) as an example to study the effects of POP-impaired microbial composition and metabolome homeostasis on metabolic syndrome. The results showed that DDE exposure increased body weight and fat content and impaired glucose homeostasis. Further investigation revealed that DDE induced gut dysbiosis as indicated by an increased Firmicutes-to-Bacteroidetes ratio, which may impact energy harvest efficiency. Meanwhile, the plasma lipid metabolome profile was significantly altered by DDE. Furthermore, phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, and triacylglycerol were identified as key metabolites affected by DDE treatment, and these altered lipid metabolites were highly correlated with changed microbiota composition. This study provides novel insight into the underlying mechanism of POP-induced obesity and diabetes, pointing to gut microbiota as one of the targets.
Collapse
Affiliation(s)
- Yiran Liang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, PR China; College of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30, Xueyuan Road, Beijing, 100083, PR China
| | - Donghui Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Jing Zhan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Mai Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Jiajun Han
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Peng Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, PR China
| | - Zhiqiang Zhou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing, 100193, PR China.
| |
Collapse
|
50
|
NAFLD Preclinical Models: More than a Handful, Less of a Concern? Biomedicines 2020; 8:biomedicines8020028. [PMID: 32046285 PMCID: PMC7167756 DOI: 10.3390/biomedicines8020028] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/01/2020] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a spectrum of liver diseases ranging from simple steatosis to non-alcoholic steatohepatitis, fibrosis, cirrhosis, and/or hepatocellular carcinoma. Due to its increasing prevalence, NAFLD is currently a major public health concern. Although a wide variety of preclinical models have contributed to better understanding the pathophysiology of NAFLD, it is not always obvious which model is best suitable for addressing a specific research question. This review provides insights into currently existing models, mainly focusing on murine models, which is of great importance to aid in the identification of novel therapeutic options for human NAFLD.
Collapse
|