1
|
Zhao J, Shen Q, Yong X, Li X, Tian X, Sun S, Xu Z, Zhang X, Zhang L, Yang H, Shao Z, Xu H, Jiang Y, Zhang Y, Yan W. Cryo-EM reveals cholesterol binding in the lysosomal GPCR-like protein LYCHOS. Nat Struct Mol Biol 2025:10.1038/s41594-024-01470-9. [PMID: 39824976 DOI: 10.1038/s41594-024-01470-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 12/06/2024] [Indexed: 01/20/2025]
Abstract
Cholesterol plays a pivotal role in modulating the activity of mechanistic target of rapamycin complex 1 (mTOR1), thereby regulating cell growth and metabolic homeostasis. LYCHOS, a lysosome-localized G-protein-coupled receptor-like protein, emerges as a cholesterol sensor and is capable of transducing the cholesterol signal to affect the mTORC1 function. However, the precise mechanism by which LYCHOS recognizes cholesterol remains unknown. Here, using cryo-electron microscopy, we determined the three-dimensional structural architecture of LYCHOS in complex with cholesterol molecules, revealing a unique arrangement of two sequential structural domains. Through a comprehensive analysis of this structure, we elucidated the specific structural features of these two domains and their collaborative role in the process of cholesterol recognition by LYCHOS.
Collapse
Affiliation(s)
- Jie Zhao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Qingya Shen
- Department of Pathology of Sir Run Shaw Hospital, Department of Pharmacology, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, China
| | - Xihao Yong
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Xin Li
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, Metabolomics and Proteomics Technology Platform, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaowen Tian
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Suyue Sun
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Zheng Xu
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Xiaoyu Zhang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Lu Zhang
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, Metabolomics and Proteomics Technology Platform, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Yang
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, Metabolomics and Proteomics Technology Platform, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China.
| | - Haoxing Xu
- New Cornerstone Science Laboratory & Liangzhu Laboratory, the Second Affiliated Hospital & School of Basic Medical Sciences, Zhejiang University, Hangzhou, China.
| | - Yiyang Jiang
- Department of Hepatobiliary Surgery, Innovative Institute of Tumor Immunity and Medicine (ITIM), Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Yan Zhang
- Department of Pathology of Sir Run Shaw Hospital, Department of Pharmacology, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
- MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, China.
| | - Wei Yan
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
2
|
May CL, Ducheix S, Cariou B, Rimbert A. From Genetic Findings to new Intestinal Molecular Targets in Lipid Metabolism. Curr Atheroscler Rep 2025; 27:26. [PMID: 39798054 DOI: 10.1007/s11883-024-01264-w] [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] [Accepted: 12/03/2024] [Indexed: 01/13/2025]
Abstract
PURPOSE OF REVIEW While lipid-lowering therapies demonstrate efficacy, many patients still contend with significant residual risk of atherosclerotic cardiovascular diseases (ASCVD). The intestine plays a pivotal role in regulating circulating lipoproteins levels, thereby exerting influence on ASCVD pathogenesis. This review underscores recent genetic findings from the last six years that delineate new biological pathways and actors in the intestine which regulate lipid-related ASCVD risk. RECENT FINDINGS Specifically, we detail the role of LIMA1 in cholesterol absorption within enterocytes, the function of PLA2G12B in the expansion and lipidation of chylomicrons, the involvement of SURF4 in lipoprotein secretion, and the discovery of a gut-derived hormone named CHOLESIN that modulates cholesterol homeostasis through GPR146 via a gut-liver crosstalk. We further discuss the potential of these newly identified genes and pathways as novel targets for pharmaceutical intervention. Newly identified genetic and intestinal molecular mechanisms offer promising opportunities for preventing and treating ASCVD, but careful evaluation and further research are needed to optimize their clinical application.
Collapse
Affiliation(s)
- Cédric Le May
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000, Nantes, France
| | - Simon Ducheix
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000, Nantes, France
| | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000, Nantes, France
| | - Antoine Rimbert
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thorax, F-44000, Nantes, France.
| |
Collapse
|
3
|
Zheng L, Ye ZY, Ma JJ. Effect of cholesterol metabolism on hepatolithiasis. World J Gastroenterol 2025; 31:99960. [PMID: 39777239 PMCID: PMC11684189 DOI: 10.3748/wjg.v31.i1.99960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/03/2024] [Accepted: 11/05/2024] [Indexed: 12/09/2024] Open
Abstract
Surgical intervention is currently the primary treatment for hepatolithiasis; however, some patients still experience residual stones and high recurrence rates after surgery. Cholesterol metabolism seems to play an important role in hepatolithiasis pathogenesis. A high cholesterol diet is one of the significant reasons for the increasing incidence of hepatolithiasis. Therefore, regular diet and appropriate medical intervention are crucial measures to prevent hepatolithiasis and reduce recurrence rate after surgery. Reducing dietary cholesterol and drugs that increase cholesterol stone solubility are key therapeutic approaches in treating hepatolithiasis. This article discusses the cholesterol metabolic pathways related to the pathogenesis of hepatolithiasis, as well as food intake and targeted therapeutic drugs.
Collapse
Affiliation(s)
- Lin Zheng
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, No. 215 Heping West Road, Shijiazhuang 050000, Hebei Province, China
| | - Zi-Yu Ye
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, No. 215 Heping West Road, Shijiazhuang 050000, Hebei Province, China
| | - Jun-Ji Ma
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, No. 215 Heping West Road, Shijiazhuang 050000, Hebei Province, China
| |
Collapse
|
4
|
Xiao MY, Li S, Pei WJ, Gu YL, Piao XL. Natural Saponins on Cholesterol-Related Diseases: Treatment and Mechanism. Phytother Res 2025. [PMID: 39754504 DOI: 10.1002/ptr.8432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/27/2024] [Accepted: 12/14/2024] [Indexed: 01/06/2025]
Abstract
Saponins are compounds composed of lipophilic aglycones linked to hydrophilic sugars. Natural saponins are isolated from plants and some Marine organisms. As important cholesterol-lowering drugs, natural saponins have attracted wide attention for their therapeutic potential in a variety of cholesterol-related metabolic diseases. To review the effects of natural saponins on cholesterol-related metabolic diseases, and to deepen the understanding of the cholesterol-lowering mechanism of saponins. The literature related to saponins and cholesterol-lowering diseases was collected using keywords "saponins" and "cholesterol" from PubMed, Web of Science, and Google Scholar from January 2000 to May 2024. The total number of articles related to saponins and cholesterol-lowering diseases was 240 after excluding irrelevant articles. Natural saponins can regulate cholesterol to prevent and treat a variety of diseases, such as atherosclerosis, diabetes, liver disease, hyperlipidemia, cancer, and obesity. Mechanistically, natural saponins regulate cholesterol synthesis and uptake through the AMPK/SREBP2/3-hydroxy-3-methyl-glutaryl coenzyme A reductase pathway and PCSK9/LDLR pathway, and regulate cholesterol efflux and esterification targeting Liver X receptor/ABC pathway and ACAT family. Natural saponins have broad application prospects in regulating cholesterol metabolism, for the development of more cholesterol-lowering drugs provides a new train of thought. However, it is still necessary to further explore the molecular mechanism and expand clinical trials to provide more evidence.
Collapse
Affiliation(s)
- Man-Yu Xiao
- School of Pharmacy, Minzu University of China, Beijing, China
| | - Si Li
- School of Pharmacy, Minzu University of China, Beijing, China
| | - Wen-Jing Pei
- School of Pharmacy, Minzu University of China, Beijing, China
| | - Yu-Long Gu
- School of Pharmacy, Minzu University of China, Beijing, China
| | - Xiang-Lan Piao
- School of Pharmacy, Minzu University of China, Beijing, China
| |
Collapse
|
5
|
Xu G, Chu M, Shen S, Miao H, Bai Y, Liu X, Liu W, Song P, Wang L, Fu M, Dang E, Shao S, Wang G. Causal effects of circulating lipids and lipid-lowering drugs on the risk of atopic dermatitis: a mendelian randomization study. Arch Dermatol Res 2024; 317:154. [PMID: 39718588 DOI: 10.1007/s00403-024-03635-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/31/2024] [Accepted: 11/26/2024] [Indexed: 12/25/2024]
Abstract
Lipid metabolism disorders are frequently noted in atopic dermatitis (AD) patients, prompting the long-term use of lipid-lowering drugs. However, the causal effects of circulating lipids and different lipid-lowering drugs on the risk of AD are not thoroughly understood. Using publicly available genome-wide association studies (GWAS) summary data from two different cohorts, a series of Mendelian randomization (MR) analyses were conducted to explore the causal effects of genetically proxied circulating lipids and lipid-lowering drugs on the risk of AD. Statistically, the random-effects inverse-variance-weighted (IVW) model was used as main analysis and several methods were conducted for sensitivity analysis to test the robustness of our results. Our findings revealed reduced risks of AD related to genetically proxied subtilisin/kexin type 9 (PCSK9) inhibition and lipoprotein lipase (LPL) agonist, while an increased AD risk associated with Niemann-Pick C1-like 1 (NPC1L1) inhibition. Circulating lipids and other drug targets did not show significant associations with AD risk. These results were replicated in the validation cohort; sensitivity analyses confirmed the robustness. This MR study suggests that, independent of circulating lipids, the use of PCSK9 inhibitors and LPL agonists may be associated with a decreased risk of AD, while inhibition of NPC1L1 is implicated in an increased risk. These findings may help optimize personalized selection of lipid-lowering drugs for AD patients and those at risk of AD.
Collapse
Affiliation(s)
- Guangquan Xu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Mengyang Chu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Shengxian Shen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Haijun Miao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Yaxing Bai
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Xuan Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Wanting Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Pu Song
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Lei Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Meng Fu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Erle Dang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Shuai Shao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China.
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China.
| |
Collapse
|
6
|
Zhu H, Chen L, Chen ZX, Lin H, Liu J, Kwek E, Ma KY, He W, Wang G, Chen ZY. Synthesis of cholesterol analogues and comparison on their effect on plasma cholesterol with β-Sitosterol. Food Chem 2024; 461:140820. [PMID: 39153376 DOI: 10.1016/j.foodchem.2024.140820] [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: 10/19/2023] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
The application of plant sterols in the treatment of hypercholesterolemia is promising. We hypothesize that plant sterols can reduce blood cholesterol because they have a side chain of at least three branches. Three cholesterol analogues were synthesized: CA0 (no side chain), CA3 (a 3‑carbon chain with one branch), and CA14 (a 14‑carbon side chain with two branches), and then compared their effect on blood cholesterol with that of β-sitosterol. Structurally, β-sitosterol has a 10‑carbon side chain with three branches. Results demonstrated that β-sitosterol could effectively reduce plasma total cholesterol (TC) by 20.3%, whereas CA0, CA3 and CA14 did not affect plasma TC in hypercholesterolemia hamsters. β-Sitosterol was absent in the plasma and liver, indicating it was not absorbed. We concluded that β-sitosterol with three branches had plasma TC-lowering activity. In contrast, cholesterol analogues with a side chain of two or fewer branches did not affect plasma cholesterol.
Collapse
Affiliation(s)
- Hanyue Zhu
- School of Food Science and Engineering / Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan, Guangdong, China; School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Longjian Chen
- Shenzhen Bay Laboratory Pingshan Translational Medicine Center, Shenzhen, China
| | - Zi-Xing Chen
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Huiqi Lin
- School of Food Science and Engineering / Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan, Guangdong, China
| | - Jianhui Liu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Erika Kwek
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Ka Ying Ma
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Wensen He
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | | | - Zhen-Yu Chen
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China.
| |
Collapse
|
7
|
Kim JJ, Yang EJ, Molina David J, Cho S, Ficarella M, Pape N, Schiffer JE, Njeim R, Kim SS, Lo Re C, Fontanella A, Kaber M, Sloan A, Merscher S, Fornoni A. Ezetimibe Enhances Lipid Droplet and Mitochondria Contact Formation, Improving Fatty Acid Transfer and Reducing Lipotoxicity in Alport Syndrome Podocytes. Int J Mol Sci 2024; 25:13134. [PMID: 39684843 DOI: 10.3390/ijms252313134] [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: 10/29/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Mitochondrial dysfunction is a critical factor in the pathogenesis of Alport syndrome (AS), contributing to podocyte injury and disease progression. Ezetimibe, a lipid-lowering drug, is known to inhibit cholesterol and fatty acid uptake and to reduce triglyceride content in the kidney cortex of mice with AS. However, its effects on lipid droplet (LD) utilization by mitochondria have not been explored. Transmission electron microscopy (TEM) and mitochondrial functional assays (ATP production, mitochondrial membrane potential, and citrate synthase activity) were used to investigate the impact of ezetimibe on LD-mitochondria contact formation and mitochondrial function in Col4a3KO (AS) and wildtype (WT) podocytes. TEM analysis revealed significant mitochondrial abnormalities in AS podocytes, including swollen mitochondria and reduced cristae density, while mitochondrial function assays showed decreased ATP production and lowered mitochondrial membrane potential. AS podocytes also demonstrated a higher content of LD but with reduced LD-mitochondria contact sites. Ezetimibe treatment significantly increased the number of LD-mitochondria contact sites, enhanced fatty acid transfer efficiency, and reduced intracellular lipid accumulation. These changes were associated with a marked reduction in the markers of lipotoxicity, such as apoptosis and oxidative stress. Mitochondrial function was significantly improved, evidenced by increased basal respiration, ATP production, maximal respiration capacity, and the restoration of mitochondrial membrane potential. Additionally, mitochondrial swelling was significantly reduced in ezetimibe-treated AS podocytes. Our findings reveal a novel role for ezetimibe in enhancing LD-mitochondria contact formation, leading to more efficient fatty acid transfer, reduced lipotoxicity, and improved mitochondrial function in AS podocytes. These results suggest that ezetimibe could be a promising therapeutic agent for treating mitochondrial dysfunction and lipid metabolism abnormalities in AS.
Collapse
Affiliation(s)
- Jin-Ju Kim
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Eun-Jeong Yang
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Judith Molina David
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Sunjoo Cho
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Maria Ficarella
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Nils Pape
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Josephin Elizabeth Schiffer
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Rachel Njeim
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Stephanie S Kim
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Claudia Lo Re
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U "G. Martino", University of Messina, 98122 Messina, Italy
| | - Antonio Fontanella
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Maria Kaber
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alexis Sloan
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
8
|
Awazu T, Sakamoto K, Minagi Y, Ohnishi M, Bito T, Matsunaga Y, Iwasaki T, Kawano T. The small GTPase RAB-18 is involved in regulating development/diapause by recruiting the intestinal cholesterol transporter NCR-1 onto the apical side in Caenorhabditis elegans. Biochem Biophys Res Commun 2024; 734:150609. [PMID: 39232459 DOI: 10.1016/j.bbrc.2024.150609] [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: 05/07/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024]
Abstract
RAB family proteins, which are small GTPases, are integral to the process of eukaryotic membrane trafficking. In the nematode, Caenorhabditis elegans, 31 RAB proteins have been identified through genome sequencing. Using an RNAi screen specifically targeting C. elegans rab genes, we identified multiple genes that are involved in the regulation of larval development, in particular, the rab-18 gene. Our molecular genetic studies resulted in several findings. First, RAB-18 predominantly functions in the intestine to regulate larval development by modulating steroid hormone signaling. Second, the C. elegans cholesterol transporter NCR-1 is a target of RAB-18 in the intestine. Third, the membrane trafficking of NCR-1 to the apical side in intestinal cells is particularly influenced by RAB-18. Finally, RAB-18 and NCR-1 possibly co-localize on membrane vesicles. Our study is the first to demonstrate the relationship between a RAB protein and a cholesterol transporter, in which the RAB protein probably drives the transporter to the apical membrane in the intestine to regulate cholesterol uptake. This study provides insight into the molecular mechanisms underlying human disease stemming from a transport defect of cholesterol and its derivative.
Collapse
Affiliation(s)
- Toshikuni Awazu
- Department of Bioscience, Biotechnology, and Agrochemistry, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Kanato Sakamoto
- Department of Agricultural Science, Graduate School of Sustainability Science, Tottori University, Tottori, Japan
| | - Yuka Minagi
- Department of Agricultural Science, Graduate School of Sustainability Science, Tottori University, Tottori, Japan
| | - Masumi Ohnishi
- Department of Agricultural Science, Graduate School of Sustainability Science, Tottori University, Tottori, Japan
| | - Tomohiro Bito
- Department of Bioscience, Biotechnology, and Agrochemistry, Faculty of Agriculture, Tottori University, Tottori, Japan; Department of Agricultural Science, Graduate School of Sustainability Science, Tottori University, Tottori, Japan
| | | | - Takashi Iwasaki
- Department of Bioscience, Biotechnology, and Agrochemistry, Faculty of Agriculture, Tottori University, Tottori, Japan; Department of Agricultural Science, Graduate School of Sustainability Science, Tottori University, Tottori, Japan
| | - Tsuyoshi Kawano
- Department of Bioscience, Biotechnology, and Agrochemistry, Faculty of Agriculture, Tottori University, Tottori, Japan; Department of Agricultural Science, Graduate School of Sustainability Science, Tottori University, Tottori, Japan.
| |
Collapse
|
9
|
Yamanashi Y, Komine T, Hirota Y, Suzuki H, Osuga Y, Takada T. Dietary phytosterols induce infertility in female mice via epigenomic modulations. Commun Biol 2024; 7:1535. [PMID: 39562830 DOI: 10.1038/s42003-024-07233-y] [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: 05/20/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
Dietary modifications to overcome infertility have attracted attention; however, scientifically substantiated information on specific dietary components affecting fertility and their mechanisms is limited. Herein, we investigated diet-induced, reversible infertility in female mice lacking the heterodimer of ATP-binding cassette transporters G5 and G8 (ABCG5/G8), which functions as a lipid exporter in the intestine. We found that dietary phytosterols, especially β-sitosterol and brassicasterol, which are substrates of ABCG5/G8, have potent but reversible reproductive toxicities in mice. Mechanistically, these phytosterols inhibited ovarian folliculogenesis and reduced egg quality by enhancing polycomb repressive complex 2-mediated histone H3 trimethylation at lysine 27 in the ovary. Clinical analyses showed that serum phytosterol levels were significantly and negatively correlated with the blastocyst development rate of fertilized eggs in women undergoing in vitro fertilization, suggesting that phytosterols affect egg quality in both humans and mice. Thus, avoiding excessive intake of certain phytosterols would be beneficial for female reproductive health.
Collapse
Affiliation(s)
- Yoshihide Yamanashi
- Department of Pharmacy, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan.
| | - Toko Komine
- Department of Pharmacy, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Suzuki
- Department of Pharmacy, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
10
|
Dang EV, Reboldi A. Cholesterol sensing and metabolic adaptation in tissue immunity. Trends Immunol 2024; 45:861-870. [PMID: 39424470 PMCID: PMC11560508 DOI: 10.1016/j.it.2024.09.013] [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: 08/23/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/21/2024]
Abstract
Cholesterol metabolites, particularly oxidized forms known as oxysterols, play crucial roles in modulating immune and metabolic processes across various tissues. Concentrations of local cholesterol and its metabolites influence tissue-specific immune responses by shaping the metabolic and spatial organization of immune cells in barrier organs like the small intestine (SI) and lungs. We explore recent molecular and cellular evidence supporting the metabolic adaptation of innate and adaptive immune cells in the SI and lung, driven by cholesterol and cholesterol metabolites. Further research should unravel the detailed molecular mechanisms and spatiotemporal adaptations involving cholesterol metabolites in distinct mucosal tissues in homeostasis or infection. We posit that pharmacological interventions targeting the generation or sensing of cholesterol metabolites might be leveraged to enhance long-term immune protection in mucosal tissues or prevent autoinflammatory states.
Collapse
Affiliation(s)
- Eric V Dang
- Molecular Mycology and Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Andrea Reboldi
- Department of Pathology, Immunology, and Microbial Pathogenesis Program, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
11
|
de Dios-Blázquez L, Cano-Argüelles AL, Pérez-Sánchez R, González-Sánchez M, Oleaga A. First data on cholesterol metabolism in Ornithodoros argasid ticks: Molecular and functional characterization of the N-terminal domain of Niemann-Pick C1 proteins. Ticks Tick Borne Dis 2024; 15:102382. [PMID: 39032307 DOI: 10.1016/j.ttbdis.2024.102382] [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: 04/29/2024] [Revised: 06/21/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
Cholesterol is a molecule vital for tick physiology, but ticks cannot synthesize it and rely on dietary cholesterol. Therefore, tick proteins involved in cholesterol absorption and transport, such as the Niemann-Pick type C1 domain-containing (NPC1) proteins, are promising targets for anti-tick vaccine development. The aim of this study was to assess the structure, function, and protective efficacy of the NPC1 orthologues identified previously in the midgut transcriptomes of argasid ticks Ornithodoros erraticus and Ornithodoros moubata. For this purpose, their corresponding cDNA coding sequences were cloned and sequenced, their secondary and 3D structures were predicted, and their function was evaluated through RNAi-mediated gene knockdown and in vitro feeding on blood supplemented with ezetimibe, which inhibits cholesterol binding by NPC1 proteins. Subsequently, the protective efficacy of a recombinant form of NPC1 from O. moubata (rOmNPC1) was tested in a rabbit vaccine trial. While inhibiting cholesterol absorption with ezetimibe resulted in up to 77 % mortality in adult O. moubata, NPC1 gene knockdown and vaccination with rOmNPC1 decreased female reproductive performance in terms of the number and fertility of laid eggs. This study presents the initial molecular and functional insights into NPC1 proteins in soft ticks and supports the hypothesis that disrupting cholesterol metabolism diminishes tick viability and reproduction, rendering Niemann-Pick type C1 domain-containing proteins promising targets for drugs or vaccines.
Collapse
Affiliation(s)
- Lucía de Dios-Blázquez
- Fundación para la Investigación Biomédica del Hospital Universitario la Paz -FIBHULP-, Paseo de la Castellana, 261, 28046, Madrid, Spain
| | - Ana Laura Cano-Argüelles
- Parasitology, Institute of Natural Resources and Agrobiology of Salamanca (IRNASA, CSIC), Cordel de Merinas, 40-52, 37008, Salamanca, Spain
| | - Ricardo Pérez-Sánchez
- Parasitology, Institute of Natural Resources and Agrobiology of Salamanca (IRNASA, CSIC), Cordel de Merinas, 40-52, 37008, Salamanca, Spain
| | - María González-Sánchez
- Parasitology, Institute of Natural Resources and Agrobiology of Salamanca (IRNASA, CSIC), Cordel de Merinas, 40-52, 37008, Salamanca, Spain
| | - Ana Oleaga
- Fundación para la Investigación Biomédica del Hospital Universitario la Paz -FIBHULP-, Paseo de la Castellana, 261, 28046, Madrid, Spain.
| |
Collapse
|
12
|
Song J, Fang Y, Rao X, Wu L, Zhang C, Ying J, Hua F, Lin Y, Wei G. Beyond conventional treatment: ASGR1 Leading the new era of hypercholesterolemia management. Biomed Pharmacother 2024; 180:117488. [PMID: 39316974 DOI: 10.1016/j.biopha.2024.117488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/01/2024] [Accepted: 09/20/2024] [Indexed: 09/26/2024] Open
Abstract
Cardiovascular disease (CVD) remains a leading cause of mortality worldwide, with hypercholesterolemia being a major risk factor. Although various lipid-lowering therapies exist, many patients fail to achieve optimal cholesterol control, highlighting the need for novel therapeutic approaches. ASGR1 (asialoglycoprotein receptor 1), predominantly expressed on hepatocytes, has emerged as a key regulator of cholesterol metabolism and low-density lipoprotein (LDL) clearance. This receptor's ability to regulate lipid homeostasis positions it as a promising target for therapeutic intervention in hypercholesterolemia and related cardiovascular diseases. This review critically examines the biological functions and regulatory mechanisms of ASGR1 in cholesterol metabolism, with a focus on its potential as a therapeutic target for hypercholesterolemia and related cardiovascular diseases. By analyzing recent advances in ASGR1 research, this article explores its role in liver-specific pathways, the implications of ASGR1 variants in CVD risk, and the prospects for developing ASGR1-targeted therapies. This review aims to provide a foundation for future research and clinical applications in hypercholesterolemia management.
Collapse
Affiliation(s)
- Jiali Song
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, Jiangxi 330006, PR China
| | - Yang Fang
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, Jiangxi 330006, PR China
| | - Xiuqin Rao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, Jiangxi 330006, PR China
| | - Luojia Wu
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, Jiangxi 330006, PR China
| | - Chenxi Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, Jiangxi 330006, PR China
| | - Jun Ying
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, Jiangxi 330006, PR China
| | - Fuzhou Hua
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, Jiangxi 330006, PR China
| | - Yue Lin
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, Jiangxi 330006, PR China.
| | - Gen Wei
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, PR China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, Jiangxi 330006, PR China.
| |
Collapse
|
13
|
Zhang J, Tian R, Liu J, Yuan J, Zhang S, Chi Z, Yu W, Yu Q, Wang Z, Chen S, Li M, Yang D, Hu T, Deng Q, Lu X, Yang Y, Zhou R, Zhang X, Liu W, Wang D. A two-front nutrient supply environment fuels small intestinal physiology through differential regulation of nutrient absorption and host defense. Cell 2024; 187:6251-6271.e20. [PMID: 39427662 DOI: 10.1016/j.cell.2024.08.012] [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: 09/08/2023] [Revised: 03/11/2024] [Accepted: 08/07/2024] [Indexed: 10/22/2024]
Abstract
The small intestine contains a two-front nutrient supply environment created by luminal dietary and microbial metabolites (enteral side) and systemic metabolites from the host (serosal side). Yet, it is unknown how each side contributes differentially to the small intestinal physiology. Here, we generated a comprehensive, high-resolution map of the small intestinal two-front nutrient supply environment. Using in vivo tracing of macronutrients and spatial metabolomics, we visualized the spatiotemporal dynamics and cell-type tropism in nutrient absorption and the region-specific metabolic heterogeneity within the villi. Specifically, glutamine from the enteral side fuels goblet cells to support mucus production, and the serosal side loosens the epithelial barrier by calibrating fungal metabolites. Disorganized feeding patterns, akin to the human lifestyle of skipping breakfast, increase the risk of metabolic diseases by inducing epithelial memory of lipid absorption. This study improves our understanding of how the small intestine is spatiotemporally regulated by its unique nutritional environment.
Collapse
Affiliation(s)
- Jian Zhang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311113, China
| | - Ruonan Tian
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jia Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Yuan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Siwen Zhang
- Department of Endocrinology & Metabolism, The First Hospital of Jilin University, Changchun 130061, China
| | - Zhexu Chi
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Weiwei Yu
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qianzhou Yu
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhen Wang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Sheng Chen
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Mobai Li
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Dehang Yang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Tianyi Hu
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qiqi Deng
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaoyang Lu
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yidong Yang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Rongbin Zhou
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Xue Zhang
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wanlu Liu
- Department of Rheumatology and Immunology of the Second Affiliated Hospital, and Centre of Biomedical Systems and Informatics of Zhejiang University, University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China; Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh EH8 9YL, UK.
| | - Di Wang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311113, China.
| |
Collapse
|
14
|
Jiao B, Wang B, Liu B, Zhao J, Zhang Y. Potential impact of ezetimibe on patients with NAFLD/NASH: a meta-analysis of randomized controlled trials. Front Endocrinol (Lausanne) 2024; 15:1468476. [PMID: 39439571 PMCID: PMC11493694 DOI: 10.3389/fendo.2024.1468476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/20/2024] [Indexed: 10/25/2024] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is now the most common cause of chronic liver disease. Studies have found that ezetimibe may be utilized as a supplemental treatment for NAFLD. Additionally, many clinical trials reported the potential impacts of ezetimibe on patients with NAFLD, although some conclusions remain controversial. Therefore, this study aimed to evaluate the effects of ezetimibe on patients with NAFLD. Method Online search was conducted across databases including PubMed, Embase, Scopus, Web of Science, Cochrane Library, Wanfang, VIP, and CNKI to retrieve all relevant controlled studies on the treatment of NAFLD with ezetimibe from the inception of the databases until April 2024. This meta-analysis comprised 10 randomized controlled trials (RCTs). Statistical analysis was conducted using the Meta package in R v4.3.2. Results A total of ten RCTs were included in this study, encompassing 578 patients (290 in the ezetimibe group and 288 in the control group) diagnosed with NAFLD/non-alcoholic steatohepatitis (NASH). The results indicated that ezetimibe significantly reduced levels of aspartate aminotransferase (P < 0.01), glutamyl transferase (γ-GT) (P < 0.01), total cholesterol (P < 0.01), low-density lipoprotein cholesterol (P < 0.01), high-sensitivity C-reactive protein (P < 0.01), and interleukin-6 (P < 0.01), and markedly increased levels of glycated hemoglobin (P = 0.02). Conclusions Ezetimibe may partially improve transaminase levels and positively impact liver function in patients with NAFLD/NASH. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42023461467.
Collapse
|
15
|
Alruhaimi RS, Hussein OE, Alnasser SM, Germoush MO, Alotaibi M, Hassanein EHM, El Mohtadi M, Mahmoud AM. Oxidative Stress, Inflammation, and Altered Lymphocyte E-NTPDase Are Implicated in Acute Dyslipidemia in Rats: Protective Role of Arbutin. Pharmaceuticals (Basel) 2024; 17:1343. [PMID: 39458984 PMCID: PMC11509952 DOI: 10.3390/ph17101343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Dyslipidemia is frequently linked to various disorders, and its clinical relevance is now recognized. The role of inflammation and oxidative stress (OS) in dyslipidemia has been acknowledged. This study assessed the potential of arbutin (ARB) to prevent dyslipidemia and its associated OS and inflammation in rats with acute hyperlipidemia. METHODS Rats received ARB orally for 14 days and a single intraperitoneal injection of poloxamer-407 on day 15. RESULTS Poloxamer-407 elevated circulating cholesterol (CHOL), triglycerides (TG), very low-density lipoprotein (vLDL), and LDL, and reduced high-density lipoprotein (HDL)-C and lipoprotein lipase (LPL). ARB ameliorated the circulating lipids and LPL, and suppressed 3-hydroxy-3-methylglutaryl CoA reductase (HMGCR) in rat liver and in vitro. Fatty acid synthase (FAS) in rat liver and its in vitro activity were suppressed by ARB, which also upregulated the LDL receptor (LDL-R) and ABCA1, and had no effect on ABCG5 and ABCG8 mRNA. ARB ameliorated liver malondialdehyde and nitric oxide and enhanced antioxidants in rats with dyslipidemia. Liver NF-κB p65 and blood inflammatory cytokines were increased in dyslipidemic rats, effects that were reversed by ARB. Moreover, ARB effectively suppressed lymphocyte E-NTPDase and E-ADA activities in dyslipidemic rats. The biochemical findings were supported by in silico data showing the affinity of ARB to bind LDL-R PCSK9 binding domain, HMGCR, FAS, and E-NTPDase. CONCLUSIONS ARB possessed anti-dyslipidemia, anti-inflammatory, and antioxidant effects mediated via the modulation of CHOL and TG synthesis, LPL, lymphocyte E-NTPDase and E-ADA, and cytokine release in rats. Thus, ARB could be an effective agent to attenuate dyslipidemia and its associated OS and inflammation, pending further studies as well as clinical trials.
Collapse
Affiliation(s)
- Reem S. Alruhaimi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Omnia E. Hussein
- Higher Technological Institute for Applied Health Sciences, Beni-Suef 62764, Egypt
| | - Sulaiman M. Alnasser
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
| | - Mousa O. Germoush
- Biology Department, College of Science, Jouf University, Sakakah 72388, Saudi Arabia
| | - Meshal Alotaibi
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Albatin, Hafar Al Batin 39524, Saudi Arabia
| | - Emad H. M. Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University-Assiut Branch, Assiut 71524, Egypt
| | | | - Ayman M. Mahmoud
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester M1 5GD, UK
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62514, Egypt
| |
Collapse
|
16
|
Sun S, Ma J, Zuo T, Shi J, Sun L, Meng C, Shu W, Yang Z, Yao H, Zhang Z. Inhibition of PCSK9: A Promising Enhancer for Anti-PD-1/PD-L1 Immunotherapy. RESEARCH (WASHINGTON, D.C.) 2024; 7:0488. [PMID: 39324018 PMCID: PMC11423609 DOI: 10.34133/research.0488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 09/27/2024]
Abstract
Immune checkpoint therapy, such as programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) blockade, has achieved remarkable results in treating various tumors. However, most cancer patients show a low response rate to PD-1/PD-L1 blockade, especially those with microsatellite stable/mismatch repair-proficient colorectal cancer subtypes, which indicates an urgent need for new approaches to augment the efficacy of PD-1/PD-L1 blockade. Cholesterol metabolism, which involves generating multifunctional metabolites and essential membrane components, is also instrumental in tumor development. In recent years, inhibiting proprotein convertase subtilisin/kexin type 9 (PCSK9), a serine proteinase that regulates cholesterol metabolism, has been demonstrated to be a method enhancing the antitumor effect of PD-1/PD-L1 blockade to some extent. Mechanistically, PCSK9 inhibition can maintain the recycling of major histocompatibility protein class I, promote low-density lipoprotein receptor-mediated T-cell receptor recycling and signaling, and modulate the tumor microenvironment (TME) by affecting the infiltration and exclusion of immune cells. These mechanisms increase the quantity and enhance the antineoplastic effect of cytotoxic T lymphocyte, the main functional immune cells involved in anti-PD-1/PD-L1 immunotherapy, in the TME. Therefore, combining PCSK9 inhibition therapy with anti-PD-1/PD-L1 immunotherapy may provide a novel option for improving antitumor effects and may constitute a promising research direction. This review concentrates on the relationship between PCSK9 and cholesterol metabolism, systematically discusses how PCSK9 inhibition potentiates PD-1/PD-L1 blockade for cancer treatment, and highlights the research directions in this field.
Collapse
Affiliation(s)
- Shengbo Sun
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jingxin Ma
- Department of Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tingting Zuo
- College of Biological Sciences and Technology, Yili Normal University, Yining, China
| | - Jinyao Shi
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Liting Sun
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Cong Meng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Wenlong Shu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Zhengyang Yang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Hongwei Yao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
17
|
Kühn J, Schutkowski A, Rayo-Abella LM, Kiourtzidis M, Nier A, Brandsch C, Stangl GI. Dietary cholesterol increases body levels of oral administered vitamin D 3 in mice. J Nutr Sci 2024; 13:e50. [PMID: 39345242 PMCID: PMC11428076 DOI: 10.1017/jns.2024.32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/13/2024] [Accepted: 06/06/2024] [Indexed: 10/01/2024] Open
Abstract
Vitamin D and cholesterol share the same intestinal transporters. Thus, it was hypothesized that dietary cholesterol adversely affects vitamin D uptake. The current studies investigated the influence of cholesterol on the availability of oral vitamin D. First, 42 wild-type mice received a diet with 25 µg/kg labelled vitamin D3 (vitamin D3-d3), supplemented with either 0% (control), 0.2%, 0.4%, 0.6%, 0.8%, 1.0% or 2.0% cholesterol for four weeks to investigate vitamin D uptake. In a second study, 10 wild-type mice received diets containing 0% (control) or 1% cholesterol over four weeks to determine cholesterol-induced changes in bile acids. Finally, we investigated the impact of cholesterol versus bile acids on vitamin D uptake in Caco-2 cells. Surprisingly, dietary cholesterol intake was associated with 40% higher serum levels of vitamin D3-d3 and 2.3-fold higher vitamin D3-d3 concentrations in the liver compared to controls. The second study showed that cholesterol intake resulted in higher concentrations of faecal bile acids (control: 3.55 ± 1.71 mg/g dry matter; 1% dietary cholesterol: 8.95 ± 3.69 mg/g dry matter; P < 0.05) and changes in the bile acid profile with lower contents of muricholic acids (P < 0.1) and higher contents of taurodeoxycholic acid (P < 0.01) compared to controls. In-vitro analyses revealed that taurocholic acid (P < 0.001) but not cholesterol increased the cellular uptake of vitamin D by Caco-2 cells. To conclude, dietary cholesterol seems to improve the bioavailability of oral vitamin D by stimulating the release of bile acids and increasing the hydrophobicity of bile.
Collapse
Affiliation(s)
- Julia Kühn
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany
| | - Alexandra Schutkowski
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany
| | - Lina-Maria Rayo-Abella
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany
| | - Mikis Kiourtzidis
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany
| | - Anika Nier
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany
| | - Corinna Brandsch
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany
| | - Gabriele I Stangl
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany
| |
Collapse
|
18
|
Bin C, Zhang C. The association between vitamin D consumption and gallstones in US adults: A cross-sectional study from the national health and nutrition examination survey. J Formos Med Assoc 2024:S0929-6646(24)00430-3. [PMID: 39261120 DOI: 10.1016/j.jfma.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 05/13/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Gallstone disease is common in the US and Europe. Gallstones are associated with factors such as age, sex, weight, and serum cholesterol levels. A complex relationship exists between vitamin D levels and cholesterol metabolism. However, the relationship between vitamin D level and gallstones remains unclear. This study aimed to investigate whether gallstones are associated with dietary vitamin D (D2+D3) consumption (VDC) in American adults. METHODS This cross-sectional study used data from people who participated in the National Health and Nutrition Examination Survey between March 2017 and March 2020. Multivariate logistic regression models were used to determine the association between vitamin D intake and the presence of gallstones. Stratified and interaction analyses were performed to determine whether the relationship was stable across different subgroups. RESULTS 6873 participants were included. VDC (per 1 SD) was positively associated with gallstones in the crude model (OR: 1.11, 95% Confidence Interval (CI): (1.05-1.17); p < 0.001), Further adjustment did not affect the results. When vitamin D was analyzed using quartiles, with increased quartile of VDC, the incidence of gallstones increased, and the OR of Q2 (OR: 1.08, 95% CI: 0.89-1.32, p = 0.436) and Q3 (OR: 1.55, 95% CI: 1.28-1.87, p < 0.001) was higher than that of Q1 in crude model. After adjusting for covariates, there is a positive association between VDC and incidence of gallstones without statistical significance. CONCLUSION VDC was positively associated with the incidence of gallstones, however, further studies are required to gather additional evidence.
Collapse
Affiliation(s)
- Chuxuan Bin
- Department of Gastroenterology, Beijing Tongren Hospital, Capital Medical University, Address: No. 1, Dongjiaomin Road, Dongcheng District, Beijing, China.
| | - Chuan Zhang
- Department of Gastroenterology, Beijing Tongren Hospital, Capital Medical University, Address: No. 1, Dongjiaomin Road, Dongcheng District, Beijing, China.
| |
Collapse
|
19
|
Aizawa H. Impact of micelle characteristics on cholesterol absorption and ezetimibe inhibition: Insights from Niemann-Pick C1-like 1 binding and molecular structure. J Liposome Res 2024; 34:386-398. [PMID: 37905576 DOI: 10.1080/08982104.2023.2274424] [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: 09/02/2023] [Accepted: 10/18/2023] [Indexed: 11/02/2023]
Abstract
Yamanashi et al., conducted a study on the absorption of cholesterol and β-sitosterol, as well as the inhibitory effect of ezetimibe (EZE). They used CaCo-2 cells to simulate the intestines and investigated how different mixed micelles, acting as carriers, were absorbed into these cells through the Niemann-Pick C1-like 1 (NPC1L1) protein. The study focused on the impact of micelle shape, size, and zeta potential on absorption and the inhibitory effect of EZE. I utilized small-angle X-ray scattering and a zeta potential measuring device to measure these characteristics. The findings revealed a two-step mechanism: NPC1L1 selectively bound micelles based on their shape and size, and once bound, the absorption was regulated by the molecular structure of the micelle components. EZE's inhibitory effect changed with micelle composition, influencing micelle size and shape. EZE initially acted on the micelle's shape and size, and then NPC1L1 selectively bound micelles based on their shape and size, allowing EZE to directly inhibit absorption by interacting with NPC1L1. This groundbreaking discovery challenges existing concepts and holds significant implications for researchers in drug development, as well as physicians and pharmacists.
Collapse
Affiliation(s)
- Hideki Aizawa
- Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Japan
| |
Collapse
|
20
|
Maddineni G, Obulareddy SJ, Paladiya RD, Korsapati RR, Jain S, Jeanty H, Vikash F, Tummala NC, Shetty S, Ghazalgoo A, Mahapatro A, Polana V, Patel D. The role of gut microbiota augmentation in managing non-alcoholic fatty liver disease: an in-depth umbrella review of meta-analyses with grade assessment. Ann Med Surg (Lond) 2024; 86:4714-4731. [PMID: 39118769 PMCID: PMC11305784 DOI: 10.1097/ms9.0000000000002276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 06/03/2024] [Indexed: 08/10/2024] Open
Abstract
Background and aim Currently, there are no authorized medications specifically for non-alcoholic fatty liver disease (NAFLD) treatment. Studies indicate that changes in gut microbiota can disturb intestinal balance and impair the immune system and metabolism, thereby elevating the risk of developing and exacerbating NAFLD. Despite some debate, the potential benefits of microbial therapies in managing NAFLD have been shown. Methods A systematic search was undertaken to identify meta-analyses of randomized controlled trials that explored the effects of microbial therapy on the NAFLD population. The goal was to synthesize the existing evidence-based knowledge in this field. Results The results revealed that probiotics played a significant role in various aspects, including a reduction in liver stiffness (MD: -0.38, 95% CI: [-0.49, -0.26]), hepatic steatosis (OR: 4.87, 95% CI: [1.85, 12.79]), decrease in body mass index (MD: -1.46, 95% CI: [-2.43, -0.48]), diminished waist circumference (MD: -1.81, 95% CI: [-3.18, -0.43]), lowered alanine aminotransferase levels (MD: -13.40, 95% CI: [-17.02, -9.77]), decreased aspartate aminotransferase levels (MD: -13.54, 95% CI: [-17.85, -9.22]), lowered total cholesterol levels (MD: -15.38, 95% CI: [-26.49, -4.26]), decreased fasting plasma glucose levels (MD: -4.98, 95% CI: [-9.94, -0.01]), reduced fasting insulin (MD: -1.32, 95% CI: [-2.42, -0.21]), and a decline in homeostatic model assessment of insulin resistance (MD: -0.42, 95% CI: [-0.72, -0.11]) (P<0.05). Conclusion Overall, the results demonstrated that gut microbiota interventions could ameliorate a wide range of indicators including glycemic profile, dyslipidemia, anthropometric indices, and liver injury, allowing them to be considered a promising treatment strategy.
Collapse
Affiliation(s)
| | | | | | | | - Shika Jain
- MVJ Medical College and Research Hospital, Bengaluru, Karnataka, India
| | | | - Fnu Vikash
- Jacobi Medical Center, Albert Einstein College of Medicine, Bronx
| | - Nayanika C. Tummala
- Gitam Institute of Medical Sciences and Research, Visakhapatnam, Andhra Pradesh
| | | | - Arezoo Ghazalgoo
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | | | - Dhruvan Patel
- Drexel University College of Medicine, Philadelphia, Pennsylvania, PA
| |
Collapse
|
21
|
Zhang Y, Zeng L, Ouyang K, Wang W. Cholesterol-Lowering Effect of Polysaccharides from Cyclocarya paliurus In Vitro and in Hypercholesterolemia Mice. Foods 2024; 13:2343. [PMID: 39123535 PMCID: PMC11312258 DOI: 10.3390/foods13152343] [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: 06/26/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
In this study, a new component of Cyclocarya paliurus polysaccharides (CPP20) was precipitated by the gradient ethanol method, and the protective effect of CPP20 on hypercholesterolemia mice was investigated. In vitro, CPP20 had the ability to bind bile salts and inhibit cholesterol micelle solubility, and it could effectively clear free radicals (DPPH•, •OH, and ABTS+). In vivo, CPP20 effectively alleviated hypercholesterolemia and liver damage in mice. After CPP20 intervention, the activity of antioxidant enzymes (SOD, CAT, and GSH-Px) and the level of HDL-C in liver and serum were increased, and the activity of aminotransferase (ALT and AST) and the level of MDA, TC, TG, LDL-C, and TBA were decreased. Molecular experiments showed that CPP20 reduced cholesterol by regulating the mRNA expression of antioxidation-related genes (SOD, GSH-Px, and CAT) and genes related to the cholesterol metabolism (CYP7A1, CYP27A1, SREBP-2, HMGCR, and FXR) in liver. In addition, CPP20 alleviated intestinal microbiota disturbances in mice with hypercholesterolemia and increased levels of SCFAs. Therefore, CPP20 alleviates hypercholesterolemia by alleviating oxidative damage, maintaining cholesterol homeostasis, and regulating gut microbiota.
Collapse
Affiliation(s)
- Yang Zhang
- Jiangxi Province Key Laboratory of Animal Nutrition, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China;
| | - Lei Zeng
- Key Lab for Agro-Product Processing and Quality Control of Nanchang City, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China;
| | - Kehui Ouyang
- Jiangxi Province Key Laboratory of Animal Nutrition, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China;
| | - Wenjun Wang
- Key Lab for Agro-Product Processing and Quality Control of Nanchang City, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China;
| |
Collapse
|
22
|
Brown C, Kariuki W, Zhong HA, Kippes A, Sui Y. Cannabidiol promotes intestinal cholesterol uptake mediated by Pregnane X receptor. Front Endocrinol (Lausanne) 2024; 15:1398462. [PMID: 38957441 PMCID: PMC11217338 DOI: 10.3389/fendo.2024.1398462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 06/05/2024] [Indexed: 07/04/2024] Open
Abstract
Background Cannabidiol (CBD), a non-psychoactive phytocannabinoid of cannabis, is therapeutically used as an analgesic, anti-convulsant, anti-inflammatory, and anti-psychotic drug. There is a growing concern about the adverse side effects posed by CBD usage. Pregnane X receptor (PXR) is a nuclear receptor activated by a variety of dietary steroids, pharmaceutical agents, and environmental chemicals. In addition to the role in xenobiotic metabolism, the atherogenic and dyslipidemic effects of PXR have been revealed in animal models. CBD has a low affinity for cannabinoid receptors, thus it is important to elucidate the molecular mechanisms by which CBD activates cellular signaling and to assess the possible adverse impacts of CBD on pro-atherosclerotic events in cardiovascular system, such as dyslipidemia. Objective Our study aims to explore the cellular and molecular mechanisms by which exposure to CBD activates human PXR and increases the risk of dyslipidemia. Methods Both human hepatic and intestinal cells were used to test if CBD was a PXR agonist via cell-based transfection assay. The key residues within PXR's ligand-binding pocket that CBD interacted with were investigated using computational docking study together with site-directed mutagenesis assay. The C57BL/6 wildtype mice were orally fed CBD in the presence of PXR antagonist resveratrol (RES) to determine how CBD exposure could change the plasma lipid profiles in a PXR-dependent manner. Human intestinal cells were treated with CBD and/or RES to estimate the functions of CBD in cholesterol uptake. Results CBD was a selective agonist of PXR with higher activities on human PXR than rodents PXRs and promoted the dissociation of human PXR from nuclear co-repressors. The key amino acid residues Met246, Ser247, Phe251, Phe288, Trp299, and Tyr306 within PXR's ligand binding pocket were identified to be necessary for the agonistic effects of CBD. Exposure to CBD increased the circulating total cholesterol levels in mice which was partially caused by the induced expression levels of the key intestinal PXR-regulated lipogenic genes. Mechanistically, CBD induced the gene expression of key intestinal cholesterol transporters, which led to the increased cholesterol uptake by intestinal cells. Conclusion CBD was identified as a selective PXR agonist. Exposure to CBD activated PXR signaling and increased the atherogenic cholesterol levels in plasma, which partially resulted from the ascended cholesterol uptake by intestinal cells. Our study provides potential evidence for the future risk assessment of CBD on cardiovascular disease, such as dyslipidemia.
Collapse
Affiliation(s)
- Conner Brown
- Department of Biology, University of Nebraska at Kearney, Kearney, NE, United States
| | - Wangeci Kariuki
- Department of Biology, University of Nebraska at Kearney, Kearney, NE, United States
| | - Haizhen A. Zhong
- Department of Chemistry, University of Nebraska at Omaha, Omaha, NE, United States
| | - Audra Kippes
- Department of Biology, University of Nebraska at Kearney, Kearney, NE, United States
| | - Yipeng Sui
- Department of Biology, University of Nebraska at Kearney, Kearney, NE, United States
| |
Collapse
|
23
|
Deng Y, Wang J, Wang R, Wang Y, Shu X, Wang P, Chen C, Zhang F. Limosilactobacillus fermentum TY-S11 ameliorates hypercholesterolemia via promoting cholesterol excretion and regulating gut microbiota in high-cholesterol diet-fed apolipoprotein E-deficient mice. Heliyon 2024; 10:e32059. [PMID: 38882320 PMCID: PMC11180314 DOI: 10.1016/j.heliyon.2024.e32059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024] Open
Abstract
Hypercholesterolemia is a metabolic disease characterized by elevated cholesterol level in the blood, which is a risk factor for many diseases. Probiotic intervention may be one of the ways to improve hypercholesterolemia. In this study, three strains with better cholesterol removal ability were selected from 60 strains of lactic acid bacteria, and were orally administered to apolipoprotein E-deficient mice on a high-cholesterol diet. Among the three strains, only Limosilactobacillus fermentum TY-S11, which was isolated from the intestine of a longevity person, significantly improved serum and liver lipid levels in hypercholesterolemic mice. Further study found that L. fermentum TY-S11 promoted the excretion of cholesterol in the feces and inhibited the absorption of cholesterol in the small intestine. As for gut microbiota, the results showed that L. fermentum TY-S11 not only prevented the reduction of diversity caused by high-cholesterol diet, but also increased the contents of short-chain fatty acids in feces. These results confirmed the ameliorative effect of L. fermentum TY-S11 on hypercholesterolemia.
Collapse
Affiliation(s)
- Yadan Deng
- Key Laboratory of Conservation, Exploration and Utilization of Southwest Characteristic Bacterial Germplasm Resources, Chongqing Tianyou Dairy Co., Ltd., Chongqing, 401120, China
| | - Jing Wang
- Key Laboratory of Conservation, Exploration and Utilization of Southwest Characteristic Bacterial Germplasm Resources, Chongqing Tianyou Dairy Co., Ltd., Chongqing, 401120, China
| | - Ran Wang
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing, 100190, China
| | - Yuying Wang
- Key Laboratory of Conservation, Exploration and Utilization of Southwest Characteristic Bacterial Germplasm Resources, Chongqing Tianyou Dairy Co., Ltd., Chongqing, 401120, China
| | - Xi Shu
- Key Laboratory of Conservation, Exploration and Utilization of Southwest Characteristic Bacterial Germplasm Resources, Chongqing Tianyou Dairy Co., Ltd., Chongqing, 401120, China
| | - Pengjie Wang
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing, 100190, China
| | - Chong Chen
- Key Laboratory of Conservation, Exploration and Utilization of Southwest Characteristic Bacterial Germplasm Resources, Chongqing Tianyou Dairy Co., Ltd., Chongqing, 401120, China
| | - Feng Zhang
- Key Laboratory of Conservation, Exploration and Utilization of Southwest Characteristic Bacterial Germplasm Resources, Chongqing Tianyou Dairy Co., Ltd., Chongqing, 401120, China
| |
Collapse
|
24
|
Jiang Y, Pang S, Liu X, Wang L, Liu Y. The Gut Microbiome Affects Atherosclerosis by Regulating Reverse Cholesterol Transport. J Cardiovasc Transl Res 2024; 17:624-637. [PMID: 38231373 DOI: 10.1007/s12265-024-10480-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 01/07/2024] [Indexed: 01/18/2024]
Abstract
The human system's secret organ, the gut microbiome, has received considerable attention. Emerging research has yielded substantial scientific evidence indicating that changes in gut microbial composition and microbial metabolites may contribute to the development of atherosclerotic cardiovascular disease. The burden of cardiovascular disease on healthcare systems is exacerbated by atherosclerotic cardiovascular disease, which continues to be the leading cause of mortality globally. Reverse cholesterol transport is a powerful protective mechanism that effectively prevents excessive accumulation of cholesterol for atherosclerotic cardiovascular disease. It has been revealed how the gut microbiota modulates reverse cholesterol transport in patients with atherosclerotic risk. In this review, we highlight the complex interactions between microbes, their metabolites, and their potential impacts in reverse cholesterol transport. We also explore the feasibility of modulating gut microbes and metabolites to facilitate reverse cholesterol transport as a novel therapy for atherosclerosis.
Collapse
Affiliation(s)
- Yangyang Jiang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shuchao Pang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.
| | - Xiaoyu Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lixin Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.
| |
Collapse
|
25
|
Schade DS, Nagamallika Godasi B, Duro T, Adolphe A, Eaton RP. New Insights Into the Importance of Dietary Cholesterol in Preventing Cardiovascular Disease. Endocr Pract 2024; 30:598-600. [PMID: 38522824 DOI: 10.1016/j.eprac.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 03/26/2024]
Affiliation(s)
- David S Schade
- Division of Endocrinology, University of New Mexico School of Medicine, Albuquerque, New Mexico.
| | | | - Teodor Duro
- Division of Endocrinology, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Allen Adolphe
- General Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - R Philip Eaton
- Division of Endocrinology, University of New Mexico School of Medicine, Albuquerque, New Mexico
| |
Collapse
|
26
|
Amini-Salehi E, Samethadka Nayak S, Maddineni G, Mahapatro A, Keivanlou MH, Soltani Moghadam S, Vakilpour A, Aleali MS, Joukar F, Hashemi M, Norouzi N, Bakhshi A, Bahrampourian A, Mansour-Ghanaei F, Hassanipour S. Can modulation of gut microbiota affect anthropometric indices in patients with non-alcoholic fatty liver disease? An umbrella meta-analysis of randomized controlled trials. Ann Med Surg (Lond) 2024; 86:2900-2910. [PMID: 38694388 PMCID: PMC11060227 DOI: 10.1097/ms9.0000000000001740] [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: 09/06/2023] [Accepted: 01/08/2024] [Indexed: 05/04/2024] Open
Abstract
Background and aim Modulating the gut microbiota population by administration of probiotics, prebiotics, and synbiotics has shown to have a variety of health benefits in different populations, particularly those with metabolic disorders. Although the promising effects of these compounds have been observed in the management of patients with non-alcoholic fatty liver disease (NAFLD), the exact effects and the mechanisms of action are yet to be understood. In the present study, we aimed to evaluate how gut microbiota modulation affects anthropometric indices of NAFLD patients to achieve a comprehensive summary of current evidence-based knowledge. Methods Two researchers independently searched international databases, including PubMed, Scopus, and Web of Science, from inception to June 2023. Meta-analysis studies that evaluated the effects of probiotics, prebiotics, and synbiotics on patients with NAFLD were entered into our umbrella review. The data regarding anthropometric indices, including body mass index, weight, waist circumference (WC), and waist-to-hip ratio (WHR), were extracted by the investigators. The authors used random effect model for conducting the meta-analysis. Subgroup analysis and sensitivity analysis were also performed. Results A total number of 13 studies were finally included in our study. Based on the final results, BMI was significantly decreased in NAFLD patients by modulation of gut microbiota [effect size (ES): -0.18, 05% CI: -0.25, -0.11, P<0.001]; however, no significant alteration was observed in weight and WC (ES: -1.72, 05% CI: -3.48, 0.03, P=0.055, and ES: -0.24, 05% CI: -0.75, 0.26, P=0.353, respectively). The results of subgroup analysis showed probiotics had the most substantial effect on decreasing BMI (ES: -0.77, 95% CI: -1.16, -0.38, P<0.001) followed by prebiotics (ES: -0.51, 95% CI: -0.76, -0.27, P<0.001) and synbiotics (ES: -0.12, 95% CI: -0.20, -0.04, P=0.001). Conclusion In conclusion, the present umbrella meta-analysis showed that although modulation of gut microbiota by administration of probiotics, prebiotics, and synbiotics had promising effects on BMI, no significant change was observed in the WC and weight of the patients. No sufficient data were available for other anthropometric indices including waist-to-hip ratio and waist-to-height ratio and future meta-analyses should be done in this regard.
Collapse
Affiliation(s)
- Ehsan Amini-Salehi
- Gastrointestinal and Liver Diseases Research Center
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | | | | | | | | | | | - Azin Vakilpour
- School of Medicine, Guilan University of Medical Sciences, Rasht
| | | | | | - Mohammad Hashemi
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | | | | | | | | |
Collapse
|
27
|
Lv X, Liu X, Peng Y, Li W, Wang J, Chen X, Lei J, Tang C, Luo S, Mai W, Cai Y, Fan Q, Liu C, Zhang L. Medium-intensity statin with ezetimibe versus high-intensity statin in acute ischemic cerebrovascular disease (MESIA): A randomized clinical trial. J Stroke Cerebrovasc Dis 2024; 33:107647. [PMID: 38431112 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107647] [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: 10/17/2023] [Revised: 01/12/2024] [Accepted: 02/21/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND High-risk stroke patients are recommended to receive high-intensity statin therapy to reduce the risk of stroke recurrence. However, doubling the dosage of statin drugs did not increase the achievement rate of LDL-C target or provide additional clinical benefits, but significantly increased the risk of adverse reactions. Statins and ezetimibe work through different mechanisms and the combined use of statins and ezetimibe significantly improves outcomes with comparable safety profiles. We tested the hypothesis that moderate-intensity statin with ezetimibe may offer advantages over the conventional high-intensity statin regimen in terms of efficacy and safety. METHODS We conducted a randomized controlled trial. Eligible participants were aged 18 years or older with acute ischemic cerebrovascular disease. We randomly assigned (1:1) participants within the acute phase of ischemic stroke, i.e., within 1 week after the onset of mild ischemic stroke (NIHSS score ≤ 5), within 1 month for severe cases (NIHSS score ≥ 16), and within 2 weeks for the rest, as well as patients with TIA within 1 week of symptom onset, to receive either moderate-intensity statin with ezetimibe (either 10-20 mg atorvastatin calcium tablets plus a 10 mg ezetimibe tablet, or 5-10 mg rosuvastatin calcium tablets once per day plus a 10 mg ezetimibe tablet once per day) or high-intensity statin (40 mg atorvastatin calcium tablets or 20 mg rosuvastatin calcium tablets once per day) for 3 months. Randomization was performed using a random number table method. The primary efficacy outcome was the level and achievement rate of LDL-C after 3 months of treatment, specifically LDL-C ≤ 1.8 mmol/L or a reduction in LDL-C ≥ 50 %. The secondary outcome was the incidence of new stroke or transient ischemic attack (TIA) within 3 months. The safety outcome was liver and renal function tests, and the occurrence of statin-related muscle events within 3 months. FINDINGS This trial took place between March 15, 2022, and March 7, 2023. Among 382 patients screened, 150 patients were randomly assigned to receive either medium-intensity statins with ezetimibe (n = 75) or high-intensity statins (n = 75). Median age was 60.0 years (IQR 52.75-70.25); 49 (36.6 %) were women and 85 (63.4 %) were men. The target achievement of LDL-C at 3 months occurred in 62 (89.86 %) of 69 patients in the medium-intensity statin with ezetimibe group and 46 (70.77 %) of 65 patients in the high-intensity statin group (P=0.005, OR=0.273, 95 % CI: 0.106, 0.705). The reduction magnitude of LDL-C in moderate-intensity statin with ezetimibe group was significantly higher (-56.540 % vs -47.995 %, P=0.001). Moderate-intensity statin with ezetimibe group showing a trend of a greater reduction in LDL-C absolute value than high-intensity statin group but without statistical significance (-1.77±0.90 vs -1.50±0.89, P=0.077). New AIS or TIA within 3 months, liver and renal function tests, and the occurrence of statin-related muscle events within 3 months were also statistically insignificant. Multivariate logistic regression analysis showed that both gender and lipid-lowering regimen as independent risk factors influencing the rate of LDL-C achievement in individuals diagnosed with acute ischemic cerebrovascular disease, but only lipid-lowering regimen had predictive value. INTERPRETATION Compared to guideline-recommended high-intensity statin therapy, moderate-intensity statin with ezetimibe further improved the achievement rate of LDL-C in patients with acute ischemic cerebrovascular disease, with a higher reduction magnitude in LDL-C. In terms of safety, there was no significant difference between the two regimens, suggesting that moderate-intensity statin with ezetimibe can also be considered as an initial treatment option for patients with acute ischemic cerebrovascular disease.
Collapse
Affiliation(s)
- Xuxian Lv
- The Fifth Affiliated Hospital, Department of Cerebrovascular Disease, Sun Yat-Sen University, China
| | - Xudong Liu
- The Fifth Affiliated Hospital, Department of Cerebrovascular Disease, Sun Yat-Sen University, China
| | - Yanfang Peng
- The Fifth Affiliated Hospital, Department of Cerebrovascular Disease, Sun Yat-Sen University, China
| | - Wenbin Li
- The Fifth Affiliated Hospital, Department of Cerebrovascular Disease, Sun Yat-Sen University, China
| | - Jianing Wang
- The Fifth Affiliated Hospital, Department of Cerebrovascular Disease, Sun Yat-Sen University, China
| | - Xiaofeng Chen
- The Fifth Affiliated Hospital, Department of Cerebrovascular Disease, Sun Yat-Sen University, China
| | - Junjie Lei
- The Fifth Affiliated Hospital, Department of Cerebrovascular Disease, Sun Yat-Sen University, China
| | - Chaogang Tang
- The Fifth Affiliated Hospital, Department of Cerebrovascular Disease, Sun Yat-Sen University, China
| | - Shijian Luo
- The Fifth Affiliated Hospital, Department of Cerebrovascular Disease, Sun Yat-Sen University, China
| | - Weihua Mai
- The Fifth Affiliated Hospital, Department of Cerebrovascular Disease, Sun Yat-Sen University, China
| | - Yiming Cai
- The Fifth Affiliated Hospital, Department of Cerebrovascular Disease, Sun Yat-Sen University, China
| | - Qian Fan
- The Fifth Affiliated Hospital, Department of Cerebrovascular Disease, Sun Yat-Sen University, China
| | - Chenhao Liu
- The Fifth Affiliated Hospital, Department of Cerebrovascular Disease, Sun Yat-Sen University, China
| | - Lei Zhang
- The Fifth Affiliated Hospital, Department of Cerebrovascular Disease, Sun Yat-Sen University, China.
| |
Collapse
|
28
|
Zhang L, Shi Y, Liang B, Li X. An overview of the cholesterol metabolism and its proinflammatory role in the development of MASLD. Hepatol Commun 2024; 8:e0434. [PMID: 38696365 PMCID: PMC11068152 DOI: 10.1097/hc9.0000000000000434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/05/2024] [Indexed: 05/04/2024] Open
Abstract
Cholesterol is an essential lipid molecule in mammalian cells. It is not only involved in the formation of cell membranes but also serves as a raw material for the synthesis of bile acids, vitamin D, and steroid hormones. Additionally, it acts as a covalent modifier of proteins and plays a crucial role in numerous life processes. Generally, the metabolic processes of cholesterol absorption, synthesis, conversion, and efflux are strictly regulated. Excessive accumulation of cholesterol in the body is a risk factor for metabolic diseases such as cardiovascular disease, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease (MASLD). In this review, we first provide an overview of the discovery of cholesterol and the fundamental process of cholesterol metabolism. We then summarize the relationship between dietary cholesterol intake and the risk of developing MASLD, and also the animal models of MASLD specifically established with a cholesterol-containing diet. In the end, the role of cholesterol-induced inflammation in the initiation and development of MASLD is discussed.
Collapse
Affiliation(s)
- Linqiang Zhang
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Yongqiong Shi
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Bin Liang
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Xi Li
- Institute of Life Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
29
|
Elste J, Cast N, Udawatte S, Adhikari K, Payen SH, Verma SC, Shukla D, Swanson-Mungerson M, Tiwari V. Co-Expression of Niemann-Pick Type C1-Like1 (NPC1L1) with ACE2 Receptor Synergistically Enhances SARS-CoV-2 Entry and Fusion. Biomedicines 2024; 12:821. [PMID: 38672177 PMCID: PMC11048565 DOI: 10.3390/biomedicines12040821] [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: 02/23/2024] [Revised: 03/20/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
The entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) into human embryonic kidney (HEK293T) cells has been shown to be a cholesterol-rich, lipid raft-dependent process. In this study, we investigated if the presence of a cholesterol uptake receptor Niemann-pick type c1-like1 (NPC1L1) impacts SARS-CoV-2 cell entry. Initially, we utilized reporter-based pseudovirus cell entry assays and a spike (S) glycoprotein-mediated cell-to-cell fusion assay. Using Chinese hamster ovary (CHO-K1) cells, which lack endogenous receptors for SARS-CoV-2 entry, our data showed that the co-expression of NPC1L1 together with the ACE2 receptor synergistically increased SARS-CoV-2 pseudovirus entry even more than the cells expressing ACE-2 receptor alone. Similar results were also found with the HEK293T cells endogenously expressing the ACE2 receptor. Co-cultures of effector cells expressing S glycoprotein together with target cells co-expressing ACE-2 receptor with NPC1L1 significantly promoted quantitative cell-to-cell fusion, including syncytia formation. Finally, we substantiated that an elevated expression of NPC1L1 enhanced entry, whereas the depletion of NPC1L1 resulted in a diminished SARS-CoV-2 entry in HEK293T-ACE2 cells using authentic SARS-CoV-2 virus in contrast to their respective control cells. Collectively, these findings underscore the pivotal role of NPC1L1 in facilitating the cellular entry of SARS-CoV-2. Importance: Niemann-Pick type C1-like1 (NPC1L1) is an endosomal membrane protein that regulates intracellular cholesterol trafficking. This protein has been demonstrated to play a crucial role in the life cycle of several clinically important viruses. Although SARS-CoV-2 exploits cholesterol-rich lipid rafts as part of its viral entry process, the role of NPC1L1 in SARS-CoV-2 entry remains unclear. Our research represents the first-ever demonstration of NPC1L1's involvement in facilitating SARS-CoV-2 entry. The observed role of NPC1L1 in human kidney cells is not only highly intriguing but also quite relevant. This relevance stems from the fact that NPC1L1 exhibits high expression levels in several organs, including the kidneys, and the fact that kidney damages are reported during severe cases of SARS-CoV-2. These findings may help us understand the new functions and mechanisms of NPC1L1 and could contribute to the identification of new antiviral targets.
Collapse
Affiliation(s)
- James Elste
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL 60515, USA; (J.E.); (N.C.); (M.S.-M.)
| | - Nicole Cast
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL 60515, USA; (J.E.); (N.C.); (M.S.-M.)
| | - Shalini Udawatte
- School of Chemistry & Biochemistry, Georgia Institute of Technology, North Ave NW, Atlanta, GA 30332, USA;
| | - Kabita Adhikari
- Department of Microbiology & Immunology, University of Reno, Reno, NV 89557, USA; (K.A.); (S.H.P.); (S.C.V.)
| | - Shannon Harger Payen
- Department of Microbiology & Immunology, University of Reno, Reno, NV 89557, USA; (K.A.); (S.H.P.); (S.C.V.)
| | - Subhash C. Verma
- Department of Microbiology & Immunology, University of Reno, Reno, NV 89557, USA; (K.A.); (S.H.P.); (S.C.V.)
| | - Deepak Shukla
- Department of Microbiology and Immunology, University of Illinois, Chicago, IL 60612, USA;
| | - Michelle Swanson-Mungerson
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL 60515, USA; (J.E.); (N.C.); (M.S.-M.)
| | - Vaibhav Tiwari
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL 60515, USA; (J.E.); (N.C.); (M.S.-M.)
| |
Collapse
|
30
|
Koehn LM, Steele JR, Schittenhelm RB, Turner BJ, Nicolazzo JA. Sex-Dependent Changes to the Intestinal and Hepatic Abundance of Drug Transporters and Metabolizing Enzymes in the SOD1 G93A Mouse Model of Amyotrophic Lateral Sclerosis. Mol Pharm 2024; 21:1756-1767. [PMID: 38415587 DOI: 10.1021/acs.molpharmaceut.3c01089] [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: 02/29/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by death and dysfunction of motor neurons that result in a rapidly progressing loss of motor function. While there are some data on alterations at the blood-brain barrier (BBB) in ALS and their potential impact on CNS trafficking of drugs, little is reported on the impact of this disease on the expression of drug-handling proteins in the small intestine and liver. This may impact the dosing of the many medicines that individuals with ALS are prescribed. In the present study, a proteomic evaluation was performed on small intestine and liver samples from postnatal day 120 SOD1G93A mice (a model of familial ALS that harbors a human mutant form of superoxide dismutase 1) and wild-type (WT) littermates (n = 7/genotype/sex). Untargeted, quantitative proteomics was undertaken using either label-based [tandem mass tag (TMT)] or label-free [data-independent acquisition (DIA)] acquisition strategies on high-resolution mass spectrometric instrumentation. Copper chaperone for superoxide dismutase (CCS) was significantly higher in SOD1G93A samples compared to the WT samples for both sexes and tissues, therefore representing a potential biomarker for ALS in this mouse model. Relative to WT mice, male SOD1G93A mice had significantly different proteins (Padj < 0.05, |fold-change|>1.2) in the small intestine (male 22, female 1) and liver (male 140, female 3). This included an up-regulation of intestinal transporters for dietary glucose [solute carrier (SLC) SLC5A1] and cholesterol (Niemann-Pick c1-like 1), as well as for several drugs (e.g., SLC15A1), in the male SOD1G93A mice. There was both an up-regulation (e.g., SLCO2A1) and down-regulation (ammonium transporter rh type b) of transporters in the male SOD1G93A liver. In addition, there was both an up-regulation (e.g., phosphoenolpyruvate carboxykinase) and down-regulation (e.g., carboxylesterase 1) of metabolizing enzymes in the male SOD1G93A liver. This proteomic data set identified male-specific changes to key small intestinal and hepatic transporters and metabolizing enzymes that may have important implications for the bioavailability of nutrients and drugs in individuals with ALS.
Collapse
Affiliation(s)
- Liam M Koehn
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Joel R Steele
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Bradley J Turner
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Victoria, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| |
Collapse
|
31
|
Hu X, Chen F, Jia L, Long A, Peng Y, Li X, Huang J, Wei X, Fang X, Gao Z, Zhang M, Liu X, Chen YG, Wang Y, Zhang H, Wang Y. A gut-derived hormone regulates cholesterol metabolism. Cell 2024; 187:1685-1700.e18. [PMID: 38503280 DOI: 10.1016/j.cell.2024.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 11/18/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024]
Abstract
The reciprocal coordination between cholesterol absorption in the intestine and de novo cholesterol synthesis in the liver is essential for maintaining cholesterol homeostasis, yet the mechanisms governing the opposing regulation of these processes remain poorly understood. Here, we identify a hormone, Cholesin, which is capable of inhibiting cholesterol synthesis in the liver, leading to a reduction in circulating cholesterol levels. Cholesin is encoded by a gene with a previously unknown function (C7orf50 in humans; 3110082I17Rik in mice). It is secreted from the intestine in response to cholesterol absorption and binds to GPR146, an orphan G-protein-coupled receptor, exerting antagonistic downstream effects by inhibiting PKA signaling and thereby suppressing SREBP2-controlled cholesterol synthesis in the liver. Therefore, our results demonstrate that the Cholesin-GPR146 axis mediates the inhibitory effect of intestinal cholesterol absorption on hepatic cholesterol synthesis. This discovered hormone, Cholesin, holds promise as an effective agent in combating hypercholesterolemia and atherosclerosis.
Collapse
Affiliation(s)
- Xiaoli Hu
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Fengyi Chen
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Liangjie Jia
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Aijun Long
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ying Peng
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xu Li
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Junfeng Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xueyun Wei
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xinlei Fang
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zihua Gao
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Mengxian Zhang
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiao Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Ye-Guang Chen
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China; Guangzhou Laboratory, Guangzhou 510005, China; School of Basic Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Yan Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Huijie Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Yiguo Wang
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
32
|
Kharve K, Engley AS, Paine MF, Sprowl JA. Impact of Drug-Mediated Inhibition of Intestinal Transporters on Nutrient and Endogenous Substrate Disposition…an Afterthought? Pharmaceutics 2024; 16:447. [PMID: 38675109 PMCID: PMC11053474 DOI: 10.3390/pharmaceutics16040447] [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: 02/07/2024] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 04/28/2024] Open
Abstract
A large percentage (~60%) of prescription drugs and new molecular entities are designed for oral delivery, which requires passage through a semi-impervious membrane bilayer in the gastrointestinal wall. Passage through this bilayer can be dependent on membrane transporters that regulate the absorption of nutrients or endogenous substrates. Several investigations have provided links between nutrient, endogenous substrate, or drug absorption and the activity of certain membrane transporters. This knowledge has been key in the development of new therapeutics that can alleviate various symptoms of select diseases, such as cholestasis and diabetes. Despite this progress, recent studies revealed potential clinical dangers of unintended altered nutrient or endogenous substrate disposition due to the drug-mediated disruption of intestinal transport activity. This review outlines reports of glucose, folate, thiamine, lactate, and bile acid (re)absorption changes and consequent adverse events as examples. Finally, the need to comprehensively expand research on intestinal transporter-mediated drug interactions to avoid the unwanted disruption of homeostasis and diminish therapeutic adverse events is highlighted.
Collapse
Affiliation(s)
- Kshitee Kharve
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA;
| | - Andrew S. Engley
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA; (A.S.E.); (M.F.P.)
| | - Mary F. Paine
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA; (A.S.E.); (M.F.P.)
| | - Jason A. Sprowl
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA;
| |
Collapse
|
33
|
Heida A, van Dijk T, Smit M, Koehorst M, Koster M, Kloosterhuis N, Havinga R, Bloks VW, Wolters JC, de Bruin A, Kuivenhoven JA, de Boer JF, Kuipers F, van de Sluis B. Changes in bile acid composition are correlated with reduced intestinal cholesterol uptake in intestine-specific WASH-deficient mice. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159445. [PMID: 38086439 DOI: 10.1016/j.bbalip.2023.159445] [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: 10/20/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
The Wiskott-Aldrich syndrome protein and SCAR homolog (WASH) complex is a pentameric protein complex localized at endosomes, where it facilitates the transport of numerous receptors from endosomes toward the plasma membrane. Recent studies have shown that the WASH complex plays an essential role in cholesterol and glucose homeostasis in humans and mice. To investigate the physiological importance of intestinal WASH, we ablated the WASH component WASHC1 specifically in murine enterocytes. Male and female intestine-specific WASHC1-deficient mice (Washc1IKO) were challenged with either a standard chow diet or a high-cholesterol (1.25 %) diet (HCD). Washc1IKO mice fed a standard diet did not present any apparent phenotype, but when fed an HCD, their hepatic cholesterol levels were ~ 50 % lower compared to those observed in control mice. The intestinal cholesterol absorption was almost 2-fold decreased in Washc1IKO mice, which translated into increased fecal neutral sterol loss. The intestinal expression of cholesterogenic genes, such as Hmgcs1, Hmgcr, and Ldlr, was significantly higher in Washc1IKO mice than in control mice and correlated with increased whole-body de novo cholesterol synthesis, likely to compensate for impaired intestinal cholesterol absorption. Unexpectedly, the ratio of biliary 12α-/non-12α-hydroxylated bile acids (BAs) was decreased in Washc1IKO mice and reversing this reduced ratio by feeding the mice with the HCD supplemented with 0.5 % (w/w) sodium cholate normalized the improvement of hepatic cholesterol levels in Washc1IKO mice. Our data indicate that the intestinal WASH complex plays an important role in intestinal cholesterol absorption, likely by modulating biliary BA composition.
Collapse
Affiliation(s)
- Andries Heida
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Theo van Dijk
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Marieke Smit
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Martijn Koehorst
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mirjam Koster
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Niels Kloosterhuis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rick Havinga
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Vincent W Bloks
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Justina C Wolters
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Alain de Bruin
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, the Netherlands
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jan Freark de Boer
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Folkert Kuipers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bart van de Sluis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
34
|
Huang P, Ran J, Zhu W, Dai W, Tang Y, Lian P, Huang X, Li R. PCSK9 dysregulates cholesterol homeostasis and triglyceride metabolism in olanzapine-induced hepatic steatosis via both receptor-dependent and receptor-independent pathways. FASEB J 2024; 38:e23464. [PMID: 38358343 DOI: 10.1096/fj.202301748r] [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: 08/29/2023] [Revised: 01/06/2024] [Accepted: 01/22/2024] [Indexed: 02/16/2024]
Abstract
Schizophrenia, affecting approximately 1% of the global population, is often treated with olanzapine. Despite its efficacy, olanzapine's prolonged use has been associated with an increased risk of cardiovascular diseases and nonalcoholic fatty liver disease (NAFLD); however, the underlying mechanism remains unclear. Proprotein convertase subtilisin kexin type 9 (PCSK9) plays a crucial role in lipid metabolism and is involved in NAFLD pathogenesis via an unknown mechanism. This study aims to investigate the role of PCSK9 in olanzapine-induced NAFLD. C57BL/6J mice and HepG2 and AML12 cell lines were treated with varying concentrations of olanzapine to examine the effects of olanzapine on PCSK9 and lipid metabolism. PCSK9 levels were manipulated using recombinant proteins, plasmids, and small interfering RNAs in vitro, and the effects on hepatic lipid accumulation and gene expression related to lipid metabolism were assessed. Olanzapine treatment significantly increased PCSK9 levels in both animal and cell line models, correlating with elevated lipid accumulation. PCSK9 manipulation demonstrated its central role in mediating hepatic steatosis through both receptor-dependent pathways (impacting NPC1L1) and receptor-independent pathways (affecting lipid synthesis, uptake, and cholesterol biosynthesis). Interestingly, upregulation of SREBP-1c, rather than SREBP-2, was identified as a key driver of PCSK9 increase in olanzapine-induced NAFLD. Our findings establish PCSK9 as a pivotal factor in olanzapine-induced NAFLD, influencing both receptor-related and metabolic pathways. This highlights PCSK9 inhibitors as potential therapeutic agents for managing NAFLD in schizophrenia patients treated with olanzapine.
Collapse
Affiliation(s)
- Piaopiao Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juanli Ran
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenqiang Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen Dai
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Yaxin Tang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Pingan Lian
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiansheng Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Rong Li
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
35
|
Dong H, Chen R, Xu F, Cheng F. Can Lipid-Lowering Drugs Reduce the Risk of Cholelithiasis? A Mendelian Randomization Study. Clin Epidemiol 2024; 16:131-141. [PMID: 38410417 PMCID: PMC10896097 DOI: 10.2147/clep.s439642] [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: 09/10/2023] [Accepted: 02/09/2024] [Indexed: 02/28/2024] Open
Abstract
Background and Aims Cholelithiasis etiology intricately involves lipid metabolism. We sought to investigate the plausible causal link between genetically proxied lipid-lowering medications-specifically HMGCR inhibitors, PCSK9 inhibitors, and NPC1L1 inhibitors-and cholelithiasis risk. Methods Our study utilized two genetic instruments for exposure to lipid-lowering drugs. These instruments encompassed genetic variants linked to low-density lipoprotein (LDL) cholesterol within or in proximity to drug target genes, along with loci governing gene expression traits of these targets. Effect estimates were derived through Inverse-variance-weighted MR (IVW-MR) and summary-data-based MR (SMR) methods. Results Higher HMGCR-mediated LDL cholesterol levels (IVW-MR, OR = 2.15, 95% CI = 1.58-2.94; P = 0.000) and increased HMGCR expression (SMR, OR = 1.19, 95% CI = 1.04-1.37; P = 0.014) are linked to elevated cholelithiasis risk, suggesting potential benefits of HMGCR inhibition. In contrast, higher PCSK9-mediated LDL cholesterol levels (IVW-MR, OR = 0.72, 95% CI = 0.56-0.94; P = 0.015) and increased PCSK9 expression (SMR, OR = 0.90, 95% CI = 0.82-0.99; P = 0.035) both correlate with lower cholelithiasis risk, indicating that PCSK9 inhibition may elevate this risk. Nevertheless, no substantial link emerged between NPC1L1-mediated LDL cholesterol or NPC1L1 expression and cholelithiasis in both IVW-MR and SMR analyses. Conclusion This MR investigation affirms the causal link between the utilization of HMGCR inhibitors and a diminished risk of cholelithiasis. Additionally, it indicates a causal link between PCSK9 inhibitors use and increased cholelithiasis risk. However, no significant correlation was found between NPC1L1 inhibitors use and cholelithiasis risk.
Collapse
Affiliation(s)
- Hao Dong
- Department of Gastroenterology and Hepatology, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
| | - Rong Chen
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, People’s Republic of China
| | - Fang Xu
- Clinical Medical Laboratory Center, Taizhou People’s Hospital, Taizhou, Jiangsu, 225300, People’s Republic of China
| | - Fang Cheng
- Department of Gastroenterology, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430023, People’s Republic of China
| |
Collapse
|
36
|
Wang Y, Gao L. Cholesterol: A friend to viruses. Int Rev Immunol 2024; 43:248-262. [PMID: 38372266 DOI: 10.1080/08830185.2024.2314577] [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: 09/17/2023] [Accepted: 01/28/2024] [Indexed: 02/20/2024]
Abstract
Cholesterol is a key life-sustaining molecule which regulates membrane fluidity and serves as a signaling mediator. Cholesterol homeostasis is closely related to various pathological conditions including tumor, obesity, atherosclerosis, Alzheimer's disease and viral infection. Viral infection disrupts host cholesterol homeostasis, facilitating their own survival. Meanwhile, the host cells strive to reduce cholesterol accessibility to limit viral infection. This review focuses on the regulation of cholesterol metabolism and the role of cholesterol in viral infection, specifically providing an overview of cholesterol as a friend to promote viral entry, replication, assembly, release and immune evasion, which might inspire valuable thinking for pathogenesis and intervention of viral infection.
Collapse
Affiliation(s)
- Yingchun Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity, and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
37
|
Guo J, Chen S, Zhang Y, Liu J, Jiang L, Hu L, Yao K, Yu Y, Chen X. Cholesterol metabolism: physiological regulation and diseases. MedComm (Beijing) 2024; 5:e476. [PMID: 38405060 PMCID: PMC10893558 DOI: 10.1002/mco2.476] [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: 07/17/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 02/27/2024] Open
Abstract
Cholesterol homeostasis is crucial for cellular and systemic function. The disorder of cholesterol metabolism not only accelerates the onset of cardiovascular disease (CVD) but is also the fundamental cause of other ailments. The regulation of cholesterol metabolism in the human is an extremely complex process. Due to the dynamic balance between cholesterol synthesis, intake, efflux and storage, cholesterol metabolism generally remains secure. Disruption of any of these links is likely to have adverse effects on the body. At present, increasing evidence suggests that abnormal cholesterol metabolism is closely related to various systemic diseases. However, the exact mechanism by which cholesterol metabolism contributes to disease pathogenesis remains unclear, and there are still unknown factors. In this review, we outline the metabolic process of cholesterol in the human body, especially reverse cholesterol transport (RCT). Then, we discuss separately the impact of abnormal cholesterol metabolism on common diseases and potential therapeutic targets for each disease, including CVD, tumors, neurological diseases, and immune system diseases. At the end of this review, we focus on the effect of cholesterol metabolism on eye diseases. In short, we hope to provide more new ideas for the pathogenesis and treatment of diseases from the perspective of cholesterol.
Collapse
Affiliation(s)
- Jiarui Guo
- Eye Center of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| | - Silong Chen
- Eye Center of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| | - Ying Zhang
- Eye Center of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
- Institute of Translational MedicineZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| | - Jinxia Liu
- Eye Center of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| | - Luyang Jiang
- Eye Center of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| | - Lidan Hu
- National Clinical Research Center for Child HealthThe Children's HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| | - Ke Yao
- Eye Center of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| | - Yibo Yu
- Eye Center of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| | - Xiangjun Chen
- Eye Center of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
- Institute of Translational MedicineZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| |
Collapse
|
38
|
Zhao J, Chen R, Luo M, Gong H, Li K, Zhao Q. Lipid-lowering drugs and inflammatory bowel disease's risk: a drug-target Mendelian randomization study. Diabetol Metab Syndr 2024; 16:12. [PMID: 38191425 PMCID: PMC10775535 DOI: 10.1186/s13098-023-01252-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 12/31/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) has been associated with lipid-lowering drugs in observational studies. Drug-target Mendelian randomization (MR) was utilized in this study to examine the causal relationship between lipid-lowering drugs and incidence of IBD, aiming to identify new preventive uses for the drugs. METHODS We identified instrumental variables for three classes of lipid-lowering drugs: HMGCR inhibitors, PCSK9 inhibitors, and NPC1L1 inhibitors, using data from the Global Lipids Genetics Consortium. Summary statistics of IBD were obtained from UK Inflammatory Bowel Disease Genetics. The summary-data-based MR (SMR) and the inverse-variance weighted (IVW) MR were used for analysis. Sensitivity analyses were performed by conventional MR methods. RESULTS The SMR analysis showed no significant genetic association between increased gene expression of HMGCR, PCSK9, and NPC1L1 and IBD, Crohn's disease (CD) and ulcerative colitis (UC). According to IVW-MR analysis, increased HMGCR expression is associated with a reduced risk of IBD (OR = 0.73, 95% confidence interval (CI) 0.59-0.90, P = 0.003) and CD (OR = 0.75, 95% CI 0.57-0.97, P = 0.03), but not with UC. Additionally, increased NPC1L1 gene expression was associated with elevated risk of IBD (OR = 1.60, 95% CI 1.07-2.40, P = 0.023), but not with CD and UC. However, no significant causal relationships were found between PCSK9 gene expression and IBD, CD, and UC. The sensitivity analysis demonstrated no evidence of heterogeneity or pleiotropy among the reported results. CONCLUSIONS The heightened expression of genetic variations in HMGCR inhibitor targets could potentially reduce the risk of IBD and CD, while genetic variation in the expression of NPC1L1 targets was positively associated with IBD.
Collapse
Affiliation(s)
- Jiaxi Zhao
- General Practice Ward/International Medical Center Ward General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Rong Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Mengqi Luo
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongping Gong
- General Practice Ward/International Medical Center Ward General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Kaixin Li
- Department of Nephrology, Huadong Hospital, Shanghai, China
| | - Qian Zhao
- General Practice Ward/International Medical Center Ward General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
39
|
Valdivia A, Luque FJ, Llabrés S. Binding of Cholesterol to the N-Terminal Domain of the NPC1L1 Transporter: Analysis of the Epimerization-Related Binding Selectivity and Loop Mutations. J Chem Inf Model 2024; 64:189-204. [PMID: 38152929 PMCID: PMC10777396 DOI: 10.1021/acs.jcim.3c01319] [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: 08/17/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 12/29/2023]
Abstract
Cholesterol is a fat-like substance with a pivotal physiological relevance in humans, and its homeostasis is tightly regulated by various cellular processes, including the import in the small intestine and the reabsorption in the biliary ducts by the Niemann-Pick C1 Like 1 (NPC1L1) importer. NPC1L1 can mediate the absorption of a variety of sterols but strikingly exhibits a large sensitivity to cholesterol epimerization. This study examines the molecular basis of the epimerization-related selective binding of cholesterol by combining extended unbiased molecular dynamics simulations of the apo and holo species of the N-terminal domain of wild-type NPC1L1, in conjunction with relative binding free energy, umbrella sampling, and well-tempered metadynamics calculations. The analysis of the results discloses the existence of two distinct binding modes for cholesterol and epi-cholesterol. The former binds deeper in the cavity, forming key hydrogen-bond interactions with Q95, S56, and a water molecule. In contrast, epi-cholesterol is shifted ca. 3 Å to the mouth of the cavity and the transition to the Q95 site is prevented by an energetic barrier of 4.1 kcal·mol-1. Thus, the configuration of the hydroxyl group of cholesterol, together with the presence of a structural water molecule, is a key feature for effective absorption. Finally, whereas these findings may seemingly be challenged by single-point mutations that impair cholesterol transport but have a mild impact on the binding of cholesterol to the Q95 binding site, our results reveal that they have a drastic influence on the conformational landscape of the α8/β7 loop in the apo species compared to the wild-type protein. Overall, the results give support to the functional role played by the α8/β7 loop in regulating the access of ligands to NPC1L1, and hence to interpreting the impact of these mutations on diseases related to disruption of sterol absorption, paving the way to understanding certain physiological dysfunctions.
Collapse
Affiliation(s)
- Aitor Valdivia
- Departament
de Nutrició, Ciències de l′Alimentació
i Gastronomia, Facultat de Farmàcia
i Ciències de l′Alimentació—Campus Torribera,
Universitat de Barcelona, Prat de la Riba 171, 08921 Santa Coloma de Gramenet, Spain
- Institut
de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - F. Javier Luque
- Departament
de Nutrició, Ciències de l′Alimentació
i Gastronomia, Facultat de Farmàcia
i Ciències de l′Alimentació—Campus Torribera,
Universitat de Barcelona, Prat de la Riba 171, 08921 Santa Coloma de Gramenet, Spain
- Institut
de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain
- Institut
de Química Teòrica i Computacional (IQTCUB), Universitat de Barcelona, 08921 Barcelona, Spain
| | - Salomé Llabrés
- Departament
de Nutrició, Ciències de l′Alimentació
i Gastronomia, Facultat de Farmàcia
i Ciències de l′Alimentació—Campus Torribera,
Universitat de Barcelona, Prat de la Riba 171, 08921 Santa Coloma de Gramenet, Spain
- Institut
de Química Teòrica i Computacional (IQTCUB), Universitat de Barcelona, 08921 Barcelona, Spain
| |
Collapse
|
40
|
Chen J, Chan TTH, Zhou J. Lipid metabolism in the immune niche of tumor-prone liver microenvironment. J Leukoc Biol 2024; 115:68-84. [PMID: 37474318 DOI: 10.1093/jleuko/qiad081] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/23/2023] [Accepted: 07/06/2023] [Indexed: 07/22/2023] Open
Abstract
The liver is a common primary site not only for tumorigenesis, but also for cancer metastasis. Advanced cancer patients with liver metastases also show reduced response rates and survival benefits when treated with immune checkpoint inhibitors. Accumulating evidence has highlighted the importance of the liver immune microenvironment in determining tumorigenesis, metastasis-organotropism, and immunotherapy resistance. Various immune cells such as T cells, natural killer and natural killer T cells, macrophages and dendritic cells, and stromal cells including liver sinusoidal endothelial cells, Kupffer cells, hepatic stellate cells, and hepatocytes are implicated in contributing to the immune niche of tumor-prone liver microenvironment. In parallel, as the major organ for lipid metabolism, the increased abundance of lipids and their metabolites is linked to processes crucial for nonalcoholic fatty liver disease and related liver cancer development. Furthermore, the proliferation, differentiation, and functions of hepatic immune and stromal cells are also reported to be regulated by lipid metabolism. Therefore, targeting lipid metabolism may hold great potential to reprogram the immunosuppressive liver microenvironment and synergistically enhance the immunotherapy efficacy in the circumstance of liver metastasis. In this review, we describe how the hepatic microenvironment adapts to the lipid metabolic alterations in pathologic conditions like nonalcoholic fatty liver disease. We also illustrate how these immunometabolic alterations promote the development of liver cancers and immunotherapy resistance. Finally, we discuss the current therapeutic options and hypothetic combination immunotherapies for the treatment of advanced liver cancers.
Collapse
Affiliation(s)
- Jintian Chen
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| | - Thomas T H Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| | - Jingying Zhou
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| |
Collapse
|
41
|
Tan J, Li Y. Revisiting the interconnection between lipids and vitamin K metabolism: insights from recent research and potential therapeutic implications: a review. Nutr Metab (Lond) 2024; 21:6. [PMID: 38172964 PMCID: PMC10763176 DOI: 10.1186/s12986-023-00779-4] [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: 06/08/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Vitamin K is a lipophilic vitamin, whose absorption, transportation, and distribution are influenced by lipids. The plasma vitamin K level after supplementation is predominantly a lipid-driven effect and independent of existing vitamin K status. However, previous studies examining the efficacy of vitamin K supplementation often overlooked the influence of lipid levels on vitamin K absorption, resulting in inconsistent outcomes. Recent research discovered that impaired transportation of vitamin K2 within uremic high-density lipoproteins (HDL) in individuals with uremia might elucidate the lack of beneficial effects in preventing calcification observed in multiple trials involving menaquinone-7 (MK-7) supplementation among patients with chronic kidney disease. Clinical findings have shown that drugs used to regulate hyperlipidemia interact with the vitamin K antagonist warfarin, because cholesterol and vitamin K share common transport receptors, such as Niemann-Pick C1-like 1 (NPC1L1) and ATP-binding cassette protein G5/G8 (ABCG5/ABCG8), in enterocytes and hepatocytes. Additionally, cholesterol and vitamin K share a common biosynthetic intermediate called geranylgeranyl pyrophosphate (GGPP). It is important to note that statins, which hinder cholesterol synthesis, can also impede vitamin K conversion, ultimately impacting the functionality of vitamin K-dependent proteins. Furthermore, certain studies have indicated that vitamin K supplementation holds potential in managing hyperlipidemia, potentially opening a novel avenue for controlling hyperlipidemia using dietary vitamin K supplements. Therefore, attaining a more comprehensive understanding of the intricate interplay between vitamin K and lipids will yield valuable insights concerning the utilization of vitamin K and lipid regulation.
Collapse
Affiliation(s)
- Jing Tan
- Department of Hematology, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China.
| | - Ying Li
- School of Medicine, North Scihuan Medical College, Nanchong, Sichuan, China
| |
Collapse
|
42
|
Li Z, Zheng D, Zhang T, Ruan S, Li N, Yu Y, Peng Y, Wang D. The roles of nuclear receptors in cholesterol metabolism and reverse cholesterol transport in nonalcoholic fatty liver disease. Hepatol Commun 2024; 8:e0343. [PMID: 38099854 PMCID: PMC10727660 DOI: 10.1097/hc9.0000000000000343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 10/28/2023] [Indexed: 12/18/2023] Open
Abstract
As the most prevalent chronic liver disease globally, NAFLD encompasses a pathological process that ranges from simple steatosis to NASH, fibrosis, cirrhosis, and HCC, closely associated with numerous extrahepatic diseases. While the initial etiology was believed to be hepatocyte injury caused by lipid toxicity from accumulated triglycerides, recent studies suggest that an imbalance of cholesterol homeostasis is of greater significance. The role of nuclear receptors in regulating liver cholesterol homeostasis has been demonstrated to be crucial. This review summarizes the roles and regulatory mechanisms of nuclear receptors in the 3 main aspects of cholesterol production, excretion, and storage in the liver, as well as their cross talk in reverse cholesterol transport. It is hoped that this review will offer new insights and theoretical foundations for the study of the pathogenesis and progression of NAFLD and provide new research directions for extrahepatic diseases associated with NAFLD.
Collapse
|
43
|
Libby P, Pinkosky SL, Nissen SE. Conquering cholesterol: a report from the front lines. Cardiovasc Res 2023; 119:e160-e163. [PMID: 38159295 PMCID: PMC10757583 DOI: 10.1093/cvr/cvad152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/24/2023] [Accepted: 09/04/2023] [Indexed: 01/03/2024] Open
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 01915, USA
| | | | - Steven E Nissen
- Cleveland Clinic Coordinating Center for Clinical Research (C5Research), Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
44
|
Xu C, Fu F, She Y, Xu C. NPC1L1 Plays a Novel Role in Nonalcoholic Fatty Liver Disease. ACS OMEGA 2023; 8:48586-48589. [PMID: 38162748 PMCID: PMC10753569 DOI: 10.1021/acsomega.3c07337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/24/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024]
Abstract
Niemann-Pick C1-Like 1 (NPC1L1) is a key protein in the transport of cholesterol, which exists in the brush marginal membrane of the intestinal epithelial cells and the timid duct membrane of the liver. It affects cholesterol absorption and plasma low-density lipoprotein levels. Cholesterol is both an important component of the cell membrane and a precursor of bile acid and steroid hormone synthesis. Abnormal cholesterol metabolism is closely related to nonalcoholic steatohepatitis (NASH). NASH can progress to fibrosis and cirrhosis, with serious consequences. NPC1L1 is involved in the regulation of cholesterol and lipid metabolism and plays an important role in maintaining the balance of cholesterol metabolism in the body. It also plays an important role in some metabolic diseases such as nonalcoholic fatty liver disease, obesity, and hypercholesterolemia. Therefore, it is necessary to elucidate the molecular pathological mechanism of NPC1L1 in the regulation of cholesterol metabolism and the occurrence and development of NASH, which can provide a target for the development of novel drugs for the treatment of NASH and other diseases. More importantly, it helps to accelerate the development of drugs that regulate lipid metabolism at multiple levels and reduce liver steatosis, which is extremely important for the prevention and treatment of NASH and related severe metabolic diseases.
Collapse
Affiliation(s)
- ChongLi Xu
- College
of Medical Technology, Chongqing Medical
and Pharmaceutical College, 82 Daxuecheng Road, Chongqing 401331, PR China
| | - Fengyang Fu
- College
of Medical Technology, Chongqing Medical
and Pharmaceutical College, 82 Daxuecheng Road, Chongqing 401331, PR China
| | - Yuhan She
- College
of Medical Technology, Chongqing Medical
and Pharmaceutical College, 82 Daxuecheng Road, Chongqing 401331, PR China
| | - ChongBo Xu
- School
of Biology and Agriculture, Shaoguan University, Shaoguan 512005, PR China
| |
Collapse
|
45
|
Yin Y, Wu C, Zhou Y, Zhang M, Mai S, Chen M, Wang HY. Ezetimibe Induces Paraptosis through Niemann-Pick C1-like 1 Inhibition of Mammalian-Target-of-Rapamycin Signaling in Hepatocellular Carcinoma Cells. Genes (Basel) 2023; 15:4. [PMID: 38275586 PMCID: PMC10815321 DOI: 10.3390/genes15010004] [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: 09/05/2023] [Revised: 11/29/2023] [Accepted: 12/06/2023] [Indexed: 01/27/2024] Open
Abstract
Currently, hepatocellular carcinoma (HCC) is characterized by its unfavorable prognosis and resistance to conventional chemotherapy and radiotherapy. Drug repositioning, an approach aimed at identifying novel therapeutic applications for existing drugs, presents a cost-effective strategy for developing new anticancer agents. We explored the anticancer properties of Ezetimibe, a widely used oral lipid-lowering drug, in the context of HCC. Our findings demonstrate that Ezetimibe effectively suppresses HCC cell proliferation through paraptosis, an apoptotic-independent cell death pathway. The examination of HCC cells lines treated with Ezetimibe using light microscopy and transmission electron microscopy (TEM) showed cytoplasmic vacuolation in the perinuclear region. Notably, the nuclear membrane remained intact in both Ezetimibe-treated and untreated HCC cell lines. Probe staining assays confirmed that the cytoplasmic vacuoles originated from dilated endoplasmic reticulum (ER) compartments rather than mitochondria. Furthermore, a dose-dependent accumulation of reactive oxygen species (ROS) was observed in Ezetimibe-treated HCC cell lines. Co-treatment with the general antioxidant NAC attenuated vacuolation and improved cell viability in Ezetimibe-treated HCC cells. Moreover, Ezetimibe induced paraptosis through proteasome activity inhibition and initiation of the unfolded protein response (UPR) in HCC cell lines. In our in vivo experiment, Ezetimibe significantly impeded the growth of HCC tumors. Furthermore, when combined with Sorafenib, Ezetimibe exhibited a synergistic antitumor effect on HCC cell lines. Mechanistically, Ezetimibe induced paraptosis by targeting NPC1L1 to inhibit the PI3K/AKT/mTOR signaling pathway. In conclusion, our study highlights the potential of Ezetimibe as an anticancer agent by triggering paraptosis in HCC cells.
Collapse
Affiliation(s)
- Yuting Yin
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China; (Y.Y.); (C.W.); (Y.Z.); (M.Z.); (S.M.)
- Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Chun Wu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China; (Y.Y.); (C.W.); (Y.Z.); (M.Z.); (S.M.)
- Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Yufeng Zhou
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China; (Y.Y.); (C.W.); (Y.Z.); (M.Z.); (S.M.)
- Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Meiyin Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China; (Y.Y.); (C.W.); (Y.Z.); (M.Z.); (S.M.)
- Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Shijuan Mai
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China; (Y.Y.); (C.W.); (Y.Z.); (M.Z.); (S.M.)
- Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Minshan Chen
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China;
| | - Hui-Yun Wang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China; (Y.Y.); (C.W.); (Y.Z.); (M.Z.); (S.M.)
- Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
46
|
Guariglia M, Saba F, Rosso C, Bugianesi E. Molecular Mechanisms of Curcumin in the Pathogenesis of Metabolic Dysfunction Associated Steatotic Liver Disease. Nutrients 2023; 15:5053. [PMID: 38140312 PMCID: PMC10745597 DOI: 10.3390/nu15245053] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/23/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a multifactorial condition characterized by insulin resistance, oxidative stress, chronic low-grade inflammation, and sometimes fibrosis. To date, no effective pharmacological therapy has been approved for the treatment of metabolic-associated steatohepatitis (MASH), the progressive form of MASLD. Recently, numerous in vitro and in vivo studies have described the efficacy of nutraceutical compounds in the diet has been tested. Among them, curcumin is the most widely used polyphenol in the diet showing potent anti-inflammatory and antifibrotic activities. This review aims to summarize the most important basic studies (in vitro and animal models studies), describing the molecular mechanisms by which curcumin acts in the context of MASLD, providing the rationale for its effective translational use in humans.
Collapse
Affiliation(s)
| | | | - Chiara Rosso
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (M.G.); (F.S.)
| | - Elisabetta Bugianesi
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (M.G.); (F.S.)
| |
Collapse
|
47
|
Katsuki S, Matoba T, Akiyama Y, Yoshida H, Kotani K, Fujii H, Harada-Shiba M, Ishibashi Y, Ishida T, Ishigaki Y, Kabata D, Kihara Y, Kurisu S, Masuda D, Matsuki K, Matsumura T, Mori K, Nakagami T, Nakazato M, Taniuchi S, Ueno H, Yamashita S, Yoshida H, Tsutsui H, Shoji T. Association of Serum Levels of Cholesterol Absorption and Synthesis Markers with the Presence of Cardiovascular Disease: The CACHE Study CVD Analysis. J Atheroscler Thromb 2023; 30:1766-1777. [PMID: 37100627 DOI: 10.5551/jat.64119] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023] Open
Abstract
AIM Serum levels of cholesterol absorption and synthesis markers have been associated with cardiovascular risk in the United States and European countries. In this study, we examined the relevance of these biomarkers and the presence of cardiovascular disease (CVD) in Japanese individuals. METHODS The CACHE consortium, comprising of 13 research groups in Japan possessing data on campesterol, an absorption marker, and lathosterol, a synthesis marker measured by gas chromatography, compiled the clinical data using the REDCap system. RESULTS Among the 2,944 individuals in the CACHE population, those with missing campesterol or lathosterol data were excluded. This cross-sectional study was able to analyze data from 2,895 individuals, including 339 coronary artery disease (CAD) patients, 108 cerebrovascular disease (CeVD) patients, and 88 peripheral artery disease (PAD) patients. The median age was 57 years, 43% were female, and the median low-density lipoprotein cholesterol and triglyceride levels were 118 mg/dL and 98 mg/dL, respectively. We assessed the associations of campesterol, lathosterol, and the ratio of campesterol to lathosterol (Campe/Latho ratio) with the odds of CVD using multivariable-adjusted nonlinear regression models. The prevalence of CVD, especially CAD, showed positive, inverse, and positive associations with campesterol, lathosterol, and the Campe/Latho ratio, respectively. These associations remained significant even after excluding individuals using statins and/or ezetimibe. The associations of the cholesterol biomarkers with PAD were determined weaker than those with CAD. Contrarily, no significant association was noted between cholesterol metabolism biomarkers and CeVD. CONCLUSION This study showed that both high cholesterol absorption and low cholesterol synthesis biomarker levels were associated with high odds of CVD, especially CAD.
Collapse
Affiliation(s)
- Shunsuke Katsuki
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University
| | - Tetsuya Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University
| | - Yusuke Akiyama
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University
| | - Hiroshi Yoshida
- Department of Laboratory Medicine, The Jikei University Kashiwa Hospital
| | - Kazuhiko Kotani
- Division of Community and Family Medicine, Jichi Medical University
| | - Hisako Fujii
- Department of Health and Medical Innovation, Osaka Metropolitan University Graduate School of Medicine
| | - Mariko Harada-Shiba
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute
| | - Yutaka Ishibashi
- Department of General Medicine, Shimane University Faculty of Medicine
- Jinjyukai Education & Training Center for Healthcare Professionals
| | - Tatsuro Ishida
- Division of Cardiovascular Medicine, Kobe University Graduate School of Medicine
| | - Yasushi Ishigaki
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University
| | - Daijiro Kabata
- Department of Medical Statistics, Osaka Metropolitan University Graduate School of Medicine
| | - Yasuki Kihara
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | - Satoshi Kurisu
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University
| | | | - Kota Matsuki
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute
- Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine
| | - Takeshi Matsumura
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University
| | - Kenta Mori
- Department of General Internal Medicine, Kobe University Hospital
| | - Tomoko Nakagami
- Division of Diabetology and Metabolism, Department of Internal Medicine, Tokyo Women fs Medical University School of Medicine
| | - Masamitsu Nakazato
- Division of Neurology, Respirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki
| | - Satsuki Taniuchi
- Department of Medical Statistics, Osaka Metropolitan University Graduate School of Medicine
| | - Hiroaki Ueno
- Division of Neurology, Respirology, Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki
| | | | - Hisako Yoshida
- Department of Medical Statistics, Osaka Metropolitan University Graduate School of Medicine
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University
| | - Tetsuo Shoji
- Department of Vascular Medicine, Osaka Metropolitan University Graduate School of Medicine
- Vascular Science Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine
| |
Collapse
|
48
|
Cesar-Silva D, Pereira-Dutra FS, Giannini ALM, Maya-Monteiro CM, de Almeida CJG. Lipid compartments and lipid metabolism as therapeutic targets against coronavirus. Front Immunol 2023; 14:1268854. [PMID: 38106410 PMCID: PMC10722172 DOI: 10.3389/fimmu.2023.1268854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/24/2023] [Indexed: 12/19/2023] Open
Abstract
Lipids perform a series of cellular functions, establishing cell and organelles' boundaries, organizing signaling platforms, and creating compartments where specific reactions occur. Moreover, lipids store energy and act as secondary messengers whose distribution is tightly regulated. Disruption of lipid metabolism is associated with many diseases, including those caused by viruses. In this scenario, lipids can favor virus replication and are not solely used as pathogens' energy source. In contrast, cells can counteract viruses using lipids as weapons. In this review, we discuss the available data on how coronaviruses profit from cellular lipid compartments and why targeting lipid metabolism may be a powerful strategy to fight these cellular parasites. We also provide a formidable collection of data on the pharmacological approaches targeting lipid metabolism to impair and treat coronavirus infection.
Collapse
Affiliation(s)
- Daniella Cesar-Silva
- Laboratory of Immunopharmacology, Department of Genetics, Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Filipe S. Pereira-Dutra
- Laboratory of Immunopharmacology, Department of Genetics, Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Ana Lucia Moraes Giannini
- Laboratory of Functional Genomics and Signal Transduction, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clarissa M. Maya-Monteiro
- Laboratory of Immunopharmacology, Department of Genetics, Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- Laboratory of Endocrinology and Department of Endocrinology and Metabolism, Amsterdam University Medical Centers (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Cecília Jacques G. de Almeida
- Laboratory of Immunopharmacology, Department of Genetics, Oswaldo Cruz Institute, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
49
|
Liu X, Lv Z, Xie Z, Wang Q, Yao W, Yu J, Jing Q, Meng X, Ma B, Xue D, Hao C. Association between the use of lipid-lowering drugs and the risk of inflammatory bowel disease. Eur J Clin Invest 2023; 53:e14067. [PMID: 37515404 DOI: 10.1111/eci.14067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/13/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Observational studies have suggested an association between lipid-lowering drugs and inflammatory bowel disease (IBD) risk. This study aimed to assess the causal influence of lipid-lowering agents on IBD risk using Mendelian randomization analysis. METHOD In a population of 173,082 individuals of European ancestry, 55 single-nucleotide polymorphisms were identified as instrumental variables for 6 lipid-lowering drug targets (HMGCR, NPC1LC, PCSK9, LDLR, CETP and APOB). Summary statistics for the genome-wide association study of IBD, ulcerative colitis (UC) and Crohn's disease (CD) were obtained from the FinnGen consortium, Program in Complex Trait Genomics and UK Biobank. Inverse-variance weighted was employed as the primary MR method, and odds ratios (ORs) with 95% confidence intervals were reported as the results. Sensitivity analyses using conventional MR methods were conducted to assess result robustness. RESULTS Gene-proxied inhibition of Niemann-Pick C1-like 1 (NPC1L1) was associated with an increased IBD risk (OR [95% CI]: 2.31 [1.38, 3.85]; p = .001), particularly in UC (OR [95% CI]: 2.40 [1.21, 4.74], p = .012), but not in CD. This finding was replicated in the validation cohort. Additionally, gene-proxied inhibition of low-density lipoprotein receptor was associated with reduced IBD (OR [95% CI]: .72 [.60, .87], p < .001) and UC risk (OR [95% CI]: .74 [.59, .92], p = .006), although this result was not replicated in the validation cohort. Other drug targets did not show significant associations with IBD, UC or CD risk. CONCLUSION Inhibition of the lipid-lowering drug-target NPC1L1 leads to an increased IBD risk, mainly in the UC population.
Collapse
Affiliation(s)
- Xuxu Liu
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhenyi Lv
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhihong Xie
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiang Wang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenchao Yao
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingjing Yu
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qingxu Jing
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xianzhi Meng
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Biao Ma
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongbo Xue
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chenjun Hao
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
50
|
He Z, Zhang Z, Xu P, Dirsch VM, Wang L, Wang K. Laminarin Reduces Cholesterol Uptake and NPC1L1 Protein Expression in High-Fat Diet (HFD)-Fed Mice. Mar Drugs 2023; 21:624. [PMID: 38132943 PMCID: PMC10744832 DOI: 10.3390/md21120624] [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: 10/27/2023] [Revised: 11/27/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Aberrantly high dietary cholesterol intake and intestinal cholesterol uptake lead to dyslipidemia, one of the risk factors for cardiovascular diseases (CVDs). Based on previous studies, laminarin, a polysaccharide found in brown algae, has hypolipidemic activity, but its underlying mechanism has not been elucidated. In this study, we investigated the effect of laminarin on intestinal cholesterol uptake in vitro, as well as the lipid and morphological parameters in an in vivo model of high-fat diet (HFD)-fed mice, and addressed the question of whether Niemann-Pick C1-like 1 protein (NPC1L1), a key transporter mediating dietary cholesterol uptake, is involved in the mechanistic action of laminarin. In in vitro studies, BODIPY-cholesterol-labeled Caco-2 cells were examined using confocal microscopy and a fluorescence reader. The results demonstrated that laminarin inhibited cholesterol uptake into Caco-2 cells in a concentration-dependent manner (EC50 = 20.69 μM). In HFD-fed C57BL/6J mice, laminarin significantly reduced the serum levels of total cholesterol (TC), total triglycerides (TG), and low-density lipoprotein cholesterol (LDL-C). It also decreased hepatic levels of TC, TG, and total bile acids (TBA) while promoting the excretion of fecal cholesterol. Furthermore, laminarin significantly reduced local villous damage in the jejunum of HFD mice. Mechanistic studies revealed that laminarin significantly downregulated NPC1L1 protein expression in the jejunum of HFD-fed mice. The siRNA-mediated knockdown of NPC1L1 attenuated the laminarin-mediated inhibition of cholesterol uptake in Caco-2 cells. This study suggests that laminarin significantly improves dyslipidemia in HFD-fed mice, likely by reducing cholesterol uptake through a mechanism that involves the downregulation of NPC1L1 expression.
Collapse
Affiliation(s)
- Zhuoqian He
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao 266073, China; (Z.H.); (P.X.); (K.W.)
| | - Zhongyin Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University Medical College, Qingdao 266073, China;
| | - Pengfei Xu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao 266073, China; (Z.H.); (P.X.); (K.W.)
| | - Verena M. Dirsch
- Department of Pharmaceutical Sciences, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria;
| | - Limei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao 266073, China; (Z.H.); (P.X.); (K.W.)
- Institute of Innovative Drugs, Qingdao University, Qingdao 266071, China
| | - Kewei Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao 266073, China; (Z.H.); (P.X.); (K.W.)
- Institute of Innovative Drugs, Qingdao University, Qingdao 266071, China
| |
Collapse
|