1
|
Lim J, Vujkovic M, Levin MG, Lorenz K, Voight BF, Zhang DY, Dudek MF, Pahl MC, Pippin JA, Su C, Manduchi E, Wells AD, Grant SF, Abramowitz S, Damrauer SM, Mukherjee S, Yang G, Kaplan DE, Penn Medicine BioBank, Rader DJ. Trans-ancestry genome-wide association meta-analysis of gallstone disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.16.25324077. [PMID: 40166541 PMCID: PMC11957090 DOI: 10.1101/2025.03.16.25324077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Gallstone disease is a highly prevalent and costly gastrointestinal disease. Yet, genetic variation in susceptibility to gallstone disease and its implication in metabolic regulatory pathways remain to be explored. We report a trans-ancestry genome-wide association meta-analysis of gallstone disease including 88,063 cases and 1,490,087 controls in the UK Biobank, FinnGen, Biobank Japan, and Million Veteran Program. We identified 91 (37 novel) risk loci across the meta-analysis and found replication in statistically compelling signals in the All of Us Research Program. A polygenic risk score constructed from trans-ancestry lead variants was positively associated with liver chemistry and alpha-1-antitrypsin deficiency and negatively associated with total cholesterol and low-density lipoprotein levels among trans-ancestry and European ancestry groups in the Penn Medicine BioBank. Cross-trait colocalization analysis between risk loci and 44 liver, metabolic, renal, and inflammatory traits yielded 350 significant colocalizations as well as 97 significant colocalizations and 65 prioritized genes from expression quantitative trait loci from eight tissues. These findings broaden our understanding of the genetic architecture of gallstone disease.
Collapse
Affiliation(s)
- Junghyun Lim
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Division of Translational Medicine and Human Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Marijana Vujkovic
- Division of Translational Medicine and Human Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Michael G. Levin
- Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kim Lorenz
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Benjamin F. Voight
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - David Y. Zhang
- Division of Translational Medicine and Human Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Max F. Dudek
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Graduate Group in Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew C. Pahl
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - James A. Pippin
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Chun Su
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Elisabetta Manduchi
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Andrew D. Wells
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sarah Abramowitz
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Scott M. Damrauer
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Samiran Mukherjee
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Guoyi Yang
- Division of Translational Medicine and Human Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - David E. Kaplan
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | | | - Daniel J. Rader
- Division of Translational Medicine and Human Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
2
|
Ma Y, Jing X, Li D, Zhang T, Xiang H, Xia Y, Xu F. Proteomics and metabolomics analyses of urine for investigation of gallstone disease in a high-altitude area. Metabolomics 2024; 20:99. [PMID: 39143352 DOI: 10.1007/s11306-024-02162-7] [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: 04/24/2024] [Accepted: 07/29/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND The incidence of gallstones is high in Qinghai Province. However, the molecular mechanisms underlying the development of gallstones remain unclear. METHODS In this study, we collected urine samples from 30 patients with gallstones and 30 healthy controls. The urine samples were analysed using multi-omics platforms. Proteomics analysis was conducted using data-independent acquisition, whereas metabolomics analysis was performed using liquid chromatography-mass spectrometry (LC-MS). RESULTS Among the patients with gallstones, we identified 49 down-regulated and 185 up-regulated differentially expressed proteins as well as 195 up-regulated and 189 down-regulated differentially expressed metabolites. Six pathways were significantly enriched: glycosaminoglycan degradation, arginine and proline metabolism, histidine metabolism, pantothenate and coenzyme A biosynthesis, drug metabolism-other enzymes, and the pentose phosphate pathway. Notably, 10 differentially expressed proteins and metabolites showed excellent predictive performance and were selected as potential biomarkers. CONCLUSION The findings of our metabolomics and proteomics analyses provide new insights into novel biomarkers for patients with cholelithiasis in high-altitude areas.
Collapse
Affiliation(s)
- Ying Ma
- Department of Hepatobiliary Surgery, Qinghai Provincial Traffic Hospital, Xining, 810001, Qinghai, China
| | - Xiaofeng Jing
- Department of Evidence-Based Medicine and Social Medicine, School of Public Health , Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Defu Li
- Department of Hepatobiliary Surgery, Qinghai Provincial Traffic Hospital, Xining, 810001, Qinghai, China
| | - Tiecheng Zhang
- Department of Evidence-Based Medicine and Social Medicine, School of Public Health , Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Haiqi Xiang
- Department of Evidence-Based Medicine and Social Medicine, School of Public Health , Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Yonghong Xia
- Department of Hepatobiliary Surgery, Qinghai Provincial Traffic Hospital, Xining, 810001, Qinghai, China.
| | - Fan Xu
- Department of Evidence-Based Medicine and Social Medicine, School of Public Health , Chengdu Medical College, Chengdu, 610500, Sichuan, China.
| |
Collapse
|
3
|
Jiang YC, Dou JY, Xuan MY, Gao C, Li ZX, Lian LH, Cui ZY, Nan JX, Wu YL. Raspberry Ketone Attenuates Hepatic Fibrogenesis and Inflammation via Regulating the Crosstalk of FXR and PGC-1α Signaling. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:15740-15754. [PMID: 38970822 DOI: 10.1021/acs.jafc.4c03286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/08/2024]
Abstract
Hepatic fibrosis is a compensatory response to chronic liver injury and inflammation, and dietary intervention is recommended as one of the fundamental prevention strategies. Raspberry ketone (RK) is an aromatic compound first isolated from raspberry and widely used to prepare food flavors. The current study investigated the hepatoprotection and potential mechanism of RK against hepatic fibrosis. In vitro, hepatic stellate cell (HSC) activation was stimulated with TGF-β and cultured with RK, farnesoid X receptor (FXR), or peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) agonist or inhibitor, respectively. In vivo, C57BL/6 mice were injected intraperitoneally with thioacetamide (TAA) at 100/200 mg/kg from the first to the fifth week. Mice were intragastrically administrated with RK or Cur once a day from the second to the fifth week. In activated HSCs, RK inhibited extracellular matrix (ECM) accumulation, inflammation, and epithelial-mesenchymal transition (EMT) process. RK both activated FXR/PGC-1α and regulated their crosstalk, which were verified by their inhibitors and agonists. Deficiency of FXR or PGC-1α also attenuated the effect of RK on the reverse of activated HSCs. RK also decreased serum ALT/AST levels, liver histopathological change, ECM accumulation, inflammation, and EMT in mice caused by TAA. Double activation of FXR/PGC-1α might be the key targets for RK against hepatic fibrosis. Above all, these discoveries supported the potential of RK as a novel candidate for the dietary intervention of hepatic fibrosis.
Collapse
Affiliation(s)
- Yu-Chen Jiang
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Jia-Yi Dou
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Mei-Yan Xuan
- School of Pharmaceutical Sciences, Josai University, Sakado, Saitama 350-0295, Japan
| | - Chong Gao
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Zhao-Xu Li
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Li-Hua Lian
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Zhen-Yu Cui
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
- Jilin Vocational and Technical College, Longjing, Jilin Province 133400, China
| | - Ji-Xing Nan
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Yan-Ling Wu
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| |
Collapse
|
4
|
Sun Y, Xu C, Luo J, Li S, Chen S, Cen Y, Xu P. Comprehensive analysis of differential long non-coding RNA and messenger RNA expression in cholelithiasis using high-throughput sequencing and bioinformatics. Front Genet 2024; 15:1375019. [PMID: 38808330 PMCID: PMC11130440 DOI: 10.3389/fgene.2024.1375019] [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: 01/23/2024] [Accepted: 04/24/2024] [Indexed: 05/30/2024] Open
Abstract
Background The etiology of gallstone disease (GSD) has not been fully elucidated. Consequently, the primary objective of this study was to scrutinize and provisionally authenticate the distinctive expression profiles of long non-coding RNAs (lncRNAs) and messenger RNAs (mRNAs) in GSD. Methods RiboNucleic Acid (RNA) sequencing was used on four paired human gallbladder samples for the purpose of this study. Differentially expressed lncRNAs (DElncRNAs) and mRNAs (DEmRNAs) were identified and subjected to analysis of their biological functions. The Pearson's correlation coefficients between DElncRNAs and DEmRNAs were computed to construct a co-expression network delineating their associations. Furthermore, both cis- and trans-regulatory networks of selected lncRNAs were established and visualized. Additionally, a competing endogenous RNA (ceRNA) regulatory network was constructed. To validate the RNA-sequencing data, we performed a Quantitative Real-time Polymerase Chain Reaction (RT-qPCR) on 10 paired human gallbladder samples, assessing the expressions of the top 4 DEmRNAs and DElncRNAs in gallstone and control samples. Results A total of 934 DEmRNAs and 304DElncRNAs were successfully identified. Functional enrichment analysis indicated a predominant involvement in metabolic-related biological functions. Correlation analysis revealed a strong association between the expressions of 597 DEmRNAs and 194 DElncRNAs. Subsequently, both a cis-lncRNA-mRNA and a trans-lncRNA-Transcription Factor (TF)-mRNA regulatory network were meticulously constructed. Additionally, a ceRNA network, comprising of 24 DElncRNAs, 201 DEmRNAs, and 120 predicted miRNAs, was established. Furthermore, using RT-qPCR, we observed significant upregulation of AC004692.4, HECW1-IT1, SFRP4, and COMP, while LINC01564, SLC26A3, RP1-27K12.2, and GSTA2 exhibited marked downregulation in gallstone samples. Importantly, these findings were consistent with the sequencing. Conclusion We conducted a screening process to identify DElncRNAs and DEmRNAs in GSD. This approach contributes to a deeper understanding of the genetic factors involved in the etiology of gallstones.
Collapse
Affiliation(s)
- Yanbo Sun
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Conghui Xu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- School of Medicine, Yunnan University, Kunming, China
| | - Jing Luo
- Department of Gastrointestinal Surgery, Qujing No. 1 People’s Hospital, Qujing, Yunnan, China
| | - Shumin Li
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shi Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yunyun Cen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Pengyuan Xu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
5
|
Sun Y, Zhang L, Jiang Z. The role of peroxisome proliferator-activated receptors in the regulation of bile acid metabolism. Basic Clin Pharmacol Toxicol 2024; 134:315-324. [PMID: 38048777 DOI: 10.1111/bcpt.13971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 12/06/2023]
Abstract
Bile acids are synthesized from cholesterol in the liver. Dysregulation of bile acid homeostasis, characterized by excessive accumulation in the liver, gallbladder and blood, can lead to hepatocellular damage and the development of cholestatic liver disease. Nuclear receptors play a crucial role in the control of bile acid metabolism by efficiently regulating bile acid synthesis and transport in the liver. Among these receptors, peroxisome proliferator-activated receptor (PPAR), a ligand-activated transcription factor belonging to the nuclear hormone receptor superfamily, controls the expression of genes involved in adipogenesis, lipid metabolism, inflammation and glucose homeostasis and has emerged as a potential therapeutic target for the treatment of the metabolic syndrome in the past two decades. Emerging evidence suggests that PPAR activation holds promise as a therapeutic target for cholestatic liver disease, as it affects both bile acid production and transport. This review provides a comprehensive overview of recent advances in elucidating the role of PPAR in the regulation of bile acid metabolism, highlighting the current position of PPAR agonists in the treatment of primary biliary cholangitis. By summarizing the specific regulatory effects of PPAR on bile acids, this review contributes to the exploration of novel therapeutic strategies for cholestatic liver diseases.
Collapse
Affiliation(s)
- Yuqing Sun
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Pharmaceutical Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Luyong Zhang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Pharmaceutical Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhenzhou Jiang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Pharmaceutical Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
6
|
Wang W, Zhang K, Liu B, Zhou T, Tang Y, Li Y. Chaihu Shugan prevents cholesterol gallstone formation by ameliorating the microbiota dysbiosis and metabolic disturbance in mice. Front Pharmacol 2024; 14:1291236. [PMID: 38357365 PMCID: PMC10866373 DOI: 10.3389/fphar.2023.1291236] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/06/2023] [Indexed: 02/16/2024] Open
Abstract
Introduction: Cholesterol gallstone (CGS) is a biliary tract disorder requiring treatment in approximately 20% of patients. The efficacy of Chaihu Shugan in preventing CGS recurrence after successful treatment remains uncertain. Methods: We examined the in vivo preventive efficacy of Chaihu Shugan using a CGS mouse model and used multi-omics to study the interplay between gut microbiota, metabolism, and gene expression. Results: The intestinal microbiota was severely dysregulated during the formation of CGS, showing a marked decrease in the abundance of beneficial microbiota, especially Lactobacillus and Akkermansia. Chaihu Shugan prevented CGS formation by restoring the composition of the gut microbiota and reversing the metabolic disturbances caused by dysbiosis. This preventive effect of Chaihu Shugan was paralleled by changes in the expression of metabolism-related genes in the liver. A network pharmacology analysis of Chaihu Shugan revealed that obacunone may be the key active metabolite in regulating bile acid metabolism. Multi-omics and correlation analyses elucidated the interplay between gut microbiota, metabolism, and gene alterations in the dose-dependent effect of Chaihu Shugan. Conclusion: Our data show that Chaihu Shugan can prevent CGS and indicate its mechanisms of action.
Collapse
Affiliation(s)
- Wei Wang
- Department of Intervention, The Second Hospital of Shandong University, Jinan, China
| | - Kun Zhang
- Shanghai Biotree Biotech Co., Ltd, Shanghai, China
| | | | - Tong Zhou
- Department of Intervention, The Second Hospital of Shandong University, Jinan, China
| | - Yu Tang
- Department of Geriatrics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuliang Li
- Department of Intervention, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
7
|
Liu HY, Li M, Li Q. De novo mutation loci and clinical analysis in a child with sodium taurocholate cotransport polypeptide deficiency: A case report. World J Clin Cases 2021; 9:11487-11494. [PMID: 35071582 PMCID: PMC8717514 DOI: 10.12998/wjcc.v9.i36.11487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/10/2021] [Accepted: 11/18/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Sodium taurocholate cotransport polypeptide (NTCP) deficiency disease is a genetic metabolic disorder due to mutations in the SLC10A1 gene and impaired bile acid salt uptake by the basolateral membrane transport protein NTCP in hepatocytes. A variety of clinical manifestations and genetic mutation loci have been reported for this disease. However, specific therapeutic measures are lacking, and the long-term effects are unknown.
CASE SUMMARY An infant with elevated bile acids and behavioral neurodevelopmental delay failed to respond to bile acid-lowering therapy. Genetic testing for metabolic liver disease revealed that the child had NTCP deficiency due to the SLC10A1 mutation: c.422dupA (p.Y141X), which is a novel mutation site. The current follow-up revealed a gradual decrease in bile acid levels after 1 year of age, but the child still had behavioral neurodevelopmental delays.
CONCLUSION The clinical manifestations, genetic characteristics, treatment and long-term prognosis due to NTCP deficiency remain poorly defined and need to be further confirmed by more studies and reports.
Collapse
Affiliation(s)
- Hui-Yan Liu
- Department of Pediatrics, the First Affiliated Hospital of Kunming Medical University, Kunming 650000, Yunnan Province, China
| | - Meng Li
- Department of Pediatrics, the First Affiliated Hospital of Kunming Medical University, Kunming 650000, Yunnan Province, China
| | - Qi Li
- Department of Pediatrics, the First Affiliated Hospital of Kunming Medical University, Kunming 650000, Yunnan Province, China
| |
Collapse
|
8
|
Fan X, Liu B, Zhou J, Gu X, Zhou Y, Yang Y, Guo F, Wei X, Wang H, Si N, Yang J, Bian B, Zhao H. High-Fat Diet Alleviates Neuroinflammation and Metabolic Disorders of APP/PS1 Mice and the Intervention With Chinese Medicine. Front Aging Neurosci 2021; 13:658376. [PMID: 34168550 PMCID: PMC8217439 DOI: 10.3389/fnagi.2021.658376] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/06/2021] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease caused by the complex interaction of multiple mechanisms. Recent studies examining the effect of high-fat diet (HFD) on the AD phenotype have demonstrated a significant influence on both inflammation and cognition. However, different studies on the effect of high-fat diet on AD pathology have reported conflicting conclusions. To explore the involvement of HFD in AD, we investigated phenotypic and metabolic changes in an AD mouse model in response to HFD. The results indicated there was no significant effect on Aβ levels or contextual memory due to HFD treatment. Of note, HFD did moderate neuroinflammation, despite spurring inflammation and increasing cholesterol levels in the periphery. In addition, diet affected gut microbiota symbiosis, altering the production of bacterial metabolites. HFD created a favorable microenvironment for bile acid alteration and arachidonic acid metabolism in APP/PS1 mice, which may be related to the observed improvement in LXR/PPAR expression. Our previous research demonstrated that Huanglian Jiedu decoction (HLJDD) significantly ameliorated impaired learning and memory. Furthermore, HLJDD may globally suppress inflammation and lipid accumulation to relieve cognitive impairment after HFD intervention. It was difficult to define the effect of HFD on AD progression because the results were influenced by confounding factors and biases. Although there was still obvious damage in AD mice treated with HFD, there was no deterioration and there was even a slight remission of neuroinflammation. Moreover, HLJDD represents a potential AD drug based on its anti-inflammatory and lipid-lowering effects.
Collapse
Affiliation(s)
- Xiaorui Fan
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China.,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bin Liu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Junyi Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinru Gu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanyan Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yifei Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Feifei Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaolu Wei
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongjie Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Nan Si
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jian Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baolin Bian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haiyu Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Han T, Lv Y, Wang S, Hu T, Hong H, Fu Z. Pioglitazone prevents cholesterol gallstone formation through the regulation of cholesterol homeostasis in guinea pigs with a lithogenic diet. Lipids Health Dis 2019; 18:218. [PMID: 31829191 PMCID: PMC6907187 DOI: 10.1186/s12944-019-1159-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/29/2019] [Indexed: 02/07/2023] Open
Abstract
Background The cholesterol gallstones diseases (CGD) is highly correlated with metabolic syndrome and type 2 diabetes. The present study aimed to investigate preventive effects of pioglitazone (PIO), an antidiabetic drug, on the CGD in guinea pigs fed with a lithogenic diet (LD). Methods The guinea pigs were fed with the LD for 8 weeks. All guinea pigs were grouped as follows: low fat diet; LD; LD plus PIO (4 mg/kg); LD plus PIO (8 mg/kg); LD plus ezetimibe (EZE) (2 mg/kg). Gallbladder stones were observed using microscopy. The profile of biliary composition, and blood glucose, insulin and lipid were analyzed. The liver or ileum was harvested for determinations of hydroxyl-methyl-glutaryl-CoA reductase (HMGCR), sterol regulatory element-binding proteins 2 (SREBP2), 7α-hydroxylase (CYP7A1), adenosine triphosphate-binding cassette (ABC) sterol transporters G5 and G8 (ABCG5, ABCG8), bile salt export pump (BSEP), Niemann-Pick C1-Like 1 (NPC1L1) and acetyl-coenzyme A cholesterol acyltransferase (ACAT2) by Western blot. The gallbladders were used for histological examination. Results The LD successfully induced gallstone. Both pioglitazone and ezetimibe prevented gallstone formation, as well as hepatic and cholecystic damages. Pioglitazone significantly decreased HMGCR and SREBP2, but increased CYP7A1, ABCG5, ABCG8, and BSEP in the liver. Pioglitazone also remarkably decreased NPC1L1 and ACAT2, while increased ABCG5/8 in the intestine. The beneficial alterations of cholesterol and bile acids in the bile, as well as profile of glucose, insulin and lipid in the blood were found in the guinea pigs treated with pioglitazone. Conclusion Pioglitazone has a noticeable benefit towards the CGD, which is involved in changes of synthesis, transformation, absorption, and transportation of cholesterol.
Collapse
Affiliation(s)
- Tao Han
- Department of Intensive Care Unit, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Yangge Lv
- Department of Pharmacology, China Pharmaceutical University, Nanjing, 210009, China
| | - Shijia Wang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Tao Hu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Hao Hong
- Department of Pharmacology, China Pharmaceutical University, Nanjing, 210009, China
| | - Zan Fu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China.
| |
Collapse
|