1
|
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
|
2
|
Wei L, Bai J, Zhang Y, Suo H, Wang C. Comparison of in vitro fermentation characteristics of carob gum and guar gum. Int J Biol Macromol 2025; 311:143886. [PMID: 40319966 DOI: 10.1016/j.ijbiomac.2025.143886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 04/27/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Carob gum (CG) and guar gum (GG) are widely used as additives in food processing; however, their fermentation properties in the human gut and potential effects on the human body are unclear. This study used an in vitro fermentation model to evaluate the interaction of CG and GG with intestinal flora and compare their fermentation characteristics. The results showed that CG and GG could be degraded and utilized by the intestinal flora. GG can increase the production of acetic acid, propionic acid, and butyric acid and the relative abundance of beneficial microorganisms such as Prevotella and Faecalibacterium. CG promotes propionic acid production and the relative abundance of beneficial microorganisms such as Bifidobacterium and Lactobacillus. Metabolomic studies have shown that fermented CG and GG mainly affect human metabolic pathways. GG promotes amino acid and lipid metabolism, and CG promotes amino acid metabolism and biosynthesis of secondary metabolites. This research shows that despite significant differences in how CG and GG interact with the gut microbiota, both may affect the human body.
Collapse
Affiliation(s)
- Li Wei
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Junying Bai
- Citrus Research Institute, Southwest University, Chongqing 400700, China
| | - Yuyan Zhang
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Chen Wang
- College of Food Science, Southwest University, Chongqing 400715, China.
| |
Collapse
|
3
|
Liu J, Deng L, Yao B, Zhang Y, Huang J, Huang S, Liang C, Shen Y, Wang X. Carboxylesterase 2A gene knockout or enzyme inhibition alleviates steatohepatitis in rats by regulating PPARγ and endoplasmic reticulum stress. Free Radic Biol Med 2025; 232:279-291. [PMID: 40089078 DOI: 10.1016/j.freeradbiomed.2025.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/09/2025] [Accepted: 03/13/2025] [Indexed: 03/17/2025]
Abstract
Metabolic dysfunction associated steatotic liver disease (MASLD) is a widespread liver disease that progresses from simple steatosis to severe steatohepatitis stage. Despite the recognized importance of carboxylesterase 2 (CES2) in hepatic lipid metabolism, the role of CES2 in hepatic inflammation remains unclear. The rat genome encodes six Ces2 genes and Ces2a shows high expression in the liver and intestine. Lipid metabolism, inflammation, fibrosis, and endoplasmic reticulum (ER) stress were investigated in Ces2a knockout (KO) rats. KO rats showed spontaneous liver lipid accumulation due to increased lipogenesis and reduced fatty acid oxidation. Non-targeted lipidomic analysis revealed enhanced lysophosphatidylcholines (LPCs) and phosphatidylcholines (PCs) in KO rats and increased concentrations of ligands, thus activating the expression of PPARγ. Although there was simple lipid accumulation in the liver of KO rats, Ces2a deficiency showed a significant protective effect against LPS and diet-induced hepatic steatohepatitis by inhibiting ER stress regulated by PPARγ activation. In line with this, treatment with tanshinone IIA, a CES2 inhibitor, significantly alleviated the progression of steatohepatitis induced by the MCD diet. In conclusion, the increased PPARγ expression in Ces2a deficiency may counteract liver inflammation and ER stress despite the presence of simple steatosis. Therefore, CES2 inhibition represents a potential therapeutic approach for steatohepatitis.
Collapse
Affiliation(s)
- Jie Liu
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Luyao Deng
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Bingyi Yao
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Yuanjin Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Junze Huang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Shengbo Huang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Chenmeizi Liang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Yifei Shen
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Xin Wang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China.
| |
Collapse
|
4
|
Xue Y, Xu C, Lv M, Li J, Wen K, Geng T, Gong D, Liu L, Fallahshahroudi A, Zheng Y. PRIAM1 participates in the inhibition of inflammation and acetylcholinesterase activity in goose fatty liver formation. Poult Sci 2025; 104:105219. [PMID: 40344705 DOI: 10.1016/j.psj.2025.105219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/11/2025] Open
Abstract
Goose fatty liver, a product of short-term overfeeding, is notable for its high nutritional value and unique tolerance to severe steatosis without inflammation, in contrast to human nonalcoholic fatty liver disease (NAFLD). It is known that choline can alleviate nonalcoholic steatohepatitis and liver cirrhosis, inhibit cell apoptosis, promote lecithin synthesis and fat transportation out of the liver, and relieve cardiovascular disease-related symptoms (e.g., hyperlipidemia and hypercholesterolemia). This study investigates the role of Proline Rich Membrane Anchor 1 (PRIMA1) in inhibiting inflammation through choline metabolism during goose fatty liver formation. Overfeeding of geese resulted in increased body and liver weights, elevated fat content, and significantly higher expression of PRIMA1, accompanied by decreased LITAF and CRP (important pro-inflammatory cytokines) expression and reduced acetylcholinesterase (AchE) activity, which was assessed by the amount of produced choline. The overexpression of PRIMA1 in goose primary hepatocytes (GPHs) enhanced AchE activity. Consistently, glucose-induced upregulation of PRIMA1 expression in GPHs was accompanied by increased AchE activity. Further combined treatment with glucose and PRIMA1 knockdown in GPHs showed that downregulation of PRIMA1 attenuated the suppression of LITAF and CRP expression caused by glucose addition, but had no effect on the rise in AchE activity. These findings indicate that PRIMA1 may play a role in promoting choline metabolism and protecting against liver inflammation, offering insights into potential therapeutic targets for NAFLD.
Collapse
Affiliation(s)
- Ying Xue
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China.
| | - Cheng Xu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - Mengqing Lv
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - JiaHui Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - Kang Wen
- Yangzhou Animal Husbandry Veterinary and Fishery Technical Guidance Station, Yangzhou 225009PR China
| | - Tuoyu Geng
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - Daoqing Gong
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - Long Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China
| | - Amir Fallahshahroudi
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala 75256, Sweden.
| | - Yun Zheng
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China.
| |
Collapse
|
5
|
Chen L, Zhong J, Niu Y, Li C, Li J, Diao Z, Yan H, Xu M, Huang W, Xu Z, Su C, Liu D. Association of Dietary Choline Intake With Incidence of Frailty: A Nationwide Prospective Cohort Study From China. J Cachexia Sarcopenia Muscle 2025; 16:e13796. [PMID: 40189218 PMCID: PMC11972690 DOI: 10.1002/jcsm.13796] [Citation(s) in RCA: 0] [Impact Index Per Article: 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: 10/21/2024] [Revised: 02/12/2025] [Accepted: 02/27/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Emerging evidence suggests that dietary choline is a modifiable nutritional factor linked to various health outcomes. However, most existing studies have focused on isolated health conditions, lacking a comprehensive assessment of overall health status. This study aimed to investigate the association between total dietary choline intake and frailty incidence among Chinese adults, considering its derivatives, soluble forms (water-soluble and lipid-soluble) and food sources (animal-derived and plant-derived). METHODS Participants without frailty at baseline were enrolled from the China Health and Nutrition Survey (CHNS), with follow-up from 2004 to 2016. Dietary intake was assessed using three consecutive 24-h dietary recalls to estimate total dietary choline intake, its derivatives, soluble forms and food sources. Frailty status was evaluated using a frailty index (FI), with frailty defined as an FI > 0.21. Cox proportional hazards regression and restricted cubic splines were used to analyse the associations between dietary choline intake and frailty incidence. RESULTS A total of 10 310 participants (mean age: 46.4 years [SD: 14.5]; 52.6% female) were eligible. During a median follow-up of 6.1 years, 1150 incident frailty cases were recorded. Cox models with penalized splines showed an L-shaped association between total dietary choline intake and frailty incidence. Compared with participants in the lowest quartile of total choline intake, those in the 2nd to 4th quartiles had lower odds of frailty, with hazard ratios (HRs) of 0.84 (95% CI: 0.71, 0.98), 0.80 (95% CI: 0.67, 0.95) and 0.75 (95% CI: 0.61, 0.93), respectively. Intake of lipid-soluble choline in the 2nd to 4th quartiles was associated with an 18% (HR: 0.82; 95% CI: 0.69, 0.98) to 23% (HR: 0.77; 95% CI: 0.63, 0.95) reduction in the odds of frailty. Participants in the 3rd to 4th quartiles of phosphatidylcholine intake exhibited 19% (HR: 0.81; 95% CI: 0.68, 0.96) to 23% (HR: 0.77; 95% CI: 0.63, 0.94) lower odds of frailty. Choline intake from plant-derived food sources was significantly associated with reduced odds of frailty (HR: 0.83; 95% CI: 0.69, 0.99). CONCLUSIONS Moderate to high dietary choline intake (171.00-464.99 mg/day), particularly phosphatidylcholine (145.20-304.93 mg/day), may be associated with reduced odds of frailty.
Collapse
Affiliation(s)
- Lian‐hong Chen
- Department of Public Health and Preventive Medicine, School of MedicineJinan UniversityGuangzhouGuangdongChina
| | - Jian‐feng Zhong
- Department of Public Health and Preventive Medicine, School of MedicineJinan UniversityGuangzhouGuangdongChina
| | - Ying‐ying Niu
- Department of Public Health and Preventive Medicine, School of MedicineJinan UniversityGuangzhouGuangdongChina
| | - Cheng‐ping Li
- Department of Public Health and Preventive Medicine, School of MedicineJinan UniversityGuangzhouGuangdongChina
| | - Jing Li
- Department of Public Health and Preventive Medicine, School of MedicineJinan UniversityGuangzhouGuangdongChina
| | - Zhi‐quan Diao
- Department of Public Health and Preventive Medicine, School of MedicineJinan UniversityGuangzhouGuangdongChina
| | - Hao‐yu Yan
- Department of Public Health and Preventive Medicine, School of MedicineJinan UniversityGuangzhouGuangdongChina
| | - Miao Xu
- Department of Public Health and Preventive Medicine, School of MedicineJinan UniversityGuangzhouGuangdongChina
| | - Wen‐qi Huang
- Department of Public Health and Preventive Medicine, School of MedicineJinan UniversityGuangzhouGuangdongChina
| | - Zhi‐tong Xu
- Department of Public Health and Preventive Medicine, School of MedicineJinan UniversityGuangzhouGuangdongChina
| | - Chang Su
- National Institute for Nutrition and HealthChinese Center for Disease Control and PreventionBeijingChina
- Key Laboratory of Trace Element NutritionNational Health CommissionBeijingChina
| | - Dan Liu
- Department of Epidemiology, School of Public HealthSouthern Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
6
|
Yu M, Liu G, Chen W, Qiu Y, You N, Chen S, Wei Z, Ji L, Han M, Qin Z, Sun T, Wang D. Choline metabolism in ischemic stroke: An underappreciated "two-edged sword". Pharmacol Res 2025; 214:107685. [PMID: 40054542 DOI: 10.1016/j.phrs.2025.107685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 03/23/2025]
Abstract
Ischemic stroke (IS) is an important cause of death and disability worldwide, but the molecular mechanisms involved are not fully understood. In this context, choline metabolism plays an increasingly important role in IS due to its multifaceted mechanisms involving neuroprotection, neuroregeneration, inflammatory response, immune regulation, and long-term health effects. With the deepening of the research on choline and its metabolites, such as trimethylamine-N-oxide (TMAO), scientists have gradually realized its key role in the occurrence, development and potential treatment of IS. This review summarizes the importance of choline in neuroprotection and long-term disease management, highlighting the complexity of choline metabolism affecting cerebrovascular health through gut microbes. Although choline and its metabolites exhibit a protective effect, excessive intake and increases in some metabolites may confer risk, suggesting the need to carefully balance dietary choline intake. The purpose of this review is to integrate the existing research results and provide a theoretical basis for further exploring the mechanism, prognosis evaluation and clinical intervention of choline metabolism in ischemic IS, hoping to provide a new perspective and enlightenment for the formulation of effective stroke prevention and treatment strategies, and promote a comprehensive understanding of heart and brain health and optimize intervention methods.
Collapse
Affiliation(s)
- Mengchen Yu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China
| | - Guohao Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China
| | - Wenbo Chen
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China
| | - Yanmei Qiu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Nanlin You
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China
| | - Sui Chen
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Zhaosheng Wei
- Department of Neurosurgery, Qilu Hospital (Qingdao), Shandong University, Qingdao 266035, China
| | - Longxin Ji
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China
| | - Mengtao Han
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China
| | - Zhen Qin
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China
| | - Tao Sun
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China
| | - Donghai Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China; Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan 250012, China; Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong 253000, China.
| |
Collapse
|
7
|
Mikołajczyk-Stecyna J, Zuk E, Chmurzynska A, Blatkiewicz M, Jopek K, Rucinski M. Exposure to a choline-deficient diet during pregnancy and lactation alters the liver transcriptome profile in offspring of dams with fatty liver. Clin Nutr ESPEN 2025; 66:9-23. [PMID: 39800134 DOI: 10.1016/j.clnesp.2025.01.028] [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: 10/28/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND & AIMS The developmental origin of health and disease hypothesis shows that early adverse exposures can have lifelong health effects. Thus, the aim of this study was to analyze the impact of choline intake during pregnancy and/or lactation on gene expression profiles in the liver of 24-day-old male rat offspring from dams with non-alcoholic fatty liver disease (NAFLD). METHODS Phenotypic characteristic, histological examination and global transcriptome pattern of liver tissue specimens obtained from offspring of dams suffering from fatty liver, provided with proper choline intake during pregnancy and lactation (NN), fed a choline-deficient diet during both periods (DD), deprived of choline only during pregnancy (DN), or only during lactation (ND), was performed. The global gene expression profile was analyzed by using microarray approach (Affymetrix® Rat Gene 2.1 ST Array Strip). The relative expression of selected genes was validated by real-time polymerase chain reaction (qPCR). RESULTS The histological examination of rat liver sections indicated alternations typical for fatty liver in all analyzed groups with increased progression among groups deprived of choline. Choline deficiency in the maternal diet was associated with changes in body mass and composition but not with biochemical marker levels, except for the high density lipoprotein fraction of cholesterol (HDL). Enhanced expression of genes involved in oxidative stress, cell proliferation, activation of catabolic processes related to hepatocyte dysfunction and cell membrane composition were simultaneously observed in all choline-deficient groups. CONCLUSIONS An adequate amount of choline in the diet of a mother with fatty liver during pregnancy and/or lactation can regulate gene expression in the offspring's liver and contribute to a milder stage of the disease in the progeny. Moreover, proper choline supply during the postpartum period is as crucial as during the prenatal period.
Collapse
Affiliation(s)
- Joanna Mikołajczyk-Stecyna
- Poznań University of Life Sciences, Department of Human Nutrition and Dietetics, Wojska Polskiego 31, 60-624 Poznań, Poland.
| | - Ewelina Zuk
- Poznań University of Life Sciences, Department of Human Nutrition and Dietetics, Wojska Polskiego 31, 60-624 Poznań, Poland
| | - Agata Chmurzynska
- Poznań University of Life Sciences, Department of Human Nutrition and Dietetics, Wojska Polskiego 31, 60-624 Poznań, Poland
| | | | - Karol Jopek
- University of Medical Sciences, Department of Histology and Embryology, Poznań, Poland
| | - Marcin Rucinski
- University of Medical Sciences, Department of Histology and Embryology, Poznań, Poland
| |
Collapse
|
8
|
Hawesa H, Alghumaiz M, Alghamdi R, Alrabiah N, Alfaifi B, Allam H, Gamalalddin M, Alshegri H, Shanawani M. Impact of dietary intake on brain choline levels: A 3 Tesla magnetic resonance spectroscopy study. Saudi Med J 2025; 46:254-260. [PMID: 40096978 PMCID: PMC11918667 DOI: 10.15537/smj.2025.46.3.20240698] [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] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/04/2025] [Indexed: 03/19/2025] Open
Abstract
OBJECTIVES To investigate the relationship between dietary choices and brain choline (Cho) levels using magnetic resonance spectroscopy (MRS). METHODS A total of 88 female students from the radiology department at King Abdullah bin Abdulaziz University Hospital, Riyadh, Saudi Arabia, participated in this study. Brain total choline (tCho) levels were estimated using MRS single volume sequence at a 3 Tesla field, with an echo time of 30 ms, repetition time of 2000 ms, voxel size of 15x15x15 mm, and water suppression bandwidth of 50 Hz. Participants' food consumption habits were assessed using a dietary questionnaire to quantify the amount of protein in their daily servings, as protein intake affects Cho levels in the brain. RESULTS Linear regression test applied using the Statistical Package for the Social Sciences, and the result showed significant impact of diet protein intake on the brain tCho level (p=0.000). CONCLUSION The study's findings indicated that dietary choices significantly affect the levels of tCho in the brain. This research can serve as a baseline for health education, highlighting the close connection between dietary decisions and brain Cho levels. Understanding this relationship is essential for promoting a healthy lifestyle among younger generations.
Collapse
Affiliation(s)
- Halima Hawesa
- From the Department of Radiological Sciences (Hawesa, Alghumaiz, Alghamdi, Alrabiah, Alfaifi, Allam, Gamalalddin), College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, and from the Department of Radiology (Alshegri, Shanawani), King Abdullah bin Abdulaziz University Hospital, Riyadh, Kingdom of Saudi Arabia.
| | - Mayar Alghumaiz
- From the Department of Radiological Sciences (Hawesa, Alghumaiz, Alghamdi, Alrabiah, Alfaifi, Allam, Gamalalddin), College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, and from the Department of Radiology (Alshegri, Shanawani), King Abdullah bin Abdulaziz University Hospital, Riyadh, Kingdom of Saudi Arabia.
| | - Renad Alghamdi
- From the Department of Radiological Sciences (Hawesa, Alghumaiz, Alghamdi, Alrabiah, Alfaifi, Allam, Gamalalddin), College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, and from the Department of Radiology (Alshegri, Shanawani), King Abdullah bin Abdulaziz University Hospital, Riyadh, Kingdom of Saudi Arabia.
| | - Nourah Alrabiah
- From the Department of Radiological Sciences (Hawesa, Alghumaiz, Alghamdi, Alrabiah, Alfaifi, Allam, Gamalalddin), College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, and from the Department of Radiology (Alshegri, Shanawani), King Abdullah bin Abdulaziz University Hospital, Riyadh, Kingdom of Saudi Arabia.
| | - Bayader Alfaifi
- From the Department of Radiological Sciences (Hawesa, Alghumaiz, Alghamdi, Alrabiah, Alfaifi, Allam, Gamalalddin), College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, and from the Department of Radiology (Alshegri, Shanawani), King Abdullah bin Abdulaziz University Hospital, Riyadh, Kingdom of Saudi Arabia.
| | - Hind Allam
- From the Department of Radiological Sciences (Hawesa, Alghumaiz, Alghamdi, Alrabiah, Alfaifi, Allam, Gamalalddin), College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, and from the Department of Radiology (Alshegri, Shanawani), King Abdullah bin Abdulaziz University Hospital, Riyadh, Kingdom of Saudi Arabia.
| | - Mahasin Gamalalddin
- From the Department of Radiological Sciences (Hawesa, Alghumaiz, Alghamdi, Alrabiah, Alfaifi, Allam, Gamalalddin), College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, and from the Department of Radiology (Alshegri, Shanawani), King Abdullah bin Abdulaziz University Hospital, Riyadh, Kingdom of Saudi Arabia.
| | - Haya Alshegri
- From the Department of Radiological Sciences (Hawesa, Alghumaiz, Alghamdi, Alrabiah, Alfaifi, Allam, Gamalalddin), College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, and from the Department of Radiology (Alshegri, Shanawani), King Abdullah bin Abdulaziz University Hospital, Riyadh, Kingdom of Saudi Arabia.
| | - Mansour Shanawani
- From the Department of Radiological Sciences (Hawesa, Alghumaiz, Alghamdi, Alrabiah, Alfaifi, Allam, Gamalalddin), College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, and from the Department of Radiology (Alshegri, Shanawani), King Abdullah bin Abdulaziz University Hospital, Riyadh, Kingdom of Saudi Arabia.
| |
Collapse
|
9
|
Zhang X, Lau HCH, Yu J. Pharmacological treatment for metabolic dysfunction-associated steatotic liver disease and related disorders: Current and emerging therapeutic options. Pharmacol Rev 2025; 77:100018. [PMID: 40148030 DOI: 10.1016/j.pharmr.2024.100018] [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: 03/29/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD; formerly known as nonalcoholic fatty liver disease) is a chronic liver disease affecting over a billion individuals worldwide. MASLD can gradually develop into more severe liver pathologies, including metabolic dysfunction-associated steatohepatitis (MASH), cirrhosis, and liver malignancy. Notably, although being a global health problem, there are very limited therapeutic options against MASLD and its related diseases. While a thyroid hormone receptor agonist (resmetirom) is recently approved for MASH treatment, other efforts to control these diseases remain unsatisfactory. Given the projected rise in MASLD and MASH incidence, it is urgent to develop novel and effective therapeutic strategies against these prevalent liver diseases. In this article, the pathogenic mechanisms of MASLD and MASH, including insulin resistance, dysregulated nuclear receptor signaling, and genetic risk factors (eg, patatin-like phospholipase domain-containing 3 and hydroxysteroid 17-β dehydrogenase-13), are introduced. Various therapeutic interventions against MASH are then explored, including approved medication (resmetirom), drugs that are currently in clinical trials (eg, glucagon-like peptide 1 receptor agonist, fibroblast growth factor 21 analog, and PPAR agonist), and those failed in previous trials (eg, obeticholic acid and stearoyl-CoA desaturase 1 antagonist). Moreover, given that the role of gut microbes in MASLD is increasingly acknowledged, alterations in the gut microbiota and microbial mechanisms in MASLD development are elucidated. Therapeutic approaches that target the gut microbiota (eg, dietary intervention and probiotics) against MASLD and related diseases are further explored. With better understanding of the multifaceted pathogenic mechanisms, the development of innovative therapeutics that target the root causes of MASLD and MASH is greatly facilitated. The possibility of alleviating MASH and achieving better patient outcomes is within reach. SIGNIFICANCE STATEMENT: Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease worldwide, and it can progress to more severe pathologies, including steatohepatitis, cirrhosis, and liver cancer. Better understanding of the pathogenic mechanisms of these diseases has facilitated the development of innovative therapeutic strategies. Moreover, increasing evidence has illustrated the crucial role of gut microbiota in the pathogenesis of MASLD and related diseases. It may be clinically feasible to target gut microbes to alleviate MASLD in the future.
Collapse
Affiliation(s)
- Xiang Zhang
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Harry Cheuk-Hay Lau
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
10
|
Yiannakou I, Long MT, Jacques PF, Beiser A, Pickering RT, Moore LL. Eggs, Dietary Choline, and Nonalcoholic Fatty Liver Disease in the Framingham Heart Study. J Nutr 2025; 155:923-935. [PMID: 39424072 PMCID: PMC11934245 DOI: 10.1016/j.tjnut.2024.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/18/2024] [Accepted: 10/02/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Eggs are rich in bioactive compounds, including choline and carotenoids that may benefit cardiometabolic outcomes. However, little is known about their relationship with nonalcoholic fatty liver disease (NAFLD). OBJECTIVES We investigated the association between intakes of eggs and selected egg-rich nutrients (choline, lutein, and zeaxanthin) and NAFLD risk and changes in liver fat over ∼6 y of follow-up in the Framingham Offspring and Third Generation cohorts. METHODS On 2 separate occasions (2002-2005 and 2008-2011), liver fat was assessed using a computed tomography scan to estimate the average liver fat attenuation relative to a control phantom to create the liver phantom ratio (LPR). In 2008-2011, cases of incident NAFLD were identified as an LPR ≤0.33 in the absence of heavy alcohol use, after excluding prevalent NAFLD (LPR ≤0.33) in 2002-2005. Food frequency questionnaires were used to estimate egg intakes (classified as <1, 1, and ≥2 per week), dietary choline (adjusted for body weight using the residual method), and the combined intakes of lutein and zeaxanthin. Multivariable modified Poisson regression and general linear models were used to compute incident risk ratios (RR) of NAFLD and adjusted mean annualized liver fat change. RESULTS NAFLD cumulative incidence was 19% among a total of 1414 participants. We observed no associations between egg intake or the combined intakes of lutein and zeaxanthin with an incident NAFLD risk or liver fat change. Other diet and cardiometabolic risk factors did not modify the association between egg intake and NAFLD risk. However, dietary choline intakes were inversely associated with NAFLD risk (RR for tertile 3 compared with tertile 1: 0.69, 95% CI: 0.51, 0.94). CONCLUSIONS Although egg intake was not directly associated with NAFLD risk, eggs are a major source of dietary choline, which was strongly inversely associated with NAFLD risk in this community-based cohort.
Collapse
Affiliation(s)
- Ioanna Yiannakou
- Department of Medicine/Preventive Medicine and Epidemiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Michelle T Long
- Department of Medicine/Section of Gastroenterology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States; Novo Nordisk A/S, Vandtårnsvej 108-110 Søborg Denmark
| | - Paul F Jacques
- Nutritional Epidemiology, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Alexa Beiser
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, United States; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Richard T Pickering
- Department of Medicine/Preventive Medicine and Epidemiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Lynn L Moore
- Department of Medicine/Preventive Medicine and Epidemiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States.
| |
Collapse
|
11
|
Donaldson J, Jacek T, Wychowański P, Zaworski K, Szkopek D, Woliński J, Grujic D, Pierzynowski S, Pierzynowska K. Rat Model of Endogenous and Exogenous Hyperammonaemia Induced by Different Diets. Int J Mol Sci 2025; 26:1818. [PMID: 40076444 PMCID: PMC11899528 DOI: 10.3390/ijms26051818] [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] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Two different diets able to induce dietary hyperammonaemia (a methionine-choline-deficient (MCD) diet and a methionine-deficient diet enriched with ammonium acetate (MAD + 20% ammonium acetate)) were tested in a rat model. The diets were shown to have different modes of action, inducing significant hyperammonaemia (HA) and growth retardation in the rats, with different metabolic consequences. The MCD diet resulted in the development of endogenous HA, with a decrease in bilirubin levels and an increase in hepatic fat content. In contrast, the MAD + 20% ammonium acetate diet increased circulating ALP and haptoglobin levels and decreased liver mass. The above results suggest that the MCD diet deteriorated the liver function of the rats, resulting in the development of endogenous HA, while the MAD diet caused moderate changes in liver metabolism, resulting in the development of exogenous HA. Interestingly, the commonly used oral treatments Lactulose and Rifaximin did not ameliorate hyperammonaemia during or after the treatment period. In conclusion, even though the diets used in the current study caused somewhat similar hyperammonaemia, they seemed to provoke different metabolic consequences. The latter can have an impact on the severity of the resulting hyperammonaemia and thus on the hyperammonaemia-induced encephalopathy, resulting in the development of distinguishing cognitive and metabolic (liver) effects compared to other forms of encephalopathy. We hypothesized that these rat models, with significantly increased serum ammonia levels, along with different liver injuries, could serve as a suitable double animal model for the testing of new, oral enzyme therapies for hepatic encephalopathy in future studies.
Collapse
Affiliation(s)
- Janine Donaldson
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Parktown, Johannesburg 2193, South Africa
- Anara AB, 23132 Trelleborg, Sweden; (D.G.); (S.P.)
| | - Tomasz Jacek
- National Research Institute of Animal Production, 32-083 Balice, Poland;
| | - Piotr Wychowański
- Department of Head and Neck and Sensory Organs, Division of Oral Surgery and Implantology, Institute of Clinical Dentistry, Gemelli Foundation for the University Policlinic, Catholic University of the “Sacred Heart”, 00168 Rome, Italy;
- Department of Interventional Dentistry, Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland
- Specialized Private Implantology Clinic Wychowański Stomatologia, 02-517 Warsaw, Poland
| | - Kamil Zaworski
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland;
| | - Dominika Szkopek
- Large Animal Models Laboratory, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland; (D.S.); (J.W.)
| | - Jarosław Woliński
- Large Animal Models Laboratory, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland; (D.S.); (J.W.)
| | | | - Stefan Pierzynowski
- Anara AB, 23132 Trelleborg, Sweden; (D.G.); (S.P.)
- Department of Biology, Lund University, 223 62 Lund, Sweden
- Department of Medical Biology, Institute of Rural Health, 20-090 Lublin, Poland
| | - Kateryna Pierzynowska
- Anara AB, 23132 Trelleborg, Sweden; (D.G.); (S.P.)
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland;
- Department of Biology, Lund University, 223 62 Lund, Sweden
| |
Collapse
|
12
|
Boyd SS, Slawson C, Thompson JA. AMEND 2.0: module identification and multi-omic data integration with multiplex-heterogeneous graphs. BMC Bioinformatics 2025; 26:39. [PMID: 39910456 PMCID: PMC11800622 DOI: 10.1186/s12859-025-06063-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/22/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Multi-omic studies provide comprehensive insight into biological systems by evaluating cellular changes between normal and pathological conditions at multiple levels of measurement. Biological networks, which represent interactions or associations between biomolecules, have been highly effective in facilitating omic analysis. However, current network-based methods lack generalizability to accommodate multiple data types across a range of diverse experiments. RESULTS We present AMEND 2.0, an updated active module identification method which can analyze multiplex and/or heterogeneous networks integrated with multi-omic data in a highly generalizable framework, in contrast to existing methods, which are mostly appropriate for at most two specific omic types. It is powered by Random Walk with Restart for multiplex-heterogeneous networks, with additional capabilities including degree bias adjustment and biased random walk for multi-objective module identification. AMEND was applied to two real-world multi-omic datasets: renal cell carcinoma data from The cancer genome atlas and an O-GlcNAc Transferase knockout study. Additional analyses investigate the performance of various subroutines of AMEND on tasks of node ranking and degree bias adjustment. CONCLUSIONS While the analysis of multi-omic datasets in a network context is poised to provide deeper understanding of health and disease, new methods are required to fully take advantage of this increasingly complex data. The current study combines several network analysis techniques into a single versatile method for analyzing biological networks with multi-omic data that can be applied in many diverse scenarios. Software is freely available in the R programming language at https://github.com/samboyd0/AMEND .
Collapse
Affiliation(s)
- Samuel S Boyd
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| | - Chad Slawson
- Department of Biochemistry, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- University of Kansas Cancer Center, Kansas City, KS, 66160, USA
- University of Kansas Alzheimer's Disease Research Center, Fairway, KS, 66205, USA
| | - Jeffrey A Thompson
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- University of Kansas Cancer Center, Kansas City, KS, 66160, USA
| |
Collapse
|
13
|
Dong J, Shelp GV, Poole EM, Cook WJJ, Michaud J, Cho CE. Prenatal choline supplementation enhances metabolic outcomes with differential impact on DNA methylation in Wistar rat offspring and dams. J Nutr Biochem 2025; 136:109806. [PMID: 39547266 DOI: 10.1016/j.jnutbio.2024.109806] [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: 08/13/2024] [Revised: 10/21/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
Choline is an essential nutrient required for proper functioning of organs and serves as a methyl donor. In liver where choline metabolism primarily occurs, glucose homeostasis is regulated through insulin receptor substrates (IRS) 1 and 2. The objective of this research was to determine the role of prenatal choline as a modulator of metabolic health and DNA methylation in liver of offspring and dams. Pregnant Wistar rat dams were fed an AIN-93G diet and received drinking water either with supplemented 0.25% choline (w/w) as choline bitartrate or untreated control. All offspring were weaned to a high-fat diet for 12 weeks. Prenatal choline supplementation led to higher insulin sensitivity in female offspring at weaning as well as lower body weight and food intake and higher insulin sensitivity in female and male adult offspring compared to offspring from untreated dams. Higher hepatic betaine concentrations were observed in dams and female offspring of choline-supplemented dams at weaning and higher glycerophosphocholine in female and male offspring at postweaning compared to the untreated control, suggestive of sustaining different choline pathways. Hepatic gene expression of Irs2 was higher in dams at weaning and female offspring at weaning and postweaning, whereas Irs1 was lower in male offspring at postweaning. Gene-specific DNA methylation of Irs2 was lower in female offspring at postweaning and Irs1 methylation was higher in male offspring at postweaning that exhibited an inverse relationship between methylation and gene expression. In conclusion, prenatal choline supplementation contributes to improved parameters of insulin signaling but these effects varied across time and offspring sex.
Collapse
Affiliation(s)
- Jianzhang Dong
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Gia V Shelp
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Elizabeth M Poole
- Department of Family Relations and Applied Nutrition, University of Guelph, Guelph, Ontario, Canada
| | - William J J Cook
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Jana Michaud
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Clara E Cho
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada.
| |
Collapse
|
14
|
Wan YP, Li S, Li D, Huang XM, Wu JH, Jian J. Study on the molecular mechanisms of rifaximin in the treatment of non‑alcoholic steatohepatitis based on the Helicobacter‑DCA‑Fxr‑Hnf1α signalling pathway. Mol Med Rep 2025; 31:42. [PMID: 39611479 PMCID: PMC11632295 DOI: 10.3892/mmr.2024.13407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/24/2024] [Indexed: 11/30/2024] Open
Abstract
Non‑alcoholic steatohepatitis (NASH), the more progressive form of non‑alcoholic fatty liver disease, has become a major cause of cirrhosis and liver cancer. The aim of the present study was to investigate the anti‑NASH effect of the nonabsorbable antibiotic rifaximin and its specific molecular mechanisms. A methionine‑choline deficient (MCD) diet was used to induce NASH formation in mice. The mice with NASH were treated with rifaximin to observe its effects on liver fat deposition, hepatocyte inflammation and liver fibrosis. Furthermore, the intestinal microbiota of mice with NASH was analysed by 16S rRNA sequencing and terminal ileal bile acid levels were assessed using liquid chromatography‑electrospray ionization‑tandem mass spectrometry analysis. Furthermore, the correlation between the intestinal microflora and bile acid levels in the terminal ileum was investigated, and the effects of rifaximin on the intestinal Helicobacter‑deoxycholic acid (DCA)‑farnesoid X receptor (Fxr)‑hepatocyte nuclear factor 1α (Hnf1α) signalling pathway were examined. Moreover, analyses of mice after intestinal decontamination with broad‑spectrum antibiotics and of hepatocyte‑specific Hnf1α knockout (Hnf1αH‑KO) mice were used to elucidate the molecular mechanisms by which rifaximin improves NASH. Notably, treatment with rifaximin markedly ameliorated liver steatosis, hepatocyte inflammation and liver fibrosis in mice with MCD diet‑induced NASH. Rifaximin modulated the gut microbiota, especially Helicobacter hepaticus, in mice with NASH. In addition, rifaximin inhibited the intestinal Helicobacter‑DCA‑Fxr‑Hnf1α signalling pathway in mice with NASH. By contrast, rifaximin did not exert an anti‑NASH effect on decontamination‑treated mice or Hnf1αH‑KO mice. Taken together, these results indicated that rifaximin can ameliorate NASH in mice by modulating the Helicobacter‑DCA‑Fxr‑Hnf1α signalling pathway, providing a theoretical basis for the clinical treatment of patients with NASH with rifaximin.
Collapse
Affiliation(s)
- Yi-Peng Wan
- Department of Gastroenterology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shuang Li
- Department of Teaching, Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, P.R. China
| | - Dan Li
- Department of Gastroenterology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiao-Mei Huang
- Department of Gastroenterology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jian-Hua Wu
- Department of Gastroenterology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jie Jian
- Department of Gastroenterology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
15
|
Bang S, Shin YH, Park SM, Deng L, Williamson RT, Graham DB, Xavier RJ, Clardy J. Unusual Phospholipids from Morganella morganii Linked to Depression. J Am Chem Soc 2025; 147:2998-3002. [PMID: 39818770 PMCID: PMC11783507 DOI: 10.1021/jacs.4c15158] [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: 10/28/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/19/2025]
Abstract
A multifactorial association study detected a probable causal connection between the prevalence of Morganella morganii in the gut microbiome and the incidence of major depressive disorder (MDD) in the human host. A bioassay-guided fractionation approach identified bacterially produced metabolites that induced pro-inflammatory immune responses. The metabolites are unusual phospholipids that resemble conventional cardiolipins, in which diethanolamine (DEA) replaces the central glycerol. These molecular chimeras of endogenous metabolites from phospholipid biosynthetic pathways and the industrially produced micropollutant DEA activate TLR2/TLR1 receptors and induce the production of pro-inflammatory cytokines, especially IL-6. Their activity in conventional immunomodulatory assays largely parallels that of immunogenic cardiolipins with conventional structures. The molecular mechanism connecting these chimeric cardiolipins to MDD is supported by other studies and has implications for conditions other than MDD.
Collapse
Affiliation(s)
- Sunghee Bang
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| | - Yern-Hyerk Shin
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| | - Sung-Moo Park
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for the Study of Inflammatory Bowel
Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Lei Deng
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for the Study of Inflammatory Bowel
Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - R. Thomas Williamson
- Department
of Chemistry and Biochemistry, University
of North Carolina Wilmington, Wilmington, North Carolina 28409, United States
| | - Daniel B. Graham
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for the Study of Inflammatory Bowel
Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Ramnik J. Xavier
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for the Study of Inflammatory Bowel
Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Jon Clardy
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| |
Collapse
|
16
|
Gousias F, Stylianaki I, Giannenas I, Kallitsis T, Papaioannou N, Chaitidis E, Squires C, Arsenos G, Tsiouris V, Papadopoulos GA. Effects of Milk Thistle Extract Supplementation on Performance, Egg Quality, and Liver Pathology of Laying Hens' Fed Diets Lacking Supplemental Choline Chloride. Vet Sci 2025; 12:77. [PMID: 40005837 PMCID: PMC11860362 DOI: 10.3390/vetsci12020077] [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: 10/17/2024] [Revised: 01/08/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
The current study evaluated the effects of milk thistle extract supplementation in laying hens' fed diets lacking choline chloride addition. A total of 60 Isa-brown laying hens were randomly allocated into T1: control diet, 0% extract supplementation; T2: control diet with 1% extract; T3: control diet with 2.5% extract; and T4: control diet with 4% extract. Egg quality parameters, yolk lipid oxidation, malondialdehyde (MDA) content, and fatty acid profile were assessed. Livers were examined grossly and histologically to evaluate hepatocellular lesions such as vacuolization (lipidosis), reticular stromal architecture, the amount of collagenous connective tissue, and vascular wall changes. Groups T3 and T4 showed darker yolks compared to both control group and T2 (p = 0.001) and redness of the egg yolk (p < 0.001). The MDA was lowest in T2 group which had improved gross appearance with lower degrees of hepatic vacuolization than other groups. Liver discoloration was milder in T3 (43.8%) compared to the T1 and T4 groups (18.8% and 12.5%, respectively, p = 0.013). Reticulin loss was correlated with the degree of hepatic vacuolization (r = 0.751, p < 0.001). Supplementation with MT extract in diets lacking choline chloride may influence certain egg quality indices and liver gross macroscopic lesions in laying hens.
Collapse
Affiliation(s)
- Fotis Gousias
- Laboratory of Animal Husbandry, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (T.K.); (C.S.); (G.A.); (G.A.P.)
| | - Ioanna Stylianaki
- Laboratory of Pathology, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (I.S.); (N.P.); (E.C.)
| | - Ilias Giannenas
- Laboratory of Nutrition, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | - Theodoros Kallitsis
- Laboratory of Animal Husbandry, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (T.K.); (C.S.); (G.A.); (G.A.P.)
| | - Nikolaos Papaioannou
- Laboratory of Pathology, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (I.S.); (N.P.); (E.C.)
| | - Efstratios Chaitidis
- Laboratory of Pathology, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (I.S.); (N.P.); (E.C.)
| | - Clare Squires
- Laboratory of Animal Husbandry, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (T.K.); (C.S.); (G.A.); (G.A.P.)
| | - Georgios Arsenos
- Laboratory of Animal Husbandry, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (T.K.); (C.S.); (G.A.); (G.A.P.)
| | - Vasilios Tsiouris
- Unit of Avian Medicine, Clinic of Farm Animals, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, 546 27 Thessaloniki, Greece;
| | - Georgios A. Papadopoulos
- Laboratory of Animal Husbandry, Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (T.K.); (C.S.); (G.A.); (G.A.P.)
| |
Collapse
|
17
|
Kenny TC, Scharenberg S, Abu-Remaileh M, Birsoy K. Cellular and organismal function of choline metabolism. Nat Metab 2025; 7:35-52. [PMID: 39779890 PMCID: PMC11990872 DOI: 10.1038/s42255-024-01203-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 12/09/2024] [Indexed: 01/11/2025]
Abstract
Choline is an essential micronutrient critical for cellular and organismal homeostasis. As a core component of phospholipids and sphingolipids, it is indispensable for membrane architecture and function. Additionally, choline is a precursor for acetylcholine, a key neurotransmitter, and betaine, a methyl donor important for epigenetic regulation. Consistent with its pleiotropic role in cellular physiology, choline metabolism contributes to numerous developmental and physiological processes in the brain, liver, kidney, lung and immune system, and both choline deficiency and excess are implicated in human disease. Mutations in the genes encoding choline metabolism proteins lead to inborn errors of metabolism, which manifest in diverse clinical pathologies. While the identities of many enzymes involved in choline metabolism were identified decades ago, only recently has the field begun to understand the diverse mechanisms by which choline availability is regulated and fuelled via metabolite transport/recycling and nutrient acquisition. This review provides a comprehensive overview of choline metabolism, emphasizing emerging concepts and their implications for human health and disease.
Collapse
Affiliation(s)
- Timothy C Kenny
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Samantha Scharenberg
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA, USA
- Stanford Medical Scientist Training Program, Stanford University, Stanford, CA, USA
- Stanford Biophysics Program, Stanford University, Stanford, CA, USA
| | - Monther Abu-Remaileh
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA.
- Department of Genetics, Stanford University, Stanford, CA, USA.
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA, USA.
- The Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| | - Kıvanç Birsoy
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
18
|
Calzoni E, Bertoldi A, Cesaretti A, Alabed HBR, Cerrotti G, Pellegrino RM, Buratta S, Urbanelli L, Emiliani C. Aloe Extracellular Vesicles as Carriers of Photoinducible Metabolites Exhibiting Cellular Phototoxicity. Cells 2024; 13:1845. [PMID: 39594594 PMCID: PMC11592872 DOI: 10.3390/cells13221845] [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: 09/23/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
The growing interest in plant-origin active molecules with medicinal properties has led to a revaluation of plants in the pharmaceutical field. Plant-derived extracellular vesicles (PDEVs) have emerged as promising candidates for next-generation drug delivery systems due to their ability to concentrate and deliver a plethora of bioactive molecules. These bilayer membranous vesicles, whose diameter ranges from 30 to 1000 nm, are released by different cell types and play a crucial role in cross-kingdom communication between plants and humans. Notably, PDEVs have demonstrated efficacy in treating various diseases, including cancer, alcoholic liver disease, and inflammatory bowel disease. However, further research on plant vesicles is necessary to fully understand their traits and purposes. This study investigates the phototoxic effects of extracellular vesicles (EVs) from Aloe arborescens, Aloe barbadensis, and Aloe chinensis on the human melanoma cell line SK-MEL-5, focusing on their anthraquinone content, recognized as natural photosensitizers. The phototoxic impact of Aloe EVs is associated with ROS production, leading to significant oxidative stress in melanoma cells, as validated by a metabolome analysis. These findings suggest that EVs from Aloe arborescens, Aloe barbadensis, and Aloe chinensis hold promise as potential photosensitizers, thus highlighting their potential for future application in photodynamic cancer therapy and providing valuable insights into the possible utilization of PDEVs for therapeutic purposes.
Collapse
Affiliation(s)
- Eleonora Calzoni
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
| | - Agnese Bertoldi
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
| | - Alessio Cesaretti
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
- Centro di Eccellenza Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Husam B. R. Alabed
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
| | - Giada Cerrotti
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
| | - Roberto Maria Pellegrino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
| | - Sandra Buratta
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
| | - Lorena Urbanelli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (E.C.); (A.B.); (H.B.R.A.); (G.C.); (R.M.P.); (S.B.); (L.U.); (C.E.)
- Centro di Eccellenza Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| |
Collapse
|
19
|
Setayesh T, Hu Y, Vaziri F, Wei D, Wan YJY. The spatial impact of a Western diet in enriching Galectin-1-regulated Rho, ECM, and SASP signaling in a novel MASH-HCC mouse model. Biomark Res 2024; 12:122. [PMID: 39402682 PMCID: PMC11476289 DOI: 10.1186/s40364-024-00660-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) arising from metabolic dysfunction-associated steatohepatitis (MASH) presents a significant clinical challenge, particularly given the prevalence of the Western diet (WD). The influence of diet on the tumor microenvironment remains poorly understood. Galectin-1 (Gal-1) is a biomarker for HCC and has a crucial role in liver carcinogenesis. Our previous studies demonstrated that silencing Gal-1 effectively treats mouse HCC. However, the impacts of a WD on Gal-1 signaling on MASH to HCC progression are unknown, and this study addresses these knowledge gaps. METHODS We developed a novel MASH-HCC mouse model. Using spatial transcriptomics and multiplex immunohistochemistry (IHC), we studied the effects of a WD on the liver and tumor microenvironment. By modulating Gal-1 expression through silencing and overexpression, we explored the location-specific impacts of WD on Gal-1 signaling. RESULTS Pathways such as Rho signaling, extracellular matrix (ECM) remodeling, and senescence-associated secretory phenotypes (SASP) were prominently activated in WD-induced metabolic dysfunction-associated fatty liver disease (MAFLD) and MASH-HCC, compared to healthy livers controls. Furthermore, Rho GTPase effectors, ECM remodeling, neutrophil degranulation, cellular stress, and cell cycle pathways were consistently enriched in human and mouse MASH-HCC. Spatially, these pathways were enriched in the tumor and tumor margins of mouse MASH-HCC. Additionally, there was a notable increase in CD11c and PD-L1-positive cells from non-tumor tissues to the tumor margin and inside the tumor of MASH-HCC, suggesting compromised immune surveillance due to WD intake. Moreover, MASH-HCC exhibited significant Gal-1 induction in N-Cadherin-positive cells, indicating enhanced epithelial-to-mesenchymal transition (EMT). Modulating Gal-1 expression in MASH-HCC further established its specific roles in regulating Rho signaling and SASP in the tumor margin and non-tumor tissues in MASH-HCC. CONCLUSION WD intake significantly influences vital cellular processes involved in Gal-1-mediated signaling, including Rho signaling and ECM remodeling, in the tumor microenvironment, thereby contributing to the development of MASH-HCC.
Collapse
Affiliation(s)
- Tahereh Setayesh
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Room 3400B, Research Building III, 4645 2nd Ave, Sacramento, CA, 95817, USA
| | - Ying Hu
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Room 3400B, Research Building III, 4645 2nd Ave, Sacramento, CA, 95817, USA
| | - Farzam Vaziri
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Room 3400B, Research Building III, 4645 2nd Ave, Sacramento, CA, 95817, USA
| | - Dongguang Wei
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Room 3400B, Research Building III, 4645 2nd Ave, Sacramento, CA, 95817, USA
| | - Yu-Jui Yvonne Wan
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Room 3400B, Research Building III, 4645 2nd Ave, Sacramento, CA, 95817, USA.
| |
Collapse
|
20
|
Ahlawat S, Mohan H, Sharma KK. Proteome profiling, biochemical and histological analysis of diclofenac-induced liver toxicity in Yersinia enterocolitica and Lactobacillus fermentum fed rat model: a comparative analysis. Biotechnol Lett 2024; 46:807-826. [PMID: 38985258 DOI: 10.1007/s10529-024-03510-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/23/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024]
Abstract
Diclofenac is a hepatotoxic non-steroidal anti-inflammatory drug (NSAID) that affects liver histology and its protein expression levels. Here, we studied the effect of diclofenac on rat liver when co-administrated with either Yersinia enterocolitica strain 8081 serotype O:8 biovar 1B (D*Y) or Lactobacillus fermentum strain 9338 (D*L). Spectroscopic analysis of stool samples showed biotransformation of diclofenac. When compared with each other, D*Y rats lack peaks at 1709 and 1198 cm-1, while D*L rats lack peaks at 1411 cm-1. However, when compared to control, both groups lack peaks at 1379 and 1170 cm-1. Assessment of serum biomarkers of hepatotoxicity indicated significantly altered activities of AST (D*Y: 185.65 ± 8.575 vs Control: 61.9 ± 2.607, D*L: 247.5 ± 5.717 vs Control: 61.9 ± 2.607), ALT (D*Y: 229.8 ± 6.920 vs Control: 70.7 ± 3.109, D*L: 123.75 ± 6.068 vs Control: 70.7 ± 3.109), and ALP (D*Y: 276.4 ± 18.154 vs Control: 320.6 ± 9.829, D*L: 298.5 ± 12.336 vs Control: 320.6 ± 9.829) in IU/L. The analysis of histological alterations showed hepatic sinusoidal dilation with vein congestion and cell infiltration exclusively in D*Y rats along with other histological changes that are common to both test groups, thereby suggesting more pronounced alterations in D*Y rats. Further, LC-MS/MS based label-free quantitation of proteins from liver tissues revealed 74.75% up-regulated, 25.25% down-regulated in D*Y rats and 51.16% up-regulated, 48.84% down-regulated in D*L experiments. The proteomics-identified proteins majorly belonged to metabolism, apoptosis, stress response and redox homeostasis, and detoxification and antioxidant defence that demonstrated the potential damage of rat liver, more pronounced in D*Y rats. Altogether the results are in favor that the administration of lactobacilli somewhat protected the rat hepatic cells against the diclofenac-induced toxicity.
Collapse
Affiliation(s)
- Shruti Ahlawat
- Laboratory of Enzymology and Recombinant DNA Technology, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
- Department of Microbiology, Faculty of Allied Health Sciences, SGT University, Gurgaon-Badli Road Chandu, Budhera, Gurugram, Haryana, 122505, India
| | - Hari Mohan
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Krishna Kant Sharma
- Laboratory of Enzymology and Recombinant DNA Technology, Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, 124001, India.
| |
Collapse
|
21
|
Tabone T, Mooney P, Donnellan C. Intestinal failure-associated liver disease: Current challenges in screening, diagnosis, and parenteral nutrition considerations. Nutr Clin Pract 2024; 39:1003-1025. [PMID: 38245851 DOI: 10.1002/ncp.11116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/19/2023] [Accepted: 12/25/2023] [Indexed: 01/22/2024] Open
Abstract
Intestinal failure-associated liver disease (IFALD) is a serious life-limiting complication that can occur throughout the clinical course of intestinal failure and its management by parenteral nutrition (PN). Despite this, there is a lack of a standardized definition for IFALD, which makes this insidious condition increasingly difficult to screen and diagnose in clinical practice. Attenuating the progression of liver disease before the onset of liver failure is key to improving morbidity and mortality in these patients. This requires timely detection and promptly addressing reversible factors. Although there are various noninvasive tools available to the clinician to detect early fibrosis or cirrhosis in various chronic liver disease states, these have not been validated in the patient population with IFALD. Such tools include biochemical composite scoring systems for fibrosis, transient elastography, and dynamic liver function tests. This review article aims to highlight the existing real need for an accurate, reproducible method to detect IFALD in its early stages. In addition, we also explore the role PN plays in the pathogenesis of this complex multifactorial condition. Various aspects of PN administration have been implicated in the etiology of IFALD, including the composition of the lipid component, nutrient excess and deficiency, and infusion timing. We aim to highlight the clinical relevance of these PN-associated factors in the development of IFALD and how these can be managed to mitigate the progression of IFALD.
Collapse
Affiliation(s)
- Trevor Tabone
- Department of Gastroenterology, St James University Hospital, Leeds, United Kingdom
| | - Peter Mooney
- Department of Gastroenterology, St James University Hospital, Leeds, United Kingdom
| | - Clare Donnellan
- Department of Gastroenterology, St James University Hospital, Leeds, United Kingdom
| |
Collapse
|
22
|
Lu J, Shataer D, Yan H, Dong X, Zhang M, Qin Y, Cui J, Wang L. Probiotics and Non-Alcoholic Fatty Liver Disease: Unveiling the Mechanisms of Lactobacillus plantarum and Bifidobacterium bifidum in Modulating Lipid Metabolism, Inflammation, and Intestinal Barrier Integrity. Foods 2024; 13:2992. [PMID: 39335920 PMCID: PMC11431124 DOI: 10.3390/foods13182992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
In recent years, the prevalence of non-alcoholic fatty liver disease (NAFLD) has risen annually, yet due to the intricacies of its pathogenesis and therapeutic challenges, there remains no definitive medication for this condition. This review explores the intricate relationship between the intestinal microbiome and the pathogenesis of NAFLD, emphasizing the substantial roles played by Lactobacillus plantarum and Bifidobacterium bifidum. These probiotics manipulate lipid synthesis genes and phosphorylated proteins through pathways such as the AMPK/Nrf2, LPS-TLR4-NF-κB, AMPKα/PGC-1α, SREBP-1/FAS, and SREBP-1/ACC signaling pathways to reduce hepatic lipid accumulation and oxidative stress, key components of NAFLD progression. By modifying the intestinal microbial composition and abundance, they combat the overgrowth of harmful bacteria, alleviating the inflammatory response precipitated by dysbiosis and bolstering the intestinal mucosal barrier. Furthermore, they participate in cellular immune regulation, including CD4+ T cells and Treg cells, to suppress systemic inflammation. L. plantarum and B. bifidum also modulate lipid metabolism and immune reactions by adjusting gut metabolites, including propionic and butyric acids, which inhibit liver inflammation and fat deposition. The capacity of probiotics to modulate lipid metabolism, immune responses, and gut microbiota presents an innovative therapeutic strategy. With a global increase in NAFLD prevalence, these insights propose a promising natural method to decelerate disease progression, avert liver damage, and tackle associated metabolic issues, significantly advancing microbiome-focused treatments for NAFLD.
Collapse
Affiliation(s)
- Jing Lu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China; (J.L.); (D.S.); (H.Y.); (M.Z.); (Y.Q.)
| | - Dilireba Shataer
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China; (J.L.); (D.S.); (H.Y.); (M.Z.); (Y.Q.)
| | - Huizhen Yan
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China; (J.L.); (D.S.); (H.Y.); (M.Z.); (Y.Q.)
| | - Xiaoxiao Dong
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China; (J.L.); (D.S.); (H.Y.); (M.Z.); (Y.Q.)
| | - Minwei Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China; (J.L.); (D.S.); (H.Y.); (M.Z.); (Y.Q.)
| | - Yanan Qin
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China; (J.L.); (D.S.); (H.Y.); (M.Z.); (Y.Q.)
| | - Jie Cui
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Liang Wang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China; (J.L.); (D.S.); (H.Y.); (M.Z.); (Y.Q.)
| |
Collapse
|
23
|
Fu Y, Hua Y, Alam N, Liu E. Progress in the Study of Animal Models of Metabolic Dysfunction-Associated Steatotic Liver Disease. Nutrients 2024; 16:3120. [PMID: 39339720 PMCID: PMC11435380 DOI: 10.3390/nu16183120] [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] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has recently been proposed as an alternative term to NAFLD. MASLD is a globally recognized chronic liver disease that poses significant health concerns and is frequently associated with obesity, insulin resistance, and hyperlipidemia. To better understand its pathogenesis and to develop effective treatments, it is essential to establish suitable animal models. Therefore, attempts have been made to establish modelling approaches that are highly similar to human diet, physiology, and pathology to better replicate disease progression. Here, we reviewed the pathogenesis of MASLD disease and summarised the used animal models of MASLD in the last 7 years through the PubMed database. In addition, we have summarised the commonly used animal models of MASLD and describe the advantages and disadvantages of various models of MASLD induction, including genetic models, diet, and chemically induced models, to provide directions for research on the pathogenesis and treatment of MASLD.
Collapse
Affiliation(s)
- Yu Fu
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China; (Y.F.); (Y.H.)
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, 76 Yanta West Road, Xi’an 710061, China;
| | - Yuxin Hua
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China; (Y.F.); (Y.H.)
| | - Naqash Alam
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, 76 Yanta West Road, Xi’an 710061, China;
| | - Enqi Liu
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, 76 Yanta West Road, Xi’an 710061, China;
| |
Collapse
|
24
|
Yaghmaei H, Nojoumi SA, Soltanipur M, Yarmohammadi H, Mirhosseini SM, Rezaei M, Jalali Nadoushan M, Siadat SD. The role of gut microbiota in non-alcoholic fatty liver disease pathogenesis. OBESITY MEDICINE 2024; 50:100551. [DOI: 10.1016/j.obmed.2024.100551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
|
25
|
Kütük T, Onbaşilar İ, Oskay-Halaçli S, Babaoğlu B, Ayhan S, Yalçin SS. Investigation of the Hepatitis-B Vaccine's Immune Response in a Non-Alcoholic Fatty Liver Disease Mouse Model. Vaccines (Basel) 2024; 12:934. [PMID: 39204057 PMCID: PMC11359425 DOI: 10.3390/vaccines12080934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
This study aimed to investigate the immunogenicity of the hepatitis B virus (HBV) vaccine by applying a normal and high-dose hepatitis B virus vaccination program in the mice modeling of non-alcoholic fatty liver disease (NAFLD). NAFLD was induced in mouse livers via diet. At the 10-week mark, both groups were divided into 3 subgroups. While the standard dose vaccination program was applied on days 0, 7, and 21, two high-dose programs were applied: one was applied on days 0 and 7, and the other was applied on days 0, 7, and 21. All mice were euthanized. Blood samples from anti-HB titers; T follicular helper, T follicular regulatory, CD27+, and CD38+ cells; and the liver, spleen, and thymus were taken for histopathologic evaluation. NAFLD subgroups receiving high doses showed higher hepatocyte ballooning scores than normal-dose subgroup. There were differences in CD27+ and CD27+CD38+ cells in animals fed on different diets, without any differences or interactions in terms of vaccine protocols. In the NAFLD group, a negative correlation was observed between anti-HB titers and T helper and CD27+ cells, while a positive correlation was observed with CD38+ cells. NAFLD induced changes in immune parameters in mice, but there was no difference in vaccine efficacy among the applied vaccine protocols. Based on this study's results, high-dose vaccination protocols are not recommended in cases of NAFLD, as they do not enhance efficacy and may lead to increased liver damage.
Collapse
Affiliation(s)
- Tuğba Kütük
- Vaccinology Department, Institute of Vaccinology, Hacettepe University, Ankara 06430, Türkiye; (T.K.); (S.S.Y.)
- Turkish Medicines and Medical Devices Agency, Ankara 06500, Türkiye
| | - İlyas Onbaşilar
- Vaccinology Department, Institute of Vaccinology, Hacettepe University, Ankara 06430, Türkiye; (T.K.); (S.S.Y.)
- Health Science Institute, Hacettepe University, Ankara 06430, Türkiye
- Transgenic Animal Technologies Research and Application Center, Hacettepe University, Ankara 06430, Türkiye
| | - Sevil Oskay-Halaçli
- Department of Basic Sciences of Pediatrics, Institute of Child Health, Hacettepe University, Ankara 06430, Türkiye; (S.O.-H.); (S.A.)
| | - Berrin Babaoğlu
- Department of Pathology, Hacettepe University, Ankara 06430, Türkiye;
| | - Selda Ayhan
- Department of Basic Sciences of Pediatrics, Institute of Child Health, Hacettepe University, Ankara 06430, Türkiye; (S.O.-H.); (S.A.)
| | - Sıddika Songül Yalçin
- Vaccinology Department, Institute of Vaccinology, Hacettepe University, Ankara 06430, Türkiye; (T.K.); (S.S.Y.)
- Department of Social Pediatrics, Institute of Child Health, Hacettepe University, Ankara 06430, Türkiye
| |
Collapse
|
26
|
Perva IT, Simina IE, Bende R, Motofelea AC, Chirita Emandi A, Andreescu N, Sima A, Vlad A, Sporea I, Zimbru C, Tutac PC, Puiu M, Niculescu MD. Use of a Micronutrient Cocktail to Improve Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) in Adults with Obesity: A Randomized, Double-Blinded Pilot Clinical Trial. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1366. [PMID: 39202647 PMCID: PMC11356300 DOI: 10.3390/medicina60081366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024]
Abstract
Background and Objectives: The goal of this study was to assess the impact of supplementation with a combination of nutrients on metabolic-dysfunction-associated steatotic liver disease (MASLD)-related liver parameters, and other parameters related to metabolic syndrome in adults with obesity. These measurements included anthropometric and lipid profiling, and FibroScan technology (controlled attenuation parameter (CAP) and transient elastography (TE) values). Materials and Methods: A double-blind, placebo-controlled pilot clinical trial was conducted over a three-month treatment period. Adults with metabolic syndrome and obesity were allocated to receive either a cocktail of nutrients with defined daily dosages (5-MTHF, betaine, alpha-linolenic acid, eicosapentaenoic acid, choline bitartrate, docosahexaenoic acid, and vitamin B12) or a placebo. The participants were evaluated at the start and the end of the three-month treatment period. Results: A total of 155 participants entered the study, comprising 84 in the treatment group and 71 in the placebo group. The administration of the nutritional supplement resulted in a notable reduction in both CAP and TE scores when compared to the placebo group. The treatment group exhibited a mean reduction in CAP of 4% (p < 0.05) and a mean reduction in TE of 7.8% (p < 0.05), indicative of a decline in liver fat content and fibrosis. Conclusions: The supplementation over a period of three months led to a significant amelioration of liver fibrosis and steatosis parameters in adults with metabolic syndrome and obesity. These findings suggest that this supplementation regimen could be a beneficial adjunct therapy for improving liver health in adults with obesity-induced MASLD.
Collapse
Affiliation(s)
- Iulia Teodora Perva
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (I.T.P.); (A.C.E.); (N.A.); (M.P.); (M.D.N.)
- Regional Center of Medical Genetics Timiș, Clinical Emergency Hospital for Children “Louis Țurcanu”, Iosif Nemoianu Street N°2, 300011 Timisoara, Romania
- Department of Medical Genetics, Asociatia Oncohelp, 300239 Timișoara, Romania
| | - Iulia Elena Simina
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (I.T.P.); (A.C.E.); (N.A.); (M.P.); (M.D.N.)
- Department of Medical Genetics, Asociatia Oncohelp, 300239 Timișoara, Romania
| | - Renata Bende
- Department of Gastroenterology and Hepatology, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (R.B.); (I.S.)
- Center of Advanced Research in Gastroenterology and Hepatology, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Alexandru Cătălin Motofelea
- Department of Internal Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania;
| | - Adela Chirita Emandi
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (I.T.P.); (A.C.E.); (N.A.); (M.P.); (M.D.N.)
- Regional Center of Medical Genetics Timiș, Clinical Emergency Hospital for Children “Louis Țurcanu”, Iosif Nemoianu Street N°2, 300011 Timisoara, Romania
| | - Nicoleta Andreescu
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (I.T.P.); (A.C.E.); (N.A.); (M.P.); (M.D.N.)
- Regional Center of Medical Genetics Timiș, Clinical Emergency Hospital for Children “Louis Țurcanu”, Iosif Nemoianu Street N°2, 300011 Timisoara, Romania
| | - Alexandra Sima
- Department of Internal Medicine II, Division of Diabetes, Nutrition and Metabolic Diseases, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (A.S.); (A.V.)
- Center for Research in Preventive Medicine, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Adrian Vlad
- Department of Internal Medicine II, Division of Diabetes, Nutrition and Metabolic Diseases, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (A.S.); (A.V.)
- Center for Molecular Research in Nephrology and Vascular Disease, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Ioan Sporea
- Department of Gastroenterology and Hepatology, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (R.B.); (I.S.)
- Center of Advanced Research in Gastroenterology and Hepatology, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Cristian Zimbru
- Department of Automation and Applied Informatics, Politehnica University Timișoara, 300223 Timișoara, Romania;
| | - Paul Calin Tutac
- Toxicology and Molecular Biology Department, “Pius Brinzeu” Clinical Emergency County Hospital, 300723 Timisoara, Romania;
| | - Maria Puiu
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (I.T.P.); (A.C.E.); (N.A.); (M.P.); (M.D.N.)
- Regional Center of Medical Genetics Timiș, Clinical Emergency Hospital for Children “Louis Țurcanu”, Iosif Nemoianu Street N°2, 300011 Timisoara, Romania
| | - Mihai Dinu Niculescu
- Department of Microscopic Morphology, Genetics Discipline, Center of Genomic Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq., 300041 Timisoara, Romania; (I.T.P.); (A.C.E.); (N.A.); (M.P.); (M.D.N.)
- Advanced Nutrigenomics LLC, Durham, NC 27703, USA
| |
Collapse
|
27
|
Al-Sulaiti H, Anwardeen N, Bashraheel SS, Naja K, Elrayess MA. Alterations in Choline Metabolism in Non-Obese Individuals with Insulin Resistance and Type 2 Diabetes Mellitus. Metabolites 2024; 14:457. [PMID: 39195553 DOI: 10.3390/metabo14080457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
The prevalence of non-obese individuals with insulin resistance (IR) and type 2 diabetes (T2D) is increasing worldwide. This study investigates the metabolic signature of phospholipid-associated metabolites in non-obese individuals with IR and T2D, aiming to identify potential biomarkers for these metabolic disorders. The study cohort included non-obese individuals from the Qatar Biobank categorized into three groups: insulin sensitive, insulin resistant, and patients with T2D. Each group comprised 236 participants, totaling 708 individuals. Metabolomic profiling was conducted using high-resolution mass spectrometry, and statistical analyses were performed to identify metabolites associated with the progression from IS to IR and T2D. The study observed significant alterations in specific phospholipid metabolites across the IS, IR, and T2D groups. Choline phosphate, glycerophosphoethanolamine, choline, glycerophosphorylcholine (GPC), and trimethylamine N-oxide showed significant changes correlated with disease progression. A distinct metabolic signature in non-obese individuals with IR and T2D was characterized by shifts in choline metabolism, including decreased levels of choline and trimethylamine N-oxide and increased levels of phosphatidylcholines, phosphatidylethanolamines, and their degradation products. These findings suggest that alterations in choline metabolism may play a critical role in the development of glucose intolerance and insulin resistance. Targeting choline metabolism could offer potential therapeutic strategies for treating T2D. Further research is needed to validate these biomarkers and understand their functional significance in the pathogenesis of IR and T2D in non-obese populations.
Collapse
Affiliation(s)
- Haya Al-Sulaiti
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
| | - Najeha Anwardeen
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
| | - Sara S Bashraheel
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Khaled Naja
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
| | - Mohamed A Elrayess
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
28
|
Lee KH, Hong M, Hur HJ, Sung MJ, Lee AS, Kim MJ, Yang HJ, Kim MS. Metabolomic profiling analysis reveals the benefits of ginseng berry intake on mitochondrial function and glucose metabolism in the liver of obese mice. Metabolomics 2024; 20:96. [PMID: 39110263 DOI: 10.1007/s11306-024-02152-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 07/10/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION Ginseng berry (GB) has previously been demonstrated to improve systemic insulin resistance and regulate hepatic glucose metabolism and steatosis in mice with diet-induced obesity (DIO). OBJECTIVES In this study, the role of GB in metabolism was assessed using metabolomics analysis on the total liver metabolites of DIO mice. METHODS Metabolomic profiling was performed using capillary electrophoresis time-of-flight mass spectrometry (CE-TOF/MS) of liver tissue from mice on a 12-wk normal chow diet (NC), high-fat diet (HFD), and HFD supplemented with 0.1% GB (HFD + GB). The detected metabolites, its pathways, and functions were analyzed through partial least square discriminant analysis (PLS-DA), the small molecular pathway database (SMPDB), and MetaboAnalyst 5.0. RESULTS The liver metabolite profiles of NC, HFD, and GB-fed mice (HFD + GB) were highly compartmentalized. Metabolites involved in major liver functions, such as mitochondrial function, gluconeogenesis/glycolysis, fatty acid metabolism, and primary bile acid biosynthesis, showed differences after GB intake. The metabolites that showed significant correlations with fasting blood glucose (FBG), insulin, and homeostatic model assessment for insulin resistance (HOMA-IR) were highly associated with mitochondrial membrane function, energy homeostasis, and glucose metabolism. Ginseng berry intake increased the levels of metabolites involved in mitochondrial membrane function, decreased the levels of metabolites related to glucose metabolism, and was highly correlated with metabolic phenotypes. CONCLUSION This study demonstrated that long-term intake of GB changed the metabolite of hepatosteatotic livers in DIO mice, normalizing global liver metabolites involved in mitochondrial function and glucose metabolism and indicating the potential mechanism of GB in ameliorating hyperglycemia in DIO mice.
Collapse
Affiliation(s)
- Kyun-Hee Lee
- Research Division of Food and Function, Korea Food Research Institute, 245 Nongsaengmyeong-ro, Wanju-gun, Jeonbuk-do, 55365, Republic of Korea
- Department of Food Biotechnology, University of Science & Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Moonju Hong
- Research Division of Food and Function, Korea Food Research Institute, 245 Nongsaengmyeong-ro, Wanju-gun, Jeonbuk-do, 55365, Republic of Korea
- Department of Food Biotechnology, University of Science & Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Haeng Jeon Hur
- Research Division of Food and Function, Korea Food Research Institute, 245 Nongsaengmyeong-ro, Wanju-gun, Jeonbuk-do, 55365, Republic of Korea
| | - Mi Jeong Sung
- Research Division of Food and Function, Korea Food Research Institute, 245 Nongsaengmyeong-ro, Wanju-gun, Jeonbuk-do, 55365, Republic of Korea
| | - Ae Sin Lee
- Research Division of Food and Function, Korea Food Research Institute, 245 Nongsaengmyeong-ro, Wanju-gun, Jeonbuk-do, 55365, Republic of Korea
| | - Min Jung Kim
- Research Division of Food and Function, Korea Food Research Institute, 245 Nongsaengmyeong-ro, Wanju-gun, Jeonbuk-do, 55365, Republic of Korea
| | - Hye Jeong Yang
- Research Division of Food and Function, Korea Food Research Institute, 245 Nongsaengmyeong-ro, Wanju-gun, Jeonbuk-do, 55365, Republic of Korea
| | - Myung-Sunny Kim
- Research Division of Food and Function, Korea Food Research Institute, 245 Nongsaengmyeong-ro, Wanju-gun, Jeonbuk-do, 55365, Republic of Korea.
- Department of Food Biotechnology, University of Science & Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
29
|
Xu C, Zhang M, Zhang S, Wang P, Lai C, Meng D, Chen Z, Yi X, Gao X. Simultaneous determination of choline, L-carnitine, betaine, trimethylamine, trimethylamine N-oxide, and creatinine in plasma, liver, and feces of hyperlipidemic rats by UHPLC-MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1243:124210. [PMID: 38936270 DOI: 10.1016/j.jchromb.2024.124210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/03/2024] [Accepted: 06/16/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND Due to the close correlation between choline, L-carnitine, betaine and their intestinal microbial metabolites, including trimethylamine (TMA) and trimethylamine N-oxide (TMAO), and creatinine, there has been an increasing interest in the study of these compounds in vivo. METHODS In this study, a rapid stable isotope dilution (SID)-UHPLC-MS/MS method was developed for the simultaneous determination of choline, L-carnitine, betaine, TMA, TMAO and creatinine in plasma, liver and feces of rats. The method was validated using quality control (QC) samples spiked at low, medium and high levels. Second, we applied the method to quantify the effects of Rosa Roxburghii Tratt juice (RRTJ) on plasma, liver, and fecal levels of choline, L-carnitine, betaine, TMA, TMAO, and creatinine in high-fat diet-induced hyperlipidemic rats, demonstrating the utility of the method. RESULTS The limits of detection (LOD) were 0.04-0.027 µM and the limits of quantification (LOQ) were 0.009-0.094 µM. The linear ranges for each metabolite in plasma were choline1.50-96 µM; L-carnitine: 2-128 µM; betaine: 3-192 µM; TMA: 0.01-40.96 µM; TMAO: 0.06-61.44 µM and creatinine: 1-64 µM (R2 ≥ 0.9954). The linear ranges for each metabolite in liver were Choline: 12-768 µM; L-carnitine: 1.5-96 µM; betaine: 10-640 µM; TMA: 0.5-32 µM; TMAO: 0.02-81.92 µM and creatinine: 0.2-204.8 µM (R2 ≥ 0.9938). The linear ranges for each metabolite in feces were choline: 1.5-96 µM; L-carnitine: 0.01-40.96 µM; Betaine: 1.5-96 µM; TMA: 1-64 µM; TMAO: 0.02-81.92 µM and Creatinine: 0.02-81.92 µM (R2 ≥ 0.998). The intra-day and inter-day coefficients of variation were < 8 % for all analytes. The samples were stabilized after multiple freeze-thaw cycles (3 freeze-thaw cycles), 24 h at room temperature, 24 h at 4 °C and 20 days at -80 °C. The samples were stable. The average recovery was 89 %-99 %. This method was used to quantify TMAO and its related metabolites and creatinine levels in hyperlipidemic rats. The results showed that high-fat diet led to the disorder of TMAO and its related metabolites and creatinine in rats, which was effectively improved after the intervention of Rosa Roxburghii Tratt juice(RRTJ). CONCLUSIONS A method for the determination of choline, L-carnitine, betaine, TMA, TMAO and creatinine in plasma, liver and feces samples was established, which is simple, time-saving, high precision, accuracy and recovery.
Collapse
Affiliation(s)
- Changqian Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang 550025, China; Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang 550025, China
| | - Min Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang 550025, China
| | - Shuo Zhang
- Experimental Animal Center of Guizhou Medical University, Guiyang 550025, China
| | - Pengjiao Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang 550025, China
| | - Chencen Lai
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang 550025, China; Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang 550025, China
| | - Duo Meng
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang 550025, China; Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang 550025, China
| | - Zhiyu Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang 550025, China; Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang 550025, China
| | - Xinxin Yi
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang 550025, China; Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang 550025, China
| | - Xiuli Gao
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang 550025, China; Center of Microbiology and Biochemical Pharmaceutical Engineering, Guizhou Medical University, Guiyang 550025, China.
| |
Collapse
|
30
|
Mohammadhasani K, Vahedi Fard M, Mottaghi Moghaddam Shahri A, Khorasanchi Z. Polyphenols improve non-alcoholic fatty liver disease via gut microbiota: A comprehensive review. Food Sci Nutr 2024; 12:5341-5356. [PMID: 39139973 PMCID: PMC11317728 DOI: 10.1002/fsn3.4178] [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: 01/29/2024] [Revised: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 08/15/2024] Open
Abstract
Polyphenols, natural micronutrients derived from plants, are valued for their anti-inflammatory and antioxidant properties. The escalating global prevalence of non-alcoholic fatty liver disease (NAFLD) underscores its status as a chronic progressive liver condition. Furthermore, the dysregulation of gut microbiota (GM) is implicated in the onset and progression of NAFLD through the actions of metabolites such as bile acids (BAs), lipopolysaccharide (LPS), choline, and short-chain fatty acids (SCFAs). Additionally, GM may influence the integrity of the intestinal barrier. This review aims to evaluate the potential effects of polyphenols on GM and intestinal barrier function, and their subsequent impact on NAFLD. We searched through a wide range of databases, such as Web of Science, PubMed, EMBASE, and Scopus to gather information for our non-systematic review of English literature. GM functions and composition can be regulated by polyphenols such as chlorogenic acid, curcumin, green tea catechins, naringenin, quercetin, resveratrol, and sulforaphane. Regulating GM composition improves NAFLD by alleviating inflammation, liver fat accumulation, and liver enzymes. Furthermore, it improves serum lipid profile and gut barrier integrity. All of these components affect NAFLD through the metabolites of GM, including SCFAs, choline, LPS, and BAs. Current evidence indicates that chlorogenic acid, resveratrol, quercetin, and curcumin can modulate GM, improving intestinal barrier integrity and positively impacting NAFLD. More studies are necessary to evaluate the safety and efficacy of naringenin, sulforaphane, and catechin.
Collapse
Affiliation(s)
- Kimia Mohammadhasani
- Department of Nutrition, Food Sciences and Clinical Biochemistry, School of Medicine, Social Determinants of Health Research CenterGonabad University of Medical SciencesGonabadIran
| | - Mohammad Vahedi Fard
- Department of Nutrition, Food Sciences and Clinical Biochemistry, School of Medicine, Social Determinants of Health Research CenterGonabad University of Medical SciencesGonabadIran
| | - Ali Mottaghi Moghaddam Shahri
- International UNESCO Center for Health‐Related Basic Sciences and Human NutritionMashhad University of Medical SciencesMashhadIran
| | - Zahra Khorasanchi
- Department of Nutrition, School of MedicineMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
31
|
Yadav KK, Boley PA, Khatiwada S, Lee CM, Bhandari M, Kenney SP. Development of fatty liver disease model using high cholesterol and low choline diet in white leghorn chickens. Vet Res Commun 2024; 48:2489-2497. [PMID: 38861204 PMCID: PMC11315703 DOI: 10.1007/s11259-024-10420-1] [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: 02/21/2024] [Accepted: 05/17/2024] [Indexed: 06/12/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD), which shows similar symptoms as fatty liver hemorrhage syndrome (FLHS) in chickens, is the most common cause of chronic liver disease and cancer in humans. NAFLD patients and FLHS in chickens have demonstrated severe liver disorders when infected by emerging strains of human hepatitis E virus (HEV) and avian HEV, respectively. We sought to develop a fatty liver disease chicken model by altering the diet of 3-week-old white leghorn chickens. The high cholesterol, and low choline (HCLC) diet included 7.6% fat with additional 2% cholesterol and 800 mg/kg choline in comparison to 5.3% fat, and 1,300 mg/kg choline in the regular diet. Our diet induced fatty liver avian model successfully recapitulates the clinical features seen during NAFLD in humans and FLHS in chickens, including hyperlipidemia and hepatic steatosis, as indicated by significantly higher serum triglycerides, serum cholesterol, liver triglycerides, cholesterol, and fatty acids. By developing this chicken model, we expect to provide a platform to explore the role of lipids in the liver pathology linked with viral infections and contribute to the development of prophylactic interventions.
Collapse
Affiliation(s)
- Kush Kumar Yadav
- Center for Food Animal Health (CFAH), Department of Animal Sciences, The Ohio State University, 1680 Madison Ave, Wooster, OH, 44691, USA
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Patricia A Boley
- Center for Food Animal Health (CFAH), Department of Animal Sciences, The Ohio State University, 1680 Madison Ave, Wooster, OH, 44691, USA
| | - Saroj Khatiwada
- Center for Food Animal Health (CFAH), Department of Animal Sciences, The Ohio State University, 1680 Madison Ave, Wooster, OH, 44691, USA
| | - Carolyn M Lee
- Center for Food Animal Health (CFAH), Department of Animal Sciences, The Ohio State University, 1680 Madison Ave, Wooster, OH, 44691, USA
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Menuka Bhandari
- Center for Food Animal Health (CFAH), Department of Animal Sciences, The Ohio State University, 1680 Madison Ave, Wooster, OH, 44691, USA
| | - Scott P Kenney
- Center for Food Animal Health (CFAH), Department of Animal Sciences, The Ohio State University, 1680 Madison Ave, Wooster, OH, 44691, USA.
- Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
32
|
Chen Q, Fan R, Song L, Wang S, You M, Cai M, Wu Y, Li Y, Xu M. Association of methyl donor nutrients dietary intake and sleep disorders in the elderly revealed by the intestinal microbiome. Food Funct 2024; 15:6335-6346. [PMID: 38832472 DOI: 10.1039/d4fo01303d] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Currently, sleep disorders (SD) in the elderly are gaining prominence globally and are becoming a significant public health concern. Methyl donor nutrients (MDNs), such as vitamin B6, vitamin B12, folate, and choline, have been reported to have the potential to improve sleep disorders. Moreover, MDNs help to maintain gut flora homeostasis, and are closely associated with the development of SD. Nevertheless, there has been a lack of comprehensive human studies examining the association between MDNs intake and SD. In our study, we comprehensively evaluated the association between MDNs intake and SD in the elderly and used 16S rRNA gene sequencing to explore the underlying mechanism. We found that the SD group (n = 91) had a lower methyl-donor nutritional quality index (MNQI) and a trend toward lower intake compared to the control group (n = 147). Based on the intestinal microbiome, the beta diversity of the intestinal flora was higher in the high methyl-donor nutritional quality (HQ) group compared to the low methyl-donor nutritional quality (LQ) group, and it was lower in the SD group compared to the control group. This suggests that MDNs may regulate sleep by modulating the abundance distribution of the microbiota. Subsequently, we performed correlation analyses between the relative abundance of the microbiota, MDNs intake, and the Pittsburgh Sleep Quality Index (PSQI), identifying five genera with potential regulatory roles. The KEGG pathway analysis indicated that energy metabolism and one-carbon metabolism might be the pathways through which MDNs modulate sleep. This study offers dietary guidance strategies for managing SD in the elderly and provides insights for targeted microbiota intervention.
Collapse
Affiliation(s)
- Qianqian Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China.
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Rui Fan
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China.
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Lixia Song
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China.
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Shuyue Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China.
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Mei You
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China.
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Meng Cai
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China.
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Yuxiao Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China.
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Yong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China.
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Meihong Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China.
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| |
Collapse
|
33
|
Bernhard W, Böckmann KA, Minarski M, Wiechers C, Busch A, Bach D, Poets CF, Franz AR. Evidence and Perspectives for Choline Supplementation during Parenteral Nutrition-A Narrative Review. Nutrients 2024; 16:1873. [PMID: 38931230 PMCID: PMC11206924 DOI: 10.3390/nu16121873] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/03/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Choline is an essential nutrient, with high requirements during fetal and postnatal growth. Tissue concentrations of total choline are tightly regulated, requiring an increase in its pool size proportional to growth. Phosphatidylcholine and sphingomyelin, containing a choline headgroup, are constitutive membrane phospholipids, accounting for >85% of total choline, indicating that choline requirements are particularly high during growth. Daily phosphatidylcholine secretion via bile for lipid digestion and very low-density lipoproteins for plasma transport of arachidonic and docosahexaenoic acid to other organs exceed 50% of its hepatic pool. Moreover, phosphatidylcholine is required for converting pro-apoptotic ceramides to sphingomyelin, while choline is the source of betaine as a methyl donor for creatine synthesis, DNA methylation/repair and kidney function. Interrupted choline supply, as during current total parenteral nutrition (TPN), causes a rapid drop in plasma choline concentration and accumulating deficit. The American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.) defined choline as critical to all infants requiring TPN, claiming its inclusion in parenteral feeding regimes. We performed a systematic literature search in Pubmed with the terms "choline" and "parenteral nutrition", resulting in 47 relevant publications. Their results, together with cross-references, are discussed. While studies on parenteral choline administration in neonates and older children are lacking, preclinical and observational studies, as well as small randomized controlled trials in adults, suggest choline deficiency as a major contributor to acute and chronic TPN-associated liver disease, and the safety and efficacy of parenteral choline administration for its prevention. Hence, we call for choline formulations suitable to be added to TPN solutions and clinical trials to study their efficacy, particularly in growing children including preterm infants.
Collapse
Affiliation(s)
- Wolfgang Bernhard
- Department of Neonatology, University Children’s Hospital, 72076 Tübingen, Germany; (W.B.); (K.A.B.); (M.M.); (C.W.); (C.F.P.)
| | - Katrin A. Böckmann
- Department of Neonatology, University Children’s Hospital, 72076 Tübingen, Germany; (W.B.); (K.A.B.); (M.M.); (C.W.); (C.F.P.)
| | - Michaela Minarski
- Department of Neonatology, University Children’s Hospital, 72076 Tübingen, Germany; (W.B.); (K.A.B.); (M.M.); (C.W.); (C.F.P.)
| | - Cornelia Wiechers
- Department of Neonatology, University Children’s Hospital, 72076 Tübingen, Germany; (W.B.); (K.A.B.); (M.M.); (C.W.); (C.F.P.)
| | - Annegret Busch
- Pharmaceutical Department, University Hospital, 72076 Tübingen, Germany; (A.B.); (D.B.)
| | - Daniela Bach
- Pharmaceutical Department, University Hospital, 72076 Tübingen, Germany; (A.B.); (D.B.)
| | - Christian F. Poets
- Department of Neonatology, University Children’s Hospital, 72076 Tübingen, Germany; (W.B.); (K.A.B.); (M.M.); (C.W.); (C.F.P.)
| | - Axel R. Franz
- Department of Neonatology, University Children’s Hospital, 72076 Tübingen, Germany; (W.B.); (K.A.B.); (M.M.); (C.W.); (C.F.P.)
- Center for Pediatric Clinical Studies, University Children’s Hospital, 72076 Tübingen, Germany
| |
Collapse
|
34
|
Mikołajczyk-Stecyna J, Zuk E, Chmurzynska A, Blatkiewicz M, Jopek K, Rucinski M. The effects of exposure to and timing of a choline-deficient diet during pregnancy and early postnatal life on the skeletal muscle transcriptome of the offspring. Clin Nutr 2024; 43:1503-1515. [PMID: 38729079 DOI: 10.1016/j.clnu.2024.05.002] [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: 11/30/2023] [Revised: 04/09/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND & AIMS Nonalcoholic fatty liver disease (NAFLD) is related to muscle loss, but the precise mechanism underlying this association remains unclear. The aim of the present study was thus to determine the influence of maternal fatty liver and dietary choline deficiency during pregnancy and/or lactation periods on the skeletal muscle gene expression profile among 24-day-old male rat offspring. METHODS Histological examination of skeletal muscle tissue specimens obtained from offspring of dams suffering from fatty liver, provided with proper choline intake during pregnancy and lactation (NN), fed a choline-deficient diet during both periods (DD), deprived of choline only during pregnancy (DN), or only during lactation (ND), was performed. The global transcriptome pattern was assessed using a microarray approach (Affymetrix® Rat Gene 2.1 ST Array Strip). The relative expression of selected genes was validated by real-time PCR (qPCR). RESULTS Morphological differences in fat accumulation in skeletal muscle related to choline supply were observed. The global gene expression profile was consistent with abnormal morphological changes. Mettl21c gene was overexpressed in all choline-deficient groups compared to the NN group, while two genes, Cdkn1a and S100a4, were downregulated. Processes of protein biosynthesis were upregulated, and processes related to cell proliferation and lipid metabolism were inhibited in DD, DN, and ND groups compared to the NN group. CONCLUSIONS Prenatal and early postnatal exposure to fatty liver and dietary choline deficiency leads to changes in the transcriptome profile in skeletal muscle of 24-day old male rat offspring and is associated with muscle damage, but the mechanism of it seems to be different at different developmental stages of life. Adequate choline intake during pregnancy and lactation can prevent severe muscle disturbance in the progeny of females suffering from fatty liver.
Collapse
Affiliation(s)
| | - Ewelina Zuk
- Poznań University of Life Sciences, Department of Human Nutrition and Dietetics, Poznań, Poland
| | - Agata Chmurzynska
- Poznań University of Life Sciences, Department of Human Nutrition and Dietetics, Poznań, Poland
| | - Malgorzata Blatkiewicz
- Poznań University of Medical Sciences, Department of Histology and Embryology, Poznań, Poland
| | - Karol Jopek
- Poznań University of Medical Sciences, Department of Histology and Embryology, Poznań, Poland
| | - Marcin Rucinski
- Poznań University of Medical Sciences, Department of Histology and Embryology, Poznań, Poland
| |
Collapse
|
35
|
Gato S, García-Fernández V, Gil-Gómez A, Rojas Á, Montero-Vallejo R, Muñoz-Hernández R, Romero-Gómez M. Navigating the Link Between Non-alcoholic Fatty Liver Disease/Non-alcoholic Steatohepatitis and Cardiometabolic Syndrome. Eur Cardiol 2024; 19:e03. [PMID: 38807856 PMCID: PMC11131154 DOI: 10.15420/ecr.2023.26] [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: 05/26/2023] [Accepted: 12/27/2023] [Indexed: 05/30/2024] Open
Abstract
The global prevalence of non-alcoholic fatty liver disease (NAFLD) is nearly 25% and is increasing rapidly. The spectrum of liver damage in NAFLD ranges from simple steatosis to non-alcoholic steatohepatitis, characterised by the presence of lobular inflammation and hepatocyte ballooning degeneration, with or without fibrosis, which can further develop into cirrhosis and hepatocellular carcinoma. Not only is NAFLD a progressive liver disease, but numerous pieces of evidence also point to extrahepatic consequences. Accumulating evidence suggests that patients with NAFLD are also at increased risk of cardiovascular disease (CVD); in fact, CVDs are the most common cause of mortality in patients with NAFLD. Obesity, type 2 diabetes and higher levels of LDL are common risk factors in both NAFLD and CVD; however, how NAFLD affects the development and progression of CVD remains elusive. In this review, we comprehensively summarise current data on the key extrahepatic manifestations of NAFLD, emphasising the possible link between NAFLD and CVD, including the role of proprotein convertase substilisin/kenin type 9, extracellular vesicles, microbiota, and genetic factors.
Collapse
Affiliation(s)
- Sheila Gato
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSeville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Madrid, Spain
| | - Vanessa García-Fernández
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSeville, Spain
| | - Antonio Gil-Gómez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSeville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Madrid, Spain
| | - Ángela Rojas
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSeville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Madrid, Spain
| | - Rocío Montero-Vallejo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSeville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Madrid, Spain
| | - Rocío Muñoz-Hernández
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSeville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Madrid, Spain
- Departamento de Fisiología, Facultad de Biología, Universidad de SevillaSeville, Spain
| | - Manuel Romero-Gómez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSeville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Madrid, Spain
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario Virgen del RocíoSeville, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de SevillaSeville, Spain
| |
Collapse
|
36
|
Son Y, Kenny TC, Khan A, Birsoy K, Hite RK. Structural basis of lipid head group entry to the Kennedy pathway by FLVCR1. Nature 2024; 629:710-716. [PMID: 38693265 PMCID: PMC11188936 DOI: 10.1038/s41586-024-07374-4] [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: 09/28/2023] [Accepted: 04/02/2024] [Indexed: 05/03/2024]
Abstract
Phosphatidylcholine and phosphatidylethanolamine, the two most abundant phospholipids in mammalian cells, are synthesized de novo by the Kennedy pathway from choline and ethanolamine, respectively1-6. Despite the essential roles of these lipids, the mechanisms that enable the cellular uptake of choline and ethanolamine remain unknown. Here we show that the protein encoded by FLVCR1, whose mutation leads to the neurodegenerative syndrome posterior column ataxia and retinitis pigmentosa7-9, transports extracellular choline and ethanolamine into cells for phosphorylation by downstream kinases to initiate the Kennedy pathway. Structures of FLVCR1 in the presence of choline and ethanolamine reveal that both metabolites bind to a common binding site comprising aromatic and polar residues. Despite binding to a common site, FLVCR1 interacts in different ways with the larger quaternary amine of choline in and with the primary amine of ethanolamine. Structure-guided mutagenesis identified residues that are crucial for the transport of ethanolamine, but dispensable for choline transport, enabling functional separation of the entry points into the two branches of the Kennedy pathway. Altogether, these studies reveal how FLVCR1 is a high-affinity metabolite transporter that serves as the common origin for phospholipid biosynthesis by two branches of the Kennedy pathway.
Collapse
Affiliation(s)
- Yeeun Son
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- BCMB Allied Program, Weill Cornell Graduate School, New York, NY, USA
| | - Timothy C Kenny
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Artem Khan
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Kıvanç Birsoy
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Richard K Hite
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
37
|
Mihajlovic M, Rosseel Z, De Waele E, Vinken M. Parenteral nutrition-associated liver injury: clinical relevance and mechanistic insights. Toxicol Sci 2024; 199:1-11. [PMID: 38383052 DOI: 10.1093/toxsci/kfae020] [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: 02/23/2024] Open
Abstract
Intestinal failure-associated liver disease (IFALD) is a relatively common complication in individuals receiving parenteral nutrition (PN). IFALD can be manifested as different types of liver injury, including steatosis, cholestasis, and fibrosis, and could result in liver failure in some cases. The onset and progression of IFALD are highly dependent on various patient and PN-related risk factors. Despite still being under investigation, several mechanisms have been proposed. Liver injury can originate due to caloric overload, nutrient deficiency, and toxicity, as well as phytosterol content, and omega-6 to omega-3 fatty acids ratio contained in lipid emulsions. Additional mechanisms include immature or defective bile acid metabolism, acute heart failure, infections, and sepsis exerting negative effects via Toll-like receptor 4 and nuclear factor κB inflammatory signaling. Furthermore, lack of enteral feeding, gut dysbiosis, and altered enterohepatic circulation that affect the farnesoid x receptor-fibroblast growth factor 19 axis can also contribute to IFALD. Various best practices can be adopted to minimize the risk of developing IFALD, such as prevention and management of central line infections and sepsis, preservation of intestine's length, a switch to oral and enteral feeding, cyclic PN, avoidance of overfeeding and soybean oil-based lipid formulations, and avoiding hepatotoxic substances. The present review thus provides a comprehensive overview of all relevant aspects inherent to IFALD. Further research focused on clinical observations, translational models, and advanced toxicological knowledge frameworks is needed to gain more insight into the molecular pathogenesis of hepatotoxicity, reduce IFALD incidence, and encourage the safe use of PN.
Collapse
Affiliation(s)
- Milos Mihajlovic
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Zenzi Rosseel
- Department of Pharmacy, Universitair Ziekenhuis Brussel (UZ Brussel), 1090 Brussels, Belgium
- Department of Clinical Nutrition, Universitair Ziekenhuis Brussel (UZ Brussel), 1090 Brussels, Belgium
| | - Elisabeth De Waele
- Department of Clinical Nutrition, Universitair Ziekenhuis Brussel (UZ Brussel), 1090 Brussels, Belgium
- Department of Intensive Care, Universitair Ziekenhuis Brussel (UZ Brussel), 1090 Brussels, Belgium
- Faculty of Medicine and Pharmacy, Department of Clinical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| |
Collapse
|
38
|
Lin Y, Zhang N, Zhang J, Lu J, Liu S, Ma G. The association between hydration state and the metabolism of phospholipids and amino acids among young adults: a metabolomic analysis. Curr Dev Nutr 2024; 8:102087. [PMID: 38425438 PMCID: PMC10904166 DOI: 10.1016/j.cdnut.2024.102087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/15/2024] [Accepted: 01/24/2024] [Indexed: 03/02/2024] Open
Abstract
Background Water is vital for humans' survival and general health, which is involved in various metabolic activities. Objectives The aim of this study was to investigate the variation in urine metabolome and associated metabolic pathways among people with different hydration states. Methods A metabolomic analysis was conducted using 24-h urine samples collected during a cross-sectional study on fluid intake behavior from December 9 to 11, 2021, in Hebei, China. Subjects were divided into the optimal hydration (OH, ≤500 mOsm/kg, n = 21), middle hydration (500-800 mOsm/kg, n = 33), and hypohydration groups (HH, >800 mOsm/kg, n = 13) based on the 3-d average 24-h urine osmolality. Collected 24-h urine samples from 67 subjects (43 males and 34 females) were analyzed for urine metabolome using liquid chromatography-MS. Results The untargeted metabolomic analysis yielded 1055 metabolites by peak intensities. Integrating the results of the orthogonal projections to latent structures discriminant analysis and fold change test, 115 differential metabolites between the OH and HH groups, including phospholipids (PLs) and lysophospholipids, were identified. Among the 115 metabolites identified as differential metabolites, 85 were recorded by the Human Metabolome Database and uploaded to the Kyoto Encyclopedia of Genes and Genomes databases for pathway analysis. Twenty-one metabolic pathways were recognized. Phenylalanine metabolism (0.50, P = 0.007), phenylalanine, tyrosine, and tryptophan biosynthesis (0.50, P = 0.051), glycerophospholipid metabolism (0.31, P < 0.001), sphingolipid metabolism (0.27, P = 0.029), and cysteine and methionine metabolism (0.10, P = 0.066) had the leading pathway impacts. Conclusions We found variations in the urinary PLs and amino acids among subjects with different hydration states. Pathways associated with these differential metabolites could further impact various physiologic and pathologic functions. A more comprehensive and in-depth investigation of the physiologic and pathologic impact of the hydration state and the underlying mechanisms to elucidate and advocate optimal fluid intake habits is needed.This trial was registered at Chinese Clinical Trial Registry as ChiCTR2100045268.
Collapse
Affiliation(s)
- Yongwei Lin
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China
| | - Na Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China
- Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, China
| | - Jianfen Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China
| | - Junbo Lu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China
| | - Shufang Liu
- School of Public Health, Hebei University Health Science Center, Baoding, China
| | - Guansheng Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing, China
- Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, China
| |
Collapse
|
39
|
Abasubong KP, Jiang GZ, Guo HX, Wang X, Huang YY, Li XF, Yan-Zou D, Liu WB, Desouky HE. Effects of a high-fat and high-carbohydrate diet on appetite regulation and central AMPK in the hypothalamus of blunt snout bream (Megalobrama amblycephala). J Anim Physiol Anim Nutr (Berl) 2024; 108:480-492. [PMID: 38014877 DOI: 10.1111/jpn.13908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/29/2023]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is a sensor of cellular energy changes and controls food intake. This study investigates the effect of a high-calorie diet (high fat diet [HFD], high carbohydrate diet [HCD] and high energy diet [HED]) on appetite and central AMPK in blunt snout bream. In the present study, fish (average initial weight 45.84 ± 0.07 g) were fed the control, HFD, HCD and HED in four replicates for 12 weeks. At the end of the feeding trial, the result showed that body mass index, specific growth rate, feed efficiency ratio and feed intake were not affected (p > 0.05) by dietary treatment. However, fish fed the HFD obtained a significantly higher (p < 0.05) lipid productive value, lipid gain and lipid intake than those fed the control diet, but no significant difference was attributed to others. Also, a significantly higher (p < 0.05) energy intake content was found in fish-fed HFD, HCD and HED than those given the control diet. Long-term HFD and HCD feeding significantly increased (p < 0.05) plasma glucose, glycated serum protein, advanced glycation end product, insulin and leptin content levels than the control group. Moreover, a significantly lower (p < 0.05) complex 1, 2 and 3 content was found in fish-fed HFD and HCD than in the control, but no differences (p > 0.05) were attributed to those in HED. Fish-fed HED significantly upregulated (p < 0.05) hypothalamic ampα 1 and ampα 2 expression, whereas the opposite trend was observed in the hypothalamic mammalian target of rapamycin than those in HFD and HCD compared to the control. However, hypothalamic neuropeptide y, peroxisome proliferator-activated receptor α (pparα), acetyl-coa oxidase and carnitine palmitoyltransferase 1 were significantly upregulated (p < 0.05) in the HCD group, while the opposite was seen in cholecystokinin expression compared to those in the control group. Our findings indicated that the central AMPK signal pathway and appetite were modulated according to the diet's energy level to regulate nutritional status and maintain energy homoeostasis in fish.
Collapse
Affiliation(s)
- Kenneth Prudence Abasubong
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Guang-Zhen Jiang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Hui-Xing Guo
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Xi Wang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Yang-Yang Huang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Xiang-Fei Li
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Dong Yan-Zou
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Wen-Bin Liu
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Hesham Eed Desouky
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
- National Laboratory of Animal Science, Nanjing Agricultural University, Nanjing, People's Republic of China
- Department of Animal and Poultry Production, Faculty of Agriculture, Damanhour University, Damanhour, Egypt
| |
Collapse
|
40
|
Sun J, Chen Y, Wang T, Ali W, Ma Y, Yuan Y, Gu J, Bian J, Liu Z, Zou H. Baicalin and N-acetylcysteine regulate choline metabolism via TFAM to attenuate cadmium-induced liver fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 125:155337. [PMID: 38241915 DOI: 10.1016/j.phymed.2024.155337] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/14/2023] [Accepted: 01/05/2024] [Indexed: 01/21/2024]
Abstract
(Background): Cadmium is an environmental pollutant associated with several liver diseases. Baicalin and N-Acetylcysteine have antioxidant and hepatoprotective effects. (Purpose): However, it is unclear whether baicalin and N-Acetylcysteine can alleviate Cadmium -induced liver fibrosis by regulating metabolism, or whether they exert a synergistic effect. (Study design): We treated Cadmium-poisoned mice with baicalin, N-Acetylcysteine, or baicalin+ N-Acetylcysteine. We studied the effects of baicalin and N-Acetylcysteine on Cadmium-induced liver fibers and their specific mechanisms. (Methods): We used C57BL/6 J mice, and AML12, and HSC-6T cells to establish in vitro assays and in vivo models. (Results): Metabolomics was used to detect the effect of baicalin and N-Acetylcysteine on liver metabolism, which showed that compared with the control group, the Cadmium group had increased fatty acid and amino acid levels, with significantly reduced choline and acetylcholine contents. Baicalin and N-Acetylcysteine alleviated these Cadmium-induced metabolic changes. We further showed that choline alleviated Cadmium -induced liver inflammation and fibrosis. In addition, cadmium significantly promoted extracellular leakage of lactic acid, while choline alleviated the cadmium -induced destruction of the cell membrane structure and lactic acid leakage. Western blotting showed that cadmium significantly reduced mitochondrial transcription factor A (TFAM) and Choline Kinase α(CHKα2) levels, and baicalin and N-Acetylcysteine reversed this effect. Overexpression of Tfam in mouse liver and AML12 cells increased the expression of CHKα2 and the choline content, alleviating and cadmium-induced lactic acid leakage, liver inflammation, and fibrosis. (Conclusion): Overall, baicalin and N-Acetylcysteine alleviated cadmium-induced liver damage, inflammation, and fibrosis to a greater extent than either drug alone. TFAM represents a target for baicalin and N-Acetylcysteine, and alleviated cadmium-induced liver inflammation and fibrosis by regulating hepatic choline metabolism.
Collapse
Affiliation(s)
- Jian Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Yan Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Tao Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Waseem Ali
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China
| | - Yonggang Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| |
Collapse
|
41
|
Garnick L, Bates C, Massarsky A, Spencer P, Sura P, Monnot AD, Maier A. Developmental and reproductive toxicity hazard characterization of 2-amino-2-methyl-1-propanol (AMP). J Appl Toxicol 2024; 44:316-332. [PMID: 37715655 DOI: 10.1002/jat.4539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/26/2023] [Accepted: 08/18/2023] [Indexed: 09/18/2023]
Abstract
2-Amino-2-methyl-1-propanol (AMP™) is a widely used pH stabilizer in personal care products (PCPs); thus, the safety implications of dermal AMP exposure remain of interest. We have previously reported that exposure to AMP in PCPs when used as intended is not anticipated to result in an increased risk of hepatotoxicity (primarily steatosis and altered phospholipid homeostasis). The current study focuses on AMP in PCP's potential for developmental and reproductive toxicity (DART) in humans, based on data from animal studies. Animal studies suggest that exposure to AMP can result in post-implantation loss. However, such effects occur at maternally toxic doses, posing a challenge for determining appropriate hazard classifications in the context of relevant consumer use scenarios. Our assessment concluded that human exposure to AMP in PCPs is not anticipated to result in DART at non-maternally toxic doses. Further, mode of action (MOA) analysis elucidated the potential biological pathways underlying DART effects observed in high-dose animal studies, such that perturbation of uterine choline synthesis was the most well-supported MOA hypothesis. Downstream uterine effects might reflect choline-dependent changes in epigenetic control of pathways important for implantation maintenance and uterine cell energetics. Since AMP-induced post-implantation loss occurs at doses higher than pathology related to liver toxicity, maintaining AMP exposures from exceeding the onset dose for maternal liver effects will also be protective of DART effects. Furthermore, dermal exposure to AMP expected from the use of PCPs is highly unlikely to result in toxicologically significant systemic AMP concentrations; thus, DART is not anticipated.
Collapse
Affiliation(s)
| | | | | | | | - Priyanka Sura
- ANGUS Chemical Company, Buffalo Grove, Illinois, USA
| | | | | |
Collapse
|
42
|
Nguyen XTA, Le TNU, Nguyen TQ, Thi Thuy Ha H, Artati A, Leong NCP, Nguyen DT, Lim PY, Susanto AV, Huang Q, Fam L, Leong LN, Bonne I, Lee A, Granadillo JL, Gooch C, Yu D, Huang H, Soong TW, Chang MW, Wenk MR, Adamski J, Cazenave-Gassiot A, Nguyen LN. MFSD7c functions as a transporter of choline at the blood-brain barrier. Cell Res 2024; 34:245-257. [PMID: 38302740 PMCID: PMC10907603 DOI: 10.1038/s41422-023-00923-y] [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: 12/18/2023] [Accepted: 12/26/2023] [Indexed: 02/03/2024] Open
Abstract
Mutations in the orphan transporter MFSD7c (also known as Flvcr2), are linked to Fowler syndrome. Here, we used Mfsd7c knockout (Mfsd7c-/-) mice and cell-based assays to reveal that MFSD7c is a choline transporter at the blood-brain barrier (BBB). We performed comprehensive metabolomics analysis and detected differential changes of metabolites in the brains and livers of Mfsd7c-/-embryos. Particularly, we found that choline-related metabolites were altered in the brains but not in the livers of Mfsd7c-/- embryos. Thus, we hypothesized that MFSD7c regulates the level of choline in the brain. Indeed, expression of human MFSD7c in cells significantly increased choline uptake. Interestingly, we showed that choline uptake by MFSD7c is greatly increased by choline-metabolizing enzymes, leading us to demonstrate that MFSD7c is a facilitative transporter of choline. Furthermore, single-cell patch clamp analysis showed that the import of choline by MFSD7c is electrogenic. Choline transport function of MFSD7c was shown to be conserved in vertebrates, but not in yeasts. We demonstrated that human MFSD7c is a functional ortholog of HNM1, the yeast choline importer. We also showed that several missense mutations identified in patients exhibiting Fowler syndrome had abolished or reduced choline transport activity. Mice lacking Mfsd7c in endothelial cells of the central nervous system suppressed the import of exogenous choline from blood but unexpectedly had increased choline levels in the brain. Stable-isotope tracing study revealed that MFSD7c was required for exporting choline derived from lysophosphatidylcholine in the brain. Collectively, our work identifies MFSD7c as a choline exporter at the BBB and provides a foundation for future work to reveal the disease mechanisms of Fowler syndrome.
Collapse
Affiliation(s)
- Xuan Thi Anh Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Thanh Nha Uyen Le
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Toan Q Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hoa Thi Thuy Ha
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Anna Artati
- Metabolomics and Proteomics Core, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Nancy C P Leong
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dat T Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Pei Yen Lim
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Adelia Vicanatalita Susanto
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Qianhui Huang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ling Fam
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lo Ngah Leong
- Electron Microscopy Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Isabelle Bonne
- Electron Microscopy Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Life Sciences Institute, Immunology Programme, National University of Singapore, Singapore, Singapore
| | - Angela Lee
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University in St Louis, Saint Louis, MO, USA
| | - Jorge L Granadillo
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University in St Louis, Saint Louis, MO, USA
| | - Catherine Gooch
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University in St Louis, Saint Louis, MO, USA
| | - Dejie Yu
- Electrophysiology Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hua Huang
- Electrophysiology Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Diseases Program, National University of Singapore, Singapore, Singapore
| | - Tuck Wah Soong
- Electrophysiology Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Diseases Program, National University of Singapore, Singapore, Singapore
| | - Matthew Wook Chang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Markus R Wenk
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Jerzy Adamski
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Amaury Cazenave-Gassiot
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Long N Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore.
- Life Sciences Institute, Immunology Programme, National University of Singapore, Singapore, Singapore.
- Cardiovascular Diseases Program, National University of Singapore, Singapore, Singapore.
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
43
|
Wen Y, Luo Y, Qiu H, Chen B, Huang J, Lv S, Wang Y, Li J, Tao L, Yang B, Li K, He L, He M, Yang Q, Yu Z, Xiao W, Zhao M, Zou X, Lu R, Gu C. Gut microbiota affects obesity susceptibility in mice through gut metabolites. Front Microbiol 2024; 15:1343511. [PMID: 38450171 PMCID: PMC10916699 DOI: 10.3389/fmicb.2024.1343511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
Abstract
Introduction It is well-known that different populations and animals, even experimental animals with the same rearing conditions, differ in their susceptibility to obesity. The disparity in gut microbiota could potentially account for the variation in susceptibility to obesity. However, the precise impact of gut microbiota on gut metabolites and its subsequent influence on susceptibility to obesity remains uncertain. Methods In this study, we established obesity-prone (OP) and obesity-resistant (OR) mouse models by High Fat Diet (HFD). Fecal contents of cecum were examined using 16S rDNA sequencing and untargeted metabolomics. Correlation analysis and MIMOSA2 analysis were used to explore the association between gut microbiota and intestinal metabolites. Results After a HFD, gut microbiota and gut metabolic profiles were significantly different between OP and OR mice. Gut microbiota after a HFD may lead to changes in eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), a variety of branched fatty acid esters of hydroxy fatty acids (FAHFAs) and a variety of phospholipids to promote obesity. The bacteria g_Akkermansia (Greengene ID: 175696) may contribute to the difference in obesity susceptibility through the synthesis of glycerophosphoryl diester phosphodiesterase (glpQ) to promote choline production and the synthesis of valyl-tRNA synthetase (VARS) which promotes L-Valine degradation. In addition, gut microbiota may affect obesity and obesity susceptibility through histidine metabolism, linoleic acid metabolism and protein digestion and absorption pathways.
Collapse
Affiliation(s)
- Yuhang Wen
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Yadan Luo
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Hao Qiu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Baoting Chen
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Jingrong Huang
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Shuya Lv
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Yan Wang
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Jiabi Li
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Lingling Tao
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Bailin Yang
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Ke Li
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Lvqin He
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Manli He
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Qian Yang
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Zehui Yu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Wudian Xiao
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Mingde Zhao
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| | - Xiaoxia Zou
- Suining First People's Hospital, Suining, China
| | - Ruilin Lu
- Suining First People's Hospital, Suining, China
| | - Congwei Gu
- Laboratory Animal Centre, Southwest Medical University, Luzhou, China
- Model Animal and Human Disease Research of Luzhou Key Laboratory, Luzhou, China
| |
Collapse
|
44
|
Zhong H, Dong J, Zhu L, Mao J, Dong J, Zhao Y, Zou Y, Guo M, Ding G. Non-alcoholic fatty liver disease: pathogenesis and models. Am J Transl Res 2024; 16:387-399. [PMID: 38463579 PMCID: PMC10918142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 01/22/2024] [Indexed: 03/12/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a complex disease characterized by a massive accumulation of lipids in the liver, with a continuous progression of simple steatosis, non-alcoholic steatohepatitis (NASH), cirrhosis, and hepatocellular carcinoma. Non-alcoholic fatty liver disease is associated with obesity, insulin resistance, and metabolic syndrome; it is a severe public health risk and is currently the most common liver disease of the world. In addition to the fatty infiltration of the liver in non-alcoholic fatty liver disease patients, the field of liver transplantation faces similar obstacles. NAFLD and NASH primarily involve lipotoxicity, inflammation, oxidative stress, and insulin resistance. However, the precise mechanisms and treatments remain unclear. Therapeutic approaches encompass exercise, weight control, as well as treatments targeting antioxidants and anti-inflammatory pathways. The role of animal models in research has become crucial as a key tool to explore the molecular mechanisms and potential treatments for non-alcoholic fatty liver disease and non-alcoholic steatohepatitis. Here, we summarized the current understanding of the pathogenesis of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis and discussed animal models commonly used in recent years.
Collapse
Affiliation(s)
- Hanxiang Zhong
- Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Navy Medical UniversityShanghai, China
| | - Jiayong Dong
- Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Navy Medical UniversityShanghai, China
| | - Liye Zhu
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical UniversityShanghai, China
| | - Jiaxi Mao
- Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Navy Medical UniversityShanghai, China
| | - Junfeng Dong
- Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Navy Medical UniversityShanghai, China
| | - Yuanyu Zhao
- Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Navy Medical UniversityShanghai, China
| | - You Zou
- Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Navy Medical UniversityShanghai, China
| | - Meng Guo
- Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Navy Medical UniversityShanghai, China
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical UniversityShanghai, China
| | - Guoshan Ding
- Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Navy Medical UniversityShanghai, China
| |
Collapse
|
45
|
Song YF, Bai ZY, Luo Z, Wang LJ, Zheng H. Choline-mediated hepatic lipid homoeostasis in yellow catfish: unravelling choline's lipotropic and methyl donor functions and significance of ire-1α signalling pathway. Br J Nutr 2024; 131:202-213. [PMID: 37642130 DOI: 10.1017/s000711452300185x] [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] [Indexed: 08/31/2023]
Abstract
Choline plays a crucial role in hepatic lipid homeostasis by acting as a major methyl-group donor. However, despite this well-accepted fact, no study has yet explored how choline's methyl-donor function contributes to preventing hepatic lipid dysregulation. Moreover, the potential regulatory role of Ire-1α, an ER-transmembrane transducer for the unfolded protein response (UPRer), in choline-mediated hepatic lipid homeostasis remains unexplored. Thus, this study investigated the mechanism by which choline prevents hepatic lipid dysregulation, focusing on its role as a methyl-donor and the involvement of Ire-1α in this process. To this end, a model animal for lipid metabolism, yellow catfish (Pelteobagrus fulvidraco) were fed two different diets (adequate or deficient choline diets) in vivo for 10 weeks. The key findings of studies are as follows: 1. Dietary choline, upregulated selected lipolytic and fatty acid β-oxidation transcripts promoting hepatic lipid homeostasis. 2. Dietary choline ameliorated UPRer and prevented hepatic lipid dysregulation mainly through ire-1α signalling, not perk or atf-6α signalling. 3. Choline inhibited the transcriptional expression level of ire-1α by activating site-specific DNA methylations in the promoter of ire-1α. 4. Choline-mediated ire-1α methylations reduced Ire-1α/Fas interactions, thereby further inhibiting Fas activity and reducing lipid droplet deposition. These results offer a novel insight into the direct and indirect regulation of choline on lipid metabolism genes and suggests a potential crosstalk between ire-1α signalling and choline-deficiency-induced hepatic lipid dysregulation, highlighting the critical contribution of choline as a methyl-donor in maintaining hepatic lipid homeostasis.
Collapse
Affiliation(s)
- Yu-Feng Song
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan430070, People's Republic of China
| | - Zhen-Yu Bai
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan430070, People's Republic of China
| | - Zhi Luo
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan430070, People's Republic of China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao266237, People's Republic of China
| | - Ling-Jiao Wang
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan430070, People's Republic of China
| | - Hua Zheng
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan430070, People's Republic of China
| |
Collapse
|
46
|
Jung IR, Ahima RS, Kim SF. Time-Restricted Feeding Ameliorates Methionine-Choline Deficient Diet-Induced Steatohepatitis in Mice. Int J Mol Sci 2024; 25:1390. [PMID: 38338668 PMCID: PMC10855189 DOI: 10.3390/ijms25031390] [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: 12/04/2023] [Revised: 01/20/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is an inflammatory form of non-alcoholic fatty liver disease (NAFLD), closely associated with disease progression, cirrhosis, liver failure, and hepatocellular carcinoma. Time-restricted feeding (TRF) has been shown to decrease body weight and adiposity and improve metabolic outcomes; however, the effect of TRF on NASH has not yet been fully understood. We had previously reported that inositol polyphosphate multikinase (IPMK) mediates hepatic insulin signaling. Importantly, we have found that TRF increases hepatic IPMK levels. Therefore, we investigated whether there is a causal link between TRF and IPMK in a mouse model of NASH, i.e., methionine- and choline-deficient diet (MCDD)-induced steatohepatitis. Here, we show that TRF alleviated markers of NASH, i.e., reduced hepatic steatosis, liver triglycerides (TG), serum alanine transaminase (ALT) and aspartate aminotransferase (AST), inflammation, and fibrosis in MCDD mice. Interestingly, MCDD led to a significant reduction in IPMK levels, and the deletion of hepatic IPMK exacerbates the NASH phenotype induced by MCDD, accompanied by increased gene expression of pro-inflammatory chemokines. Conversely, TRF restored IPMK levels and significantly reduced gene expression of proinflammatory cytokines and chemokines. Our results demonstrate that TRF attenuates MCDD-induced NASH via IPMK-mediated changes in hepatic steatosis and inflammation.
Collapse
Affiliation(s)
| | - Rexford S. Ahima
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University, Baltimore, MD 21218, USA;
| | - Sangwon F. Kim
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University, Baltimore, MD 21218, USA;
| |
Collapse
|
47
|
Oh J, Kim J, Lee S, Park G, Baritugo KAG, Han KJ, Lee S, Sung GH. 1H NMR Serum Metabolomic Change of Trimethylamine N-oxide (TMAO) Is Associated with Alcoholic Liver Disease Progression. Metabolites 2024; 14:39. [PMID: 38248842 PMCID: PMC10818766 DOI: 10.3390/metabo14010039] [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: 11/13/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
Without early detection and treatment, chronic and excessive alcohol consumption can lead to the development of alcoholic liver disease (ALD). With this in mind, we exploit the recent concept of the liver-gut axis and analyze the serum profile of ALD patients for identification of microbiome-derived metabolites that can be used as diagnostic biomarkers for onset of ALD. 1H-NMR was used to analyze serum metabolites of 38 ALD patients that were grouped according to their Child-Turcotte-Pugh scores (CTP): class A (CTP-A; 19), class B(CTP-B; 10), and class C (CTP-C; 9). A partial least squares-discriminant analysis (PLS-DA) and a variable importance of projection (VIP) score were used to identify significant metabolites. A receiver operating characteristic (ROC) curve and correlation heatmap were used to evaluate the predictability of identified metabolites as ALD biomarkers. Among 42 identified metabolites, 6 were significantly correlated to exacerbation of ALD. As ALD progressed in CTP-C, the levels of trimethylamine N-oxide (TMAO), malate, tyrosine, and 2-hydroxyisovalerate increased, while isobutyrate and isocitrate decreased. Out of six metabolites, elevated levels of TMAO and its precursors (carnitine, betaine, choline) were associated with severity of ALD. This indicates that TMAO can be used as an effective biomarker for the diagnosis of ALD progression.
Collapse
Affiliation(s)
- Junsang Oh
- Biomedical Institute of Mycological Resource, International St. Mary’s Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea; (J.O.); (J.K.); (K.-A.G.B.)
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Gang-won-do, Republic of Korea
| | - Jayoung Kim
- Biomedical Institute of Mycological Resource, International St. Mary’s Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea; (J.O.); (J.K.); (K.-A.G.B.)
- Department of Laboratory Medicine, International St. Mary’s Hospital and College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea
| | - Sanghak Lee
- Department of Biomedical Science, Graduate School, Catholic Kwandong University, Gangneung-si 25601, Gang-won-do, Republic of Korea; (S.L.); (G.P.)
| | - Gyubin Park
- Department of Biomedical Science, Graduate School, Catholic Kwandong University, Gangneung-si 25601, Gang-won-do, Republic of Korea; (S.L.); (G.P.)
| | - Kei-Anne Garcia Baritugo
- Biomedical Institute of Mycological Resource, International St. Mary’s Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea; (J.O.); (J.K.); (K.-A.G.B.)
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Gang-won-do, Republic of Korea
| | - Ki Jun Han
- Department of Internal Medicine, International St. Mary’s Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea;
| | - Sangheun Lee
- Biomedical Institute of Mycological Resource, International St. Mary’s Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea; (J.O.); (J.K.); (K.-A.G.B.)
- Department of Internal Medicine, International St. Mary’s Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea;
| | - Gi-Ho Sung
- Biomedical Institute of Mycological Resource, International St. Mary’s Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea; (J.O.); (J.K.); (K.-A.G.B.)
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Gang-won-do, Republic of Korea
| |
Collapse
|
48
|
Maj MA, Burrin DG, Manjarín R. Decreased FXR Agonism in the Bile Acid Pool Is Associated with Impaired FXR Signaling in a Pig Model of Pediatric NAFLD. Biomedicines 2023; 11:3303. [PMID: 38137523 PMCID: PMC10740974 DOI: 10.3390/biomedicines11123303] [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: 10/23/2023] [Revised: 11/28/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
The objective of this study was to investigate whether the impairment of farnesoid X receptor (FXR)-fibroblast growth factor 19 (FGF19) signaling in juvenile pigs with non-alcoholic fatty liver disease (NAFLD) is associated with changes in the composition of the enterohepatic bile acid pool. Eighteen 15-day-old Iberian pigs, pair-housed in pens, were allocated to receive either a control (CON) or high-fructose, high-fat (HFF) diet. Animals were euthanized in week 10, and liver, blood, and distal ileum (DI) samples were collected. HFF-fed pigs developed NAFLD and had decreased FGF19 expression in the DI and lower FGF19 levels in the blood. Compared with the CON, the HFF diet increased the total cholic acid (CA) and the CA to chenodeoxycholic acid (CDCA) ratio in the liver, DI, and blood. CA and CDCA levels in the DI were negatively and positively correlated with ileal FGF19 expression, respectively, and blood levels of FGF19 decreased with an increasing ileal CA to CDCA ratio. Compared with the CON, the HFF diet increased the gene expression of hepatic 12-alpha-hydrolase, which catalyzes the synthesis of CA in the liver. Since CA species are weaker FXR ligands than CDCA, our results suggest that impairment of FXR-FGF19 signaling in NAFLD pigs is associated with a decrease in FXR agonism in the bile acid pool.
Collapse
Affiliation(s)
- Magdalena A. Maj
- Department of Biological Sciences, California Polytechnic State University, San Luis Obispo, CA 93407, USA
- Center for Applications in Biotechnology, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Douglas G. Burrin
- USDA-ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Rodrigo Manjarín
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, CA 93407, USA;
| |
Collapse
|
49
|
Mims TS, Kumari R, Leathem C, Antunes K, Joseph S, Yen MI, Ferstl D, Jamieson SM, Sabbar A, Biebel C, Lazarevic N, Willis NB, Henry L, Yen CLE, Smith JP, Gosain A, Meisel M, Willis KA, Talati AJ, Elabiad MT, Hibl B, Pierre JF. Altered hepatic and intestinal homeostasis in a neonatal murine model of short-term total parenteral nutrition and antibiotics. Am J Physiol Gastrointest Liver Physiol 2023; 325:G556-G569. [PMID: 37753583 PMCID: PMC11901332 DOI: 10.1152/ajpgi.00129.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/18/2023] [Accepted: 09/24/2023] [Indexed: 09/28/2023]
Abstract
Parenteral nutrition (PN) prevents starvation and supports metabolic requirements intravenously when patients are unable to be fed enterally. Clinically, infants are frequently provided PN in intensive care settings along with exposure to antibiotics (ABX) to minimize infection during care. Unfortunately, neonates experience extremely high rates of hepatic complications. Adult rodent and piglet models of PN are well-established but neonatal models capable of leveraging the considerable transgenic potential of the mouse remain underdeveloped. Utilizing our newly established neonatal murine PN mouse model, we administered ABX or controlled drinking water to timed pregnant dams to disrupt the maternal microbiome. We randomized mouse pups to PN or sham surgery controls +/- ABX exposure. ABX or short-term PN decreased liver and brain organ weights, intestinal length, and mucosal architecture (vs. controls). PN significantly elevated evidence of hepatic proinflammatory markers, neutrophils and macrophage counts, bacterial colony-forming units, and evidence of cholestasis risk, which was blocked by ABX. However, ABX uniquely elevated metabolic regulatory genes resulting in accumulation of hepatocyte lipids, triglycerides, and elevated tauro-chenoxycholic acid (TCDCA) in serum. Within the gut, PN elevated the relative abundance of Akkermansia, Enterococcus, and Suterella with decreased Anaerostipes and Lactobacillus compared with controls, whereas ABX enriched Proteobacteria. We conclude that short-term PN elevates hepatic inflammatory stress and risk of cholestasis in early life. Although concurrent ABX exposure protects against hepatic immune activation during PN, the dual exposure modulates metabolism and may contribute toward early steatosis phenotype, sometimes observed in infants unable to wean from PN.NEW & NOTEWORTHY This study successfully established a translationally relevant, murine neonatal parenteral nutrition (PN) model. Short-term PN is sufficient to induce hepatitis-associated cholestasis in a neonatal murine model that can be used to understand disease in early life. The administration of antibiotics during PN protects animals from bacterial translocation and proinflammatory responses but induces unique metabolic shifts that may predispose the liver toward early steatosis.
Collapse
Affiliation(s)
- Tahliyah S Mims
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Roshan Kumari
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Cameron Leathem
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Karen Antunes
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Sydney Joseph
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Mei-I Yen
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Danielle Ferstl
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Sophia M Jamieson
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Austin Sabbar
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Claudia Biebel
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Nikolai Lazarevic
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Nathaniel B Willis
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Lydia Henry
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Chi-Liang E Yen
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Joseph P Smith
- Department of Pharmacy, University of Wisconsin Hospitals and Clinics, Madison, Wisconsin, United States
| | - Ankush Gosain
- Department of Pediatric Surgery, Children's Hospital of Colorado, Denver, Colorado, United States
| | - Marlies Meisel
- Department of Immunology, University of Pittsburg, Pittsburg, Pennsylvania, United States
| | - Kent A Willis
- Division of Neonatology, Department of Pediatrics, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Ajay J Talati
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Mohammad T Elabiad
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Brianne Hibl
- Department of Comparative Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Joseph F Pierre
- Department of Nutritional Sciences, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, Wisconsin, United States
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States
| |
Collapse
|
50
|
Chai C, Chen L, Deng MG, Liang Y, Liu F, Nie JQ. Dietary choline intake and non-alcoholic fatty liver disease (NAFLD) in U.S. adults: National Health and Nutrition Examination Survey (NHANES) 2017-2018. Eur J Clin Nutr 2023; 77:1160-1166. [PMID: 37634048 DOI: 10.1038/s41430-023-01336-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 08/28/2023]
Abstract
BACKGROUND Whether there is an association between dietary choline intake and non-alcoholic fatty liver disease (NAFLD) in American adults remains unclear. METHODS Data came from the National Health and Nutrition Examination Survey 2017-2018. Choline intake was defined by the mean amounts of two 24 h dietary recalls, and choline intake was categorized into three groups according to the quartiles: inadequate ( P75). Hepatic steatosis was assessed with FibroScan®, in which VCTE was employed with controlled attenuation to derive the controlled attenuation parameter (CAP), and NAFLD was defined as a CAP score ≥285 dB/m. Multivariable linear regression was performed to assess the linear relationship between choline intake and CAP. Multivariable logistics regression models were conducted to assess the association between choline intake status and NAFLD in the final sample and subgroup analysis was then performed in men and women. RESULTS The amount of dietary choline was inversely associated with CAP score (β = -0.262, 95% CI: -0.280, -0.245). Compared to inadequate choline intake, optimal choline intake was related to a lower risk of NAFLD (OR: 0.705, 95% CI: 0.704-0.706) in the final sample. Subgroup analysis by gender revealed that the highest choline intake status was associated with a lower risk of NAFLD both in females (OR: 0.764, 95% CI: 0.762-0.766), and males (OR: 0.955, 95% CI: 0.953-0.958) when compared to the lowest choline intake. CONCLUSIONS With the latest NHANES data, we found that higher dietary choline was associated with a lower risk of NAFLD in American adults, and such a relationship exists in both females and males.
Collapse
Affiliation(s)
- Chen Chai
- Emergency Center, Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Lin Chen
- Emergency Department, Xiantao First People's Hospital Affiliated to Changjiang University, Xiantao, China
| | - Ming-Gang Deng
- School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Yuehui Liang
- School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Fang Liu
- School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Jia-Qi Nie
- School of Public Health, Wuhan University, Wuhan, 430071, China
| |
Collapse
|