1
|
Chen CH, Sawamura T, Akhmedov A, Tsai MH, Akyol O, Kakino A, Chiang HH, Kraler S, Lüscher TF. Evolving concepts of low-density lipoprotein: From structure to function. Eur J Clin Invest 2025; 55:e70019. [PMID: 40045739 DOI: 10.1111/eci.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/17/2025] [Indexed: 04/23/2025]
Abstract
BACKGROUND Low-density lipoprotein (LDL) is a central player in atherogenesis and has long been referred to as 'bad cholesterol.' However, emerging evidence indicates that LDL functions in multifaceted ways beyond cholesterol transport that include roles in inflammation, immunity, and cellular signaling. Understanding LDL's structure, metabolism and function is essential for advancing cardiovascular disease research and therapeutic strategies. METHODS This narrative review examines the history, structural properties, metabolism and functions of LDL in cardiovascular health and disease. We analyze key milestones in LDL research, from its early identification to recent advancements in molecular biology and omics-based investigations. Structural and functional insights are explored through imaging, proteomic analyses and lipidomic profiling, providing a deeper understanding of LDL heterogeneity. RESULTS Low-density lipoprotein metabolism, from biosynthesis to receptor-mediated clearance, plays a crucial role in lipid homeostasis and atherogenesis. Beyond cholesterol transport, LDL contributes to plaque inflammation, modulates adaptive immunity and regulates cellular signaling pathways. Structural studies reveal its heterogeneous composition, which influences its pathogenic potential. Evolving perspectives on LDL redefine its clinical significance, affecting cardiovascular risk assessment and therapeutic interventions. CONCLUSIONS A holistic understanding of LDL biology challenges traditional perspectives and underscores its complexity in cardiovascular health. Future research should focus on further elucidating LDL's structural and functional diversity to refine risk prediction models and therapeutic strategies, ultimately improving cardiovascular outcomes.
Collapse
Affiliation(s)
- Chu-Huang Chen
- Molecular Cardiology Research Laboratories, Vascular and Medicinal Research, The Texas Heart Institute, Houston, Texas, USA
| | - Tatsuya Sawamura
- Department of Molecular Pathophysiology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Ming-Hsien Tsai
- Department of Child Care, College of Humanities and Social Sciences, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Omer Akyol
- Molecular Cardiology Research Laboratories, Vascular and Medicinal Research, The Texas Heart Institute, Houston, Texas, USA
| | - Akemi Kakino
- Department of Molecular Pathophysiology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Huan-Hsing Chiang
- Molecular Cardiology Research Laboratories, Vascular and Medicinal Research, The Texas Heart Institute, Houston, Texas, USA
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
- Department of Internal Medicine and Cardiology, Cantonal Hospital Baden, Baden, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
- Heart Division, Royal Brompton and Harefield Hospitals, GSTT and King's College, London, UK
| |
Collapse
|
2
|
Du L, Gao R, Chen Z. 5-Methylcytosine Methylation-Linked Hippo Pathway Molecular Interactions Regulate Lipid Metabolism. Int J Mol Sci 2025; 26:2560. [PMID: 40141201 PMCID: PMC11942534 DOI: 10.3390/ijms26062560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
5-methylcytosine (5mC) is a common form of DNA methylation, essentially acting as an epigenetic modification that regulates gene expression by affecting the binding of transcription factors to DNA or by recruiting proteins that make it difficult to recognize and transcribe genes. 5mC methylation is present in eukaryotes in a variety of places, such as in CpG islands, within gene bodies, and in regions of repetitive sequences, whereas in prokaryotic organisms, it is mainly present in genomic DNA. The Hippo pathway is a highly conserved signal transduction pathway, which is extremely important in cell proliferation and death, controlling the size of tissues and organs and regulating cell differentiation, in addition to its important regulatory roles in lipid synthesis, transport, and catabolism. Lipid metabolism is an important part of various metabolic pathways in the human body, and problems in lipid metabolism are related to abnormalities in key enzymes, related proteins, epigenetic inheritance, and certain specific amino acids, which are the key factors affecting its proper regulation. In this article, we will introduce the molecular mechanisms of 5mC methylation and the Hippo signaling pathway, and the possibility of their co-regulation of lipid metabolism, with the aim of providing new ideas for further research and novel therapeutic modalities for lipid metabolism and a reference for the development and exploration of related research.
Collapse
Affiliation(s)
- Lichen Du
- Agricultural College, Yangzhou University, Yangzhou 225009, China;
| | - Rui Gao
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Zhi Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
3
|
Ghiasvand T, Karimi J, Khodadadi I, Yazdi A, Khazaei S, Kichi ZA, Hosseini SK. The interplay of LDLR, PCSK9, and lncRNA- LASER genes expression in coronary artery disease: Implications for therapeutic interventions. Prostaglandins Other Lipid Mediat 2025; 177:106969. [PMID: 40020908 DOI: 10.1016/j.prostaglandins.2025.106969] [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: 10/05/2024] [Revised: 02/03/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND AND PURPOSE Coronary artery disease (CAD) is defined as stenosis of coronary arteries due to atherosclerosis. The etiology of atherosclerosis can be attributed to a disruption in lipid metabolism, specifically cholesterol and low-density lipoprotein cholesterol (LDL-C). PCSK9 is an enzyme that controls the metabolism of LDL-C by degrading the low-density lipoprotein receptor (LDLR), which in turn affects the metabolism of LDL-C. A newly discovered Long Non-coding RNA named LASER, which affects the homeostasis of cholesterol, has been identified through the evaluation of bioinformatics. The objective of this study was to assess the levels of gene expression related to cholesterol balance, specifically LDLR, PCSK9, and LASER, in peripheral blood mononuclear cells (PBMCs) of Iranian CAD patients in comparison to controls. EXPERIMENTAL APPROACH This case-control study included 49 CAD patients, with 81.63 % receiving statins, compared to 40 control subjects, of whom 40 % received statins. The qRT-PCR was used to analyze the expression levels of LDLR, PCSK9, and LASER in PBMCs. Additionally, the ELISA method was employed to determine the blood concentration of PCSK9. FINDINGS / RESULTS CAD patients demonstrated a significant reduction in PBMC gene expression levels of LDLR (P < 0.01) and a significant rise in gene expression of PCSK9 and LASER, as well as blood concentration of PCSK9 (P < 0.05) compared to controls. The gene expression of PCSK9 showed a strong positive relationship with LDLR expression in patients (P = 0.0003). Furthermore, a strong correlation was seen between PCSK9 and LASER, as well as LASER and LDLR expression (P < 0.0001) in two groups. CONCLUSION AND IMPLICATIONS PCSK9 and LASER are potential therapeutic targets for atherosclerosis-related disorders, including CAD. Given that patients receiving statins were twice that of the control subjects, and the effect of statins on the LDLR, PCSK9 and LASER, further research is required to delineate the distinct effects of coronary artery disease conditions and statin usage on the expression of the aforementioned genes.
Collapse
Affiliation(s)
- Tayebe Ghiasvand
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Jamshid Karimi
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Khodadadi
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amirhossein Yazdi
- Department of Cardiology, Faculty of Medicine, Clinical Research Development Unit of Farshchian Hospital, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Salman Khazaei
- Research Center for Health Sciences, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Zahra Abedi Kichi
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilian University, Munich 80336, Germany
| | - Seyed Kianoosh Hosseini
- Department of Cardiology, Faculty of Medicine, Clinical Research Development Unit of Farshchian Hospital, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
4
|
Moolsup F, Suttithumsatid W, Woonnoi W, Chonpathompikunlert P, Tanasawet S, Sukketsiri W. Passion Fruit Seed Extract Attenuates Hepatic Steatosis in Oleic Acid-Treated HepG2 Cells through Modulation of ERK1/2 and Akt Signaling Pathways. Cell Biochem Biophys 2025:10.1007/s12013-025-01706-5. [PMID: 40025286 DOI: 10.1007/s12013-025-01706-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2025] [Indexed: 03/04/2025]
Abstract
Hepatic steatosis, commonly referred to as fatty liver disease, is defined by the abnormal buildup of fat within liver cells. Currently, primary treatments mainly focus on lifestyle changes, underscoring a lack of direct pharmacological options. Passion fruit seed extract (PFSE) has been reported to decrease hepatosteatosis; however, the mechanism underlying this effect has not been clarified. Therefore, the objective of this research was to investigate the effects and mechanisms of action of PFSE against oleic acid (OA)-induced hepatosteatosis in HepG2 cells. OA-induced HepG2 cells were analyzed by using various cell-based experiments, including assessments of cytotoxicity, reactive oxygen species (ROS) production, apoptosis, and protein and gene expression. LC-MS-MS analysis showed that PFSE contains a variety of phytochemical compounds such as alkaloids, flavonoids, stilbenoids, coumarins, terpenoids, lipids, and fatty acid derivatives, which have the potential to exhibit various pharmacological activities. In this study, PFSE demonstrated antioxidant, anti-inflammatory, and lipid metabolism-regulating activities. It also influenced key genes related to lipid metabolism, including SREBP-1c, ACC, FASN, PPARα, CPT-1A, LPL, SCD1, and LDLR. The positive effects of PFSE on OA-induced hepatic steatosis in HepG2 cells were modulated through the Akt and ERK signaling pathways, suggesting that PFSE may offer a comprehensive approach to managing hepatic steatosis.
Collapse
Affiliation(s)
- Furoida Moolsup
- Laboratory Animal Service Center, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
| | - Wiwit Suttithumsatid
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla, Thailand
- Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai, Thailand
| | - Wanwipha Woonnoi
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Pennapa Chonpathompikunlert
- Research and Development Group for Bio-Industries, Thailand Institute of Scientific and Technological Research (TISTR), Pathumthani, Thailand
| | - Supita Tanasawet
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Wanida Sukketsiri
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand.
| |
Collapse
|
5
|
Chen S, Li J, Liang Y, Zhang M, Qiu Z, Liu S, Wang H, Zhu Y, Song S, Hou X, Liu C, Wu Q, Zhu M, Shen W, Miao J, Hou FF, Liu Y, Wang C, Zhou L. β-catenin-inhibited Sumoylation modification of LKB1 and fatty acid metabolism is critical in renal fibrosis. Cell Death Dis 2024; 15:769. [PMID: 39438470 PMCID: PMC11496881 DOI: 10.1038/s41419-024-07154-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
Liver kinase B1 (LKB1) is a serine/threonine kinase controlling cell homeostasis. Among post-translational modification, Sumoylation is vital for LKB1 activating adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK), the key regulator in energy metabolism. Of note, AMPK-regulated fatty acid metabolism is highly involved in maintaining normal renal function. However, the regulative mechanisms of LKB1 Sumoylation remain elusive. In this study, we demonstrated that β-catenin, a notorious signal in renal fibrosis, inhibited the Sumoylation of LKB1, thereby disrupting fatty acid oxidation in renal tubular cells and triggering renal fibrosis. Mechanically, we found that Sumo3 was the key mediator for LKB1 Sumoylation in renal tubular cells, which was transcriptionally inhibited by β-catenin/Transcription factor 4 (TCF4) signaling. Overexpression of Sumo3, not Sumo1 or Sumo2, restored β-catenin-disrupted fatty acid metabolism, and retarded lipid accumulation and fibrogenesis in the kidney. In vivo, conditional knockout of β-catenin in tubular cells effectively preserved fatty acid oxidation and blocked lipid accumulation by maintaining LKB1 Sumoylation and AMPK activation. Furthermore, ectopic expression of Sumo3 strongly inhibited Wnt1-aggravated lipid accumulation and fibrogenesis in unilateral ischemia-reperfusion mice. In patients with chronic kidney disease, we found a loss of Sumo3 expression, and it was highly related to LKB1 repression. This contributes to fatty acid metabolism disruption and lipid accumulation, resulting in renal fibrosis. Overall, our study revealed a new mechanism in fatty acid metabolism dysfunction and provided a new therapeutic target pathway for regulating Sumo modification in renal fibrosis.
Collapse
Affiliation(s)
- Shuangqin Chen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Division of Nephrology, Department of Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Jiemei Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Liang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Meijia Zhang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ziqi Qiu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Sirui Liu
- Division of Nephrology, Department of Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - HaoRan Wang
- Walter Johnson High School, Bethesda, MD, USA
| | - Ye Zhu
- Division of Nephrology, Department of Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Shicong Song
- Division of Nephrology, Department of Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Xiaotao Hou
- Pathology Department, Guangzhou KingMed Center for Clinical Laboratory Co., Ltd, Guangzhou, China
| | - Canzhen Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qinyu Wu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mingsheng Zhu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weiwei Shen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinhua Miao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Fan Hou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Cheng Wang
- Division of Nephrology, Department of Medicine, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China.
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
6
|
Munkhsaikhan U, Ait-Aissa K, Sahyoun AM, Apu EH, Abidi AH, Kassan A, Kassan M. Lomitapide: navigating cardiovascular challenges with innovative therapies. Mol Biol Rep 2024; 51:1082. [PMID: 39432146 DOI: 10.1007/s11033-024-10003-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/07/2024] [Indexed: 10/22/2024]
Abstract
Dyslipidemia is the most significant risk factor for cardiovascular diseases (CVDs) Secondary dyslipidemia: its treatments and association with atherosclerosis. Glob Health Med, Efficacy and safety of saroglitazar for the management of dyslipidemia: A systematic review and meta-analysis of interventional studies. The current treatment strategies for managing dyslipidemia focus on reducing low-density lipoprotein cholesterol (LDL-C) to minimize the risks of atherosclerosis and myocardial infarction (MI). Homozygous Familial Hypercholesterolemia (HoFH) is an inherited autosomal dominant disease caused by a mutation in the LDL receptor (LDLr), which can lead to extremely high levels of LDL-C The Beneficial Effect of Lomitapide on the Cardiovascular System in LDLr(-/-) Mice with Obesity, The microsomal triglyceride transfer protein inhibitor lomitapide improves vascular function in mice with obesity. Although statin therapy has been the primary treatment for dyslipidemia, HoFH patients do not respond well to statins, requiring alternative therapies. Microsomal triglyceride transfer protein (MTP) inhibition has emerged as a potential therapeutic target for treating HoFH. MTP is primarily responsible for transferring triglyceride and other lipids into apolipoprotein B (ApoB) during the assembly of very low-density lipoprotein (VLDL) particles in the liver. Lomitapide, an inhibitor of MTP, has been approved for treatingof HoFH adults. Unlike statins, lomitapide does not act on the LDLr to reduce cholesterol. Instead, lomitapide lowers the levels of ApoB-containing proteins, primarily VLDL, eventually decreasing LDL-C levels. Studies have shown that lomitapide can reduce LDL-C levels by more than 50% in patients with HoFH who have failed to respond adequately to other treatments. Lowering LDL-C levels is important for preventing atherosclerosis, reducing cardiovascular risk, improving endothelial function, and promoting overall cardiovascular health, especially for patients with HoFH Efficacy and safety of a microsomal triglyceride transfer protein inhibitor in patients with homozygous familial hypercholesterolaemia: a single-arm, open-label, phase 3 study. This review paper focuses on research findings regarding the therapeutic benefits of lomitapide, highlighting its effectiveness in lowering cholesterol levels and reducing the risk of CVDs The microsomal triglyceride transfer protein inhibitor lomitapide improves vascular function in mice with obesity.
Collapse
Affiliation(s)
- Undral Munkhsaikhan
- College of Dental Medicine, Lincoln Memorial University, LMU tower, 1705 St. Mary Street, Knoxville, TN, 37917, USA
| | - Karima Ait-Aissa
- College of Dental Medicine, Lincoln Memorial University, LMU tower, 1705 St. Mary Street, Knoxville, TN, 37917, USA
| | - Amal M Sahyoun
- College of Dental Medicine, Lincoln Memorial University, LMU tower, 1705 St. Mary Street, Knoxville, TN, 37917, USA
| | - Ehsanul Hoque Apu
- College of Dental Medicine, Lincoln Memorial University, LMU tower, 1705 St. Mary Street, Knoxville, TN, 37917, USA
| | - Ammaar H Abidi
- College of Dental Medicine, Lincoln Memorial University, LMU tower, 1705 St. Mary Street, Knoxville, TN, 37917, USA
| | - Adam Kassan
- School of Pharmacy, West Coast University, 590 N. Vermont Ave, Los Angeles, CA, 90004, USA.
| | - Modar Kassan
- College of Dental Medicine, Lincoln Memorial University, LMU tower, 1705 St. Mary Street, Knoxville, TN, 37917, USA.
| |
Collapse
|
7
|
Zhang YY, Li YJ, Xue CD, Li S, Gao ZN, Qin KR. Effects of T2DM on cancer progression: pivotal precipitating factors and underlying mechanisms. Front Endocrinol (Lausanne) 2024; 15:1396022. [PMID: 39290325 PMCID: PMC11405243 DOI: 10.3389/fendo.2024.1396022] [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: 03/05/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder affecting people worldwide. It is characterized by several key features, including hyperinsulinemia, hyperglycemia, hyperlipidemia, and dysbiosis. Epidemiologic studies have shown that T2DM is closely associated with the development and progression of cancer. T2DM-related hyperinsulinemia, hyperglycemia, and hyperlipidemia contribute to cancer progression through complex signaling pathways. These factors increase drug resistance, apoptosis resistance, and the migration, invasion, and proliferation of cancer cells. Here, we will focus on the role of hyperinsulinemia, hyperglycemia, and hyperlipidemia associated with T2DM in cancer development. Additionally, we will elucidate the potential molecular mechanisms underlying their effects on cancer progression. We aim to identify potential therapeutic targets for T2DM-related malignancies and explore relevant directions for future investigation.
Collapse
Affiliation(s)
- Yu-Yuan Zhang
- Institute of Cardio-Cerebrovascular Medicine, Central Hospital of Dalian University of Technology, Dalian, Liaoning, China
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning, China
| | - Yong-Jiang Li
- Institute of Cardio-Cerebrovascular Medicine, Central Hospital of Dalian University of Technology, Dalian, Liaoning, China
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning, China
| | - Chun-Dong Xue
- Institute of Cardio-Cerebrovascular Medicine, Central Hospital of Dalian University of Technology, Dalian, Liaoning, China
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning, China
| | - Shen Li
- Institute of Cardio-Cerebrovascular Medicine, Central Hospital of Dalian University of Technology, Dalian, Liaoning, China
| | - Zheng-Nan Gao
- Institute of Cardio-Cerebrovascular Medicine, Central Hospital of Dalian University of Technology, Dalian, Liaoning, China
| | - Kai-Rong Qin
- Institute of Cardio-Cerebrovascular Medicine, Central Hospital of Dalian University of Technology, Dalian, Liaoning, China
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning, China
| |
Collapse
|
8
|
Cao Y, Yang Y, Liang Z, Guo W, Lv X, Ni L, Chen Y. Synthesis of Ganoderic Acids Loaded Zein-Chitosan Nanoparticles and Evaluation of Their Hepatoprotective Effect on Mice Given Excessive Alcohol. Foods 2024; 13:2760. [PMID: 39272525 PMCID: PMC11394847 DOI: 10.3390/foods13172760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/24/2024] [Accepted: 08/25/2024] [Indexed: 09/15/2024] Open
Abstract
Ganoderma lucidum, used in East Asia for its health benefits, contains ganoderic acids (GA) which have various pharmacological activities but are limited by poor water solubility and low oral bioaccessibility. This study synthesized and characterized ganoderic acids loaded zein-chitosan nanoparticles (GA-NPs), and investigated its advantages in alleviating alcoholic liver injury (ALI) in mice model. The GA-NPs demonstrated high encapsulation efficiency (92.68%), small particle size (177.20 nm), and a +29.53 mV zeta potential. The experimental results of alcohol-induced liver injury mouse model showed that GA-NPs significantly improved liver metabolic function, reduced alcohol-induced liver oxidative stress in liver by decreasing lactate dehydrogenase activity and malondialdehyde level, while increasing the activities of liver antioxidant enzymes and alcohol dehydrogenase. Moreover, GA-NPs were favorable to ameliorate intestinal microbiota dysbiosis in mice exposed to alcohol by increasing the proportion of probiotics such as Romboutsia, Faecalibaculum, Bifidobacterium and Turicibacter, etc., which were highly correlated with the improvement of liver function. Furthermore, GA-NPs modulated the mRNA expression related to ethanol metabolism, oxidative stress and lipid metabolism. Conclusively, this study revealed that GA-NPs have stronger hepatoprotective effects than non-encapsulated ganoderic acids on alleviating ALI by regulating intestinal microbiota and liver metabolism.
Collapse
Affiliation(s)
- Yingjia Cao
- Department of Hepatopancreatobiliary Surgery, Fujian Research Institute of Abdominal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China
- Food Nutrition and Health Research Center, School of Advanced Manufacturing, Fuzhou University, Jinjiang 362200, China
| | - Yuheng Yang
- Department of Hepatopancreatobiliary Surgery, Fujian Research Institute of Abdominal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
- The First Affiliated Hospital of Fujian Medical University, Fujian Medical University, Fuzhou 350004, China
| | - Zihua Liang
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China
- Food Nutrition and Health Research Center, School of Advanced Manufacturing, Fuzhou University, Jinjiang 362200, China
| | - Weiling Guo
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China
- Food Nutrition and Health Research Center, School of Advanced Manufacturing, Fuzhou University, Jinjiang 362200, China
| | - Xucong Lv
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China
- Food Nutrition and Health Research Center, School of Advanced Manufacturing, Fuzhou University, Jinjiang 362200, China
| | - Li Ni
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China
- Food Nutrition and Health Research Center, School of Advanced Manufacturing, Fuzhou University, Jinjiang 362200, China
| | - Youting Chen
- Department of Hepatopancreatobiliary Surgery, Fujian Research Institute of Abdominal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
- Department of Hepatopancreatobiliary Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| |
Collapse
|
9
|
Koppula S, Wankhede NL, Sammeta SS, Shende PV, Pawar RS, Chimthanawala N, Umare MD, Taksande BG, Upaganlawar AB, Umekar MJ, Kopalli SR, Kale MB. Modulation of cholesterol metabolism with Phytoremedies in Alzheimer's disease: A comprehensive review. Ageing Res Rev 2024; 99:102389. [PMID: 38906182 DOI: 10.1016/j.arr.2024.102389] [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: 04/20/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
Alzheimer's disease (AD) is a complex neurological ailment that causes cognitive decline and memory loss. Cholesterol metabolism dysregulation has emerged as a crucial element in AD pathogenesis, contributing to the formation of amyloid-beta (Aβ) plaques and tau tangles, the disease's hallmark neuropathological characteristics. Thus, targeting cholesterol metabolism has gained attention as a potential therapeutic method for Alzheimer's disease. Phytoremedies, which are generated from plants and herbs, have shown promise as an attainable therapeutic option for Alzheimer's disease. These remedies contain bioactive compounds like phytochemicals, flavonoids, and polyphenols, which have demonstrated potential in modulating cholesterol metabolism and related pathways. This comprehensive review explores the modulation of cholesterol metabolism by phytoremedies in AD. It delves into the role of cholesterol in brain function, highlighting disruptions observed in AD. Additionally, it examines the underlying molecular mechanisms of cholesterol-related pathology in AD. The review emphasizes the significance of phytoremedies as a potential therapeutic intervention for AD. It discusses the drawbacks of current treatments and the need for alternative strategies addressing cholesterol dysregulation and its consequences. Through an in-depth analysis of specific phytoremedies, the review presents compelling evidence of their potential benefits. Molecular mechanisms underlying phytoremedy effects on cholesterol metabolism are examined, including regulation of cholesterol-related pathways, interactions with Aβ pathology, influence on tau pathology, and anti-inflammatory effects. The review also highlights challenges and future perspectives, emphasizing standardization, clinical evidence, and personalized medicine approaches to maximize therapeutic potential in AD treatment. Overall, phytoremedies offer promise as a potential avenue for AD management, but further research and collaboration are necessary to fully explore their efficacy, safety, and mechanisms of action.
Collapse
Affiliation(s)
- Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| | - Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Shivkumar S Sammeta
- National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500037, India.
| | - Prajwali V Shende
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Rupali S Pawar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | | | - Mohit D Umare
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India.
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea.
| | - Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| |
Collapse
|
10
|
Tapken I, Kuhn D, Hoffmann N, Detering NT, Schüning T, Billaud JN, Tugendreich S, Schlüter N, Green J, Krämer A, Claus P. From data to discovery: AI-guided analysis of disease-relevant molecules in spinal muscular atrophy (SMA). Hum Mol Genet 2024; 33:1367-1377. [PMID: 38704739 DOI: 10.1093/hmg/ddae076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/04/2024] [Accepted: 04/22/2024] [Indexed: 05/07/2024] Open
Abstract
Spinal Muscular Atrophy is caused by partial loss of survival of motoneuron (SMN) protein expression. The numerous interaction partners and mechanisms influenced by SMN loss result in a complex disease. Current treatments restore SMN protein levels to a certain extent, but do not cure all symptoms. The prolonged survival of patients creates an increasing need for a better understanding of SMA. Although many SMN-protein interactions, dysregulated pathways, and organ phenotypes are known, the connections among them remain largely unexplored. Monogenic diseases are ideal examples for the exploration of cause-and-effect relationships to create a network describing the disease-context. Machine learning tools can utilize such knowledge to analyze similarities between disease-relevant molecules and molecules not described in the disease so far. We used an artificial intelligence-based algorithm to predict new genes of interest. The transcriptional regulation of 8 out of 13 molecules selected from the predicted set were successfully validated in an SMA mouse model. This bioinformatic approach, using the given experimental knowledge for relevance predictions, enhances efficient targeted research in SMA and potentially in other disease settings.
Collapse
Affiliation(s)
- Ines Tapken
- SMATHERIA gGmbH - Non-Profit Biomedical Research Institute, Feodor-Lynen-Str. 31, Hannover 30625, Germany
- Center for Systems Neuroscience (ZSN), Bünteweg 2, Hannover 30559, Germany
| | - Daniela Kuhn
- SMATHERIA gGmbH - Non-Profit Biomedical Research Institute, Feodor-Lynen-Str. 31, Hannover 30625, Germany
- Hannover Medical School, Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Nico Hoffmann
- SMATHERIA gGmbH - Non-Profit Biomedical Research Institute, Feodor-Lynen-Str. 31, Hannover 30625, Germany
| | - Nora T Detering
- SMATHERIA gGmbH - Non-Profit Biomedical Research Institute, Feodor-Lynen-Str. 31, Hannover 30625, Germany
- Center for Systems Neuroscience (ZSN), Bünteweg 2, Hannover 30559, Germany
| | - Tobias Schüning
- SMATHERIA gGmbH - Non-Profit Biomedical Research Institute, Feodor-Lynen-Str. 31, Hannover 30625, Germany
| | - Jean-Noël Billaud
- QIAGEN Digital Insights, 1001 Marshall Street,Redwood City, CA 94063, United States
| | - Stuart Tugendreich
- QIAGEN Digital Insights, 1001 Marshall Street,Redwood City, CA 94063, United States
| | - Nadine Schlüter
- Hannover Medical School, Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Jeff Green
- QIAGEN Digital Insights, 1001 Marshall Street,Redwood City, CA 94063, United States
| | - Andreas Krämer
- QIAGEN Digital Insights, 1001 Marshall Street,Redwood City, CA 94063, United States
| | - Peter Claus
- SMATHERIA gGmbH - Non-Profit Biomedical Research Institute, Feodor-Lynen-Str. 31, Hannover 30625, Germany
- Center for Systems Neuroscience (ZSN), Bünteweg 2, Hannover 30559, Germany
| |
Collapse
|
11
|
Chen SK, Wei WX, Huang FY, Wang J, Li XY, Yang YT, Xing WT, Gao F, Li M, Miao F, Chen LL, Wei PF. Research on the mechanism of sea buckthorn leaf Fu tea in the treatment of hyperlipidemia. Heliyon 2024; 10:e32343. [PMID: 38984297 PMCID: PMC11231531 DOI: 10.1016/j.heliyon.2024.e32343] [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: 01/27/2024] [Revised: 05/31/2024] [Accepted: 06/02/2024] [Indexed: 07/11/2024] Open
Abstract
Background Hyperlipidemia (HLP) presents a significant challenge to global public health. Mounting evidence suggests that statins, the recommended first-line lipid-lowering agents, have significant adverse effects. Consequently, the quest for natural and efficacious alternative therapies is steadily emerging as a research priority for HLP prevention and treatment. Consumption of tea, which is rich in diverse biologically active compounds with the capacity to regulate lipid metabolism and combat obesity, has emerged as a promising alternative therapy. Sea buckthorn leaves are rich in a multitude of biologically active substances, have a hypolipidemic effect, and can be used as a raw material for tea because of their unique flavor. There is a suggestion that combining Aspergillus cristatus with tea could modify or boost the lipid-lowering active compounds present in tea, thereby increasing its efficacy in regulating lipid metabolism. Results Sea Buckthorn Leaf Fu Tea (SBLFT) was obtained by fermentation when sea buckthorn leaves contained 42 % moisture, inoculated with Aspergillus cristatus 0.2 mL/g, and incubated for 8 d at constant temperature. Animal experiments demonstrated that SBLFT significantly inhibited body weight gain in HLP rats and reduced lipid content and serum oxidative stress. In addition, liver tissue sections and functional indices showed that SBLFT can improve liver morphology and function abnormalities. Reverse transcription-polymerase chain reaction results indicated that the expression of Liver kinase B1 (LKB1), adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK), acetyl CoA carboxylase 1 (ACC1), and sterol-regulatory element binding protein-1 (SREBP1c) gene related to lipid metabolism was altered. Conclusion SBLFT improved HLP, specifically via promoting the expression of LKB1 in the liver of HLP rats, activating AMPK, and inhibiting ACC1 and SREBP1c expression, resulting in the inhibition of fatty acid and triglyceride synthesis-related enzymes at the transcriptional level.
Collapse
Affiliation(s)
- Si-Kai Chen
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Wen-Xin Wei
- Nanchang University, Nanchang, 330029, China
| | - Feng-Yu Huang
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Jing Wang
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Xing-Yu Li
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yu-Ting Yang
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Wan-Tao Xing
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Feng Gao
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Min Li
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Feng Miao
- The Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, China
| | - Liang-Liang Chen
- The Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, China
| | - Pei-Feng Wei
- Shaanxi University of Chinese Medicine, Xianyang, 712046, China
- The Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, China
| |
Collapse
|
12
|
Bai X, Wang S, Shu L, Cao Q, Hu H, Zhu Y, Chen C. Hawthorn leaf flavonoids alleviate the deterioration of atherosclerosis by inhibiting SCAP-SREBP2-LDLR pathway through sPLA2-ⅡA signaling in macrophages in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:118006. [PMID: 38442806 DOI: 10.1016/j.jep.2024.118006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/23/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hawthorn leaves are a combination of the dried leaves of the Rosaceae plants, i.e., Crataegus pinnatifida Bge. or Crataegus pinnatifida Bge. var. major N. E. Br., is primarily cultivated in East Asia, North America, and Europe. hawthorn leaf flavonoids (HLF) are the main part of extraction. The HLF have demonstrated potential in preventing hypertension, inflammation, hyperlipidemia, and atherosclerosis. However, the potential pharmacological mechanism behind its anti-atherosclerotic effect has yet to be explored. AIM OF THE STUDY The in vivo and in vitro effects of HLF on lipid-mediated foam cell formation were investigated, with a specific focus on the levels of secreted phospholipase A2 type IIA (sPLA2-II A) in macrophage cells. MATERIALS AND METHODS The primary constituents of HLF were analyzed using ultra-high performance liquid chromatography and liquid chromatography-tandem mass spectrometry. In vivo, HLF, at concentrations of 5 mg/kg, 20 mg/kg, and 40 mg/kg, were administered to apolipoprotein E knockout mice (ApoE-/-) fed by high-fat diet (HFD) for 16 weeks. Aorta and serum samples were collected to identify lesion areas and lipids through mass spectrometry analysis to dissect the pathological process. RAW264.7 cells were incubated with oxidized low-density lipoprotein (ox-LDL) alone, or ox-LDL combined with different doses of HLF (100, 50, and 25 μg/ml), or ox-LDL plus 24-h sPLA2-IIA inhibitors, for cell biology analysis. Lipids and inflammatory cytokines were detected using biochemical analyzers and ELISA, while plaque size and collagen content of plaque were assessed by HE and the Masson staining of the aorta. The lipid deposition in macrophages was observed by Oil Red O staining. The expression of sPLA2-IIA and SCAP-SREBP2-LDLR was determined by RT-qPCR and Western blot analysis. RESULTS The chemical profile of HLF was studied using UPLC-Q-TOF-MS/MS, allowing the tentative identification of 20 compounds, comprising 1 phenolic acid, 9 flavonols and 10 flavones, including isovitexin, vitexin-4″-O-glucoside, quercetin-3-O-robibioside, rutin, vitexin-2″-O-rhamnoside, quercetin, etc. HLF decreased total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), and non-high-density lipoprotein cholesterol (non-HDL-C) levels in ApoE-/- mice (P < 0.05), reduced ox-LDL uptake, inhibited level of inflammatory factors, such as IL-6, IL-8, TNF-α, and IL-1ꞵ (P < 0.001), and alleviated aortic plaques with a thicker fibrous cap. HLF effectively attenuated foam cell formation in ox-LDL-treated RAW264.7 macrophages, and reduced levels of intracellular TC, free cholesterol (FC), cholesteryl ester (CE), IL-6, TNF-α, and IL-1β (P < 0.001). In both in vivo and in vitro experiments, HLF significantly downregulated the expression of sPLA2-IIA, SCAP, SREBP2, LDLR, HMGCR, and LOX-1 (P < 0.05). Furthermore, sPLA2-IIA inhibitor effectively mitigated inflammatory release in RAW264.7 macrophages and regulated SCAP-SREBP2-LDLR signaling pathway by inhibiting sPLA2-IIA secretion (P < 0.05). CONCLUSION HLF exerted a protective effect against atherosclerosis through inhibiting sPLA2-IIA to diminish SCAP-SREBP2-LDLR signaling pathway, to reduce LDL uptake caused foam cell formation, and to slow down the progression of atherosclerosis in mice.
Collapse
Affiliation(s)
- Xufeng Bai
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Shuwen Wang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Limei Shu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Qingyu Cao
- College of Pharmacy, Nanchang Medical College, Nanchang, Jiangxi, 330052, China
| | - Huiming Hu
- College of Pharmacy, Nanchang Medical College, Nanchang, Jiangxi, 330052, China; Key Laboratory of Pharmacodynamics and Quality Evaluation on Anti-Inflammatory Chinese Herbs, Jiangxi Administration of Traditional Chinese Medicine, Jiangxi, 330052, China; School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Queensland, 4072, Australia.
| | - Yanchen Zhu
- College of Computer Science, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Queensland, 4072, Australia.
| |
Collapse
|
13
|
Lin Y, Huang H, Cao J, Zhang K, Chen R, Jiang J, Yi X, Feng S, Liu J, Zheng S, Ling Q. An integrated proteomics and metabolomics approach to assess graft quality and predict early allograft dysfunction after liver transplantation: a retrospective cohort study. Int J Surg 2024; 110:3480-3494. [PMID: 38502860 PMCID: PMC11175820 DOI: 10.1097/js9.0000000000001292] [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: 09/18/2023] [Accepted: 02/22/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND Early allograft dysfunction (EAD) is a common complication after liver transplantation (LT) and is associated with poor prognosis. Graft itself plays a major role in the development of EAD. We aimed to reveal the EAD-specific molecular profiles to assess graft quality and establish EAD predictive models. METHODS A total of 223 patients who underwent LT were enrolled and divided into training ( n =73) and validation ( n =150) sets. In the training set, proteomics was performed on graft biopsies, together with metabolomics on paired perfusates. Differential expression, enrichment analysis, and protein-protein interaction network were used to identify the key molecules and pathways involved. EAD predictive models were constructed using machine learning and verified in the validation set. RESULTS A total of 335 proteins were differentially expressed between the EAD and non-EAD groups. These proteins were significantly enriched in triglyceride and glycerophospholipid metabolism, neutrophil degranulation, and the MET-related signaling pathway. The top 12 graft proteins involved in the aforementioned processes were identified, including GPAT1, LPIN3, TGFB1, CD59, and SOS1. Moreover, downstream metabolic products, such as lactate dehydrogenase, interleukin-8, triglycerides, and the phosphatidylcholine/phosphorylethanolamine ratio in the paired perfusate displayed a close relationship with the graft proteins. To predict the occurrence of EAD, an integrated model using perfusate metabolic products and clinical parameters showed areas under the curve of 0.915 and 0.833 for the training and validation sets, respectively. It displayed superior predictive efficacy than that of currently existing models, including donor risk index and D-MELD scores. CONCLUSIONS We identified novel biomarkers in both grafts and perfusates that could be used to assess graft quality and provide new insights into the etiology of EAD. Herein, we also offer a valid tool for the early prediction of EAD.
Collapse
Affiliation(s)
- Yimou Lin
- Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Haitao Huang
- Department of Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiaying Cao
- Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Ke Zhang
- Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Ruihan Chen
- Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Jingyu Jiang
- Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Xuewen Yi
- Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
| | - Shi Feng
- Department of Pathology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jimin Liu
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | - Shusen Zheng
- Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, Hangzhou, China
| | - Qi Ling
- Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Medical Center for Infectious Diseases, Hangzhou, China
| |
Collapse
|
14
|
Zhang D, Zhou Q, Yang X, Zhang Z, Wang D, Hu D, Huang Y, Sheng J, Wang X. Gallic Acid Can Promote Low-Density Lipoprotein Uptake in HepG2 Cells via Increasing Low-Density Lipoprotein Receptor Accumulation. Molecules 2024; 29:1999. [PMID: 38731489 PMCID: PMC11085419 DOI: 10.3390/molecules29091999] [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: 03/28/2024] [Revised: 04/16/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Gallic acid (GA) is a type of polyphenolic compound that can be found in a range of fruits, vegetables, and tea. Although it has been confirmed it improves non-alcoholic fatty liver disease (NAFLD), it is still unknown whether GA can improve the occurrence of NAFLD by increasing the low-density lipoprotein receptor (LDLR) accumulation and alleviating cholesterol metabolism disorders. Therefore, the present study explored the effect of GA on LDLR and its mechanism of action. The findings indicated that the increase in LDLR accumulation in HepG2 cells induced by GA was associated with the stimulation of the epidermal growth factor receptor-extracellular regulated protein kinase (EGFR-ERK1/2) signaling pathway. When the pathway was inhibited by EGFR mab cetuximab, it was observed that the activation of the EGFR-ERK1/2 signaling pathway induced by GA was also blocked. At the same time, the accumulation of LDLR protein and the uptake of LDL were also suppressed. Additionally, GA can also promote the accumulation of forkhead box O3 (FOXO3) and suppress the accumulation of hepatocyte nuclear factor-1α (HNF1α), leading to the inhibition of proprotein convertase subtilisin/kexin 9 (PCSK9) mRNA expression and protein accumulation. This ultimately results in increased LDLR protein accumulation and enhanced uptake of LDL in cells. In summary, the present study revealed the potential mechanism of GA's role in ameliorating NAFLD, with a view of providing a theoretical basis for the dietary supplementation of GA.
Collapse
Affiliation(s)
- Dongying Zhang
- College of Science, Yunnan Agricultural University, Kunming 650201, China; (D.Z.); (D.H.)
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China; (Q.Z.); (X.Y.); (Z.Z.); (D.W.)
| | - Qixing Zhou
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China; (Q.Z.); (X.Y.); (Z.Z.); (D.W.)
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China;
| | - Xiangxuan Yang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China; (Q.Z.); (X.Y.); (Z.Z.); (D.W.)
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China;
| | - Zhen Zhang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China; (Q.Z.); (X.Y.); (Z.Z.); (D.W.)
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China;
| | - Dongxue Wang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China; (Q.Z.); (X.Y.); (Z.Z.); (D.W.)
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China;
| | - Dandan Hu
- College of Science, Yunnan Agricultural University, Kunming 650201, China; (D.Z.); (D.H.)
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China; (Q.Z.); (X.Y.); (Z.Z.); (D.W.)
| | - Yewei Huang
- College of Science, Yunnan Agricultural University, Kunming 650201, China; (D.Z.); (D.H.)
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China; (Q.Z.); (X.Y.); (Z.Z.); (D.W.)
| | - Jun Sheng
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China;
| | - Xuanjun Wang
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, Yunnan University of Chinese Medicine, Kunming 650500, China
| |
Collapse
|
15
|
Guo L, Zhou L, Wei P, Li S, He S, Li D. Emerging Roles of UDP-GalNAc Polypeptide N-Acetylgalactosaminyltransferases in Cardiovascular Disease. Aging Dis 2024; 16:AD.2024.0308. [PMID: 38502587 PMCID: PMC11745429 DOI: 10.14336/ad.2024.0308] [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: 02/02/2024] [Accepted: 03/08/2024] [Indexed: 03/21/2024] Open
Abstract
UDP-GalNAc polypeptide N-acetylgalactosaminyltransferases (GalNAc-Ts) catalyze mucin-type O-glycosylation by transferring α-N-acetylgalactosamine (GalNAc) from UDP-GalNAc to Ser or Thr residues of target proteins. This post-translational modification is common in eukaryotes, yet its biological functions remain unclear. Recent studies have identified specific receptors in the heart and vascular wall cells that can be mucin-type O-glycosylated, and there is now substantial evidence confirming that patients with various cardiovascular diseases (CVDs), such as heart failure, coronary artery disease, myocardial hypertrophy, and vascular calcification, exhibit abnormal changes in GalNAc-Ts. This review aims to highlight recent advances in GalNAc-Ts and their roles in the cardiovascular system, intending to provide evidence for clinical treatment and prevention of CVDs.
Collapse
Affiliation(s)
- Liwei Guo
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Xinxiang, Henan, China.
| | - Lulu Zhou
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Pengcheng Wei
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Shijie Li
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Shanqing He
- Department of Cardiovascular Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Duan Li
- School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan, China.
- Henan Key Biological of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan, China.
| |
Collapse
|
16
|
Zhou S, Ling X, Zhu J, Liang Y, Feng Q, Xie C, Li J, Chen Q, Chen S, Miao J, Zhang M, Li Z, Shen W, Li X, Wu Q, Wang X, Liu R, Wang C, Hou FF, Kong Y, Liu Y, Zhou L. MAGL protects against renal fibrosis through inhibiting tubular cell lipotoxicity. Theranostics 2024; 14:1583-1601. [PMID: 38389852 PMCID: PMC10879875 DOI: 10.7150/thno.92848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/20/2024] [Indexed: 02/24/2024] Open
Abstract
Rationale: Renal fibrosis, with no therapeutic approaches, is a common pathological feature in various chronic kidney diseases (CKD). Tubular cell injury plays a pivotal role in renal fibrosis. Commonly, injured tubular cells exhibit significant lipid accumulation. However, the underlying mechanisms remain poorly understood. Methods: 2-arachidonoylglycerol (2-AG) levels in CKD patients and CKD model specimens were measured using mass spectrometry. 2-AG-loaded nanoparticles were infused into unilateral ureteral obstruction (UUO) mice. Lipid accumulation and renal fibrosis were tested. Furthermore, monoacylglycerol lipase (MAGL), the hydrolyzing enzyme of 2-AG, was assessed in CKD patients and models. Tubular cell-specific MAGL knock-in mice were generated. Moreover, MAGL recombination protein was also administered to unilateral ischemia reperfusion injury (UIRI) mice. Besides, a series of methods including RNA sequencing, metabolomics, primary cell culture, lipid staining, etc. were used. Results: 2-AG was increased in the serum or kidneys from CKD patients and models. Supplement of 2-AG further induced lipid accumulation and fibrogenesis through cannabinoid receptor type 2 (CB2)/β-catenin signaling. β-catenin knockout blocked 2-AG/CB2-induced fatty acid β-oxidation (FAO) deficiency and lipid accumulation. Remarkably, MAGL significantly decreased in CKD, aligning with lipid accumulation and fibrosis. Specific transgene of MAGL in tubular cells significantly preserved FAO, inhibited lipid-mediated toxicity in tubular cells, and finally retarded fibrogenesis. Additionally, supplementation of MAGL in UIRI mice also preserved FAO function, inhibited lipid accumulation, and protected against renal fibrosis. Conclusion: MAGL is a potential diagnostic marker for kidney function decline, and also serves as a new therapeutic target for renal fibrosis through ameliorating lipotoxicity.
Collapse
Affiliation(s)
- Shan Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xian Ling
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jielin Zhu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Health Care, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Ye Liang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qijian Feng
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chao Xie
- Nephrology Department, The First People's Hospital of Foshan, Foshan, China
| | - Jiemei Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiyan Chen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nephrology Department, The First People's Hospital of Foshan, Foshan, China
| | - Shuangqin Chen
- Division of Nephrology, Department of medicine, The Fifth Affiliated Hospital Sun Yat-Sen University, Zhuhai, China
| | - Jinhua Miao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengyao Zhang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiru Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weiwei Shen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaolong Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qinyu Wu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxu Wang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruiyuan Liu
- School of Pharmaceutical Sciences and School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Cheng Wang
- Division of Nephrology, Department of medicine, The Fifth Affiliated Hospital Sun Yat-Sen University, Zhuhai, China
| | - Fan Fan Hou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yaozhong Kong
- Nephrology Department, The First People's Hospital of Foshan, Foshan, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
17
|
Ma YX, Chai YJ, Han YQ, Zhao SB, Yang GY, Wang J, Ming SL, Chu BB. Pseudorabies virus upregulates low-density lipoprotein receptors to facilitate viral entry. J Virol 2024; 98:e0166423. [PMID: 38054618 PMCID: PMC10804996 DOI: 10.1128/jvi.01664-23] [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: 10/23/2023] [Accepted: 11/17/2023] [Indexed: 12/07/2023] Open
Abstract
Pseudorabies virus (PRV) is the causative agent of Aujeszky's disease in pigs. The low-density lipoprotein receptor (LDLR) is a transcriptional target of the sterol-regulatory element-binding proteins (SREBPs) and participates in the uptake of LDL-derived cholesterol. However, the involvement of LDLR in PRV infection has not been well characterized. We observed an increased expression level of LDLR mRNA in PRV-infected 3D4/21, PK-15, HeLa, RAW264.7, and L929 cells. The LDLR protein level was also upregulated by PRV infection in PK-15 cells and in murine lung and brain. The treatment of cells with the SREBP inhibitor, fatostatin, or with SREBP2-specific small interfering RNA prevented the PRV-induced upregulation of LDLR expression as well as viral protein expression and progeny virus production. This suggested that PRV activated SREBPs to induce LDLR expression. Furthermore, interference in LDLR expression affected PRV proliferation, while LDLR overexpression promoted it. This indicated that LDLR was involved in PRV infection. The study also demonstrated that LDLR participated in PRV invasions. The overexpression of LDLR or inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9), which binds to LDLR and targets it for lysosomal degradation, significantly enhanced PRV attachment and entry. Mechanistically, LDLR interacted with PRV on the plasma membrane, and pretreatment of cells with LDLR antibodies was able to neutralize viral entry. An in vivo study indicated that the treatment of mice with the PCSK9 inhibitor SBC-115076 promoted PRV proliferation. The data from the study indicate that PRV hijacks LDLR for viral entry through the activation of SREBPs.IMPORTANCEPseudorabies virus (PRV) is a herpesvirus that primarily manifests as fever, pruritus, and encephalomyelitis in various domestic and wild animals. Owing to its lifelong latent infection characteristics, PRV outbreaks have led to significant financial setbacks in the global pig industry. There is evidence that PRV variant strains can infect humans, thereby crossing the species barrier. Therefore, gaining deeper insights into PRV pathogenesis and developing updated strategies to contain its spread are critical. This study posits that the low-density lipoprotein receptor (LDLR) could be a co-receptor for PRV infection. Hence, strategies targeting LDLR may provide a promising avenue for the development of effective PRV vaccines and therapeutic interventions.
Collapse
Affiliation(s)
- Ying-Xian Ma
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Zhengzhou, Henan, China
- Key Laboratory of Animal Growth and Development, Zhengzhou, Henan, China
| | - Ya-Jing Chai
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Zhengzhou, Henan, China
- Key Laboratory of Animal Growth and Development, Zhengzhou, Henan, China
| | - Ya-Qi Han
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Zhengzhou, Henan, China
- Key Laboratory of Animal Growth and Development, Zhengzhou, Henan, China
| | - Shi-Bo Zhao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Zhengzhou, Henan, China
- Key Laboratory of Animal Growth and Development, Zhengzhou, Henan, China
| | - Guo-Yu Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Zhengzhou, Henan, China
- Key Laboratory of Animal Growth and Development, Zhengzhou, Henan, China
- International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, Henan, China
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Zhengzhou, Henan, China
- Key Laboratory of Animal Growth and Development, Zhengzhou, Henan, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan, China
| | - Sheng-Li Ming
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Zhengzhou, Henan, China
- Key Laboratory of Animal Growth and Development, Zhengzhou, Henan, China
| | - Bei-Bei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Zhengzhou, Henan, China
- Key Laboratory of Animal Growth and Development, Zhengzhou, Henan, China
- International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, Henan, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan, China
- Longhu Advanced Immunization Laboratory, Zhengzhou, Henan, China
| |
Collapse
|
18
|
Ou-Yang YN, Deng FF, Wang YJ, Chen M, Yang PF, Yang Z, Tian Z. High-salt diet induces dyslipidemia through the SREBP2/PCSK9 pathway in dahl salt-sensitive rats. Biochimie 2024; 216:34-45. [PMID: 37844755 DOI: 10.1016/j.biochi.2023.10.001] [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: 01/18/2023] [Revised: 09/24/2023] [Accepted: 10/02/2023] [Indexed: 10/18/2023]
Abstract
A high-salt diet is known to increase serum cholesterol levels; however, the underlying mechanism of salt-induced dyslipidemia in patients with salt-sensitivity remains poorly understood. We aimed to investigate whether high-salt diet (HSD) can induce dyslipidemia and elucidate the underlying mechanism of salt-induced dyslipidemia in Dahl salt-sensitive (SS) rats. Metabolomic and biochemical analyses revealed that the consumption of an HSD (8 % NaCl) significantly increased the serum levels of total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) in SS rats. The enzyme-linked immunosorbent assay demonstrated an increase in circulating proprotein convertase subtilisin/kexin type 9 (PCSK9) levels, accompanied by a decrease in hepatic low-density lipoprotein receptor (LDLR) levels due to HSD consumption. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis revealed that HSD consumption activated sterol regulatory element-binding protein-2 (SREBP2) expression in the liver and kidney, resulting in upregulation of PCSK9 at the transcriptional level in the liver and at the translational level in the kidney, ultimately increasing circulating PCSK9 levels. The combined effects of HSD on the liver and kidney contributed to the development of hypercholesterolemia. Furthermore, an in vitro assay confirmed that high-salt exposure led to an increase in the protein expression of SREBP2 and PCSK9 secretion, thereby reducing low-density lipoprotein (LDL) uptake. This study, for the first time, shows that an HSD induces dyslipidemia through activation of the SREBP2/PCSK9 pathway, providing new insights into the prevention and treatment of dyslipidemia in patients with salt sensitivity.
Collapse
Affiliation(s)
- Ya-Nan Ou-Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Fen-Fen Deng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Yun-Jia Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Meng Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Peng-Fei Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Zhongmin Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| |
Collapse
|
19
|
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
|
20
|
Kroh A, Walter J, Fragoulis A, Möckel D, Lammers T, Kiessling F, Andruszkow J, Preisinger C, Egbert M, Jiao L, Eickhoff RM, Heise D, Berndt N, Cramer T, Neumann UP, Egners A, Ulmer TF. Hepatocellular loss of mTOR aggravates tumor burden in nonalcoholic steatohepatitis-related HCC. Neoplasia 2023; 46:100945. [PMID: 37976569 PMCID: PMC10685311 DOI: 10.1016/j.neo.2023.100945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/13/2023] [Indexed: 11/19/2023]
Abstract
Obesity and associated nonalcoholic steatohepatitis (NASH) are on the rise globally. NASH became an important driver of hepatocellular carcinoma (HCC) in recent years. Activation of the central metabolic regulator mTOR (mechanistic target of rapamycin) is frequently observed in HCCs. However, mTOR inhibition failed to improve the outcome of HCC therapies, demonstrating the need for a better understanding of the molecular and functional consequences of mTOR blockade. We established a murine NASH-driven HCC model based on long-term western diet feeding combined with hepatocellular mTOR-inactivation. We evaluated tumor load and whole-body fat percentage via µCT-scans, analyzed metabolic blood parameters and tissue proteome profiles. Additionally, we used a bioinformatic model to access liver and HCC mitochondrial metabolic functions. The tumor burden was massively increased via mTOR-knockout. Several signs argue for extensive metabolic reprogramming of glucose, fatty acid, bile acid and cholesterol metabolism. Kinetic modeling revealed reduced oxygen consumption in KO-tumors. NASH-derived HCC pathogenesis is driven by metabolic disturbances and should be considered separately from those caused by other etiologies. We conclude that mTOR functions as tumor suppressor in hepatocytes especially under long-term western diet feeding. However, some of the detrimental consequences of this diet are attenuated by mTOR blockade.
Collapse
Affiliation(s)
- Andreas Kroh
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany.
| | - Jeanette Walter
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany; Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, RWTH Aachen University Hospital, Aachen, Germany
| | - Athanassios Fragoulis
- Department of Anatomy and Cell Biology, RWTH Aachen University Hospital Aachen, Germany
| | - Diana Möckel
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen, Germany
| | - Julia Andruszkow
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Christian Preisinger
- Proteomics Facility, Interdisciplinary Center for Clinical Research (IZKF) Aachen, Medical School, RWTH Aachen University Hospital, Aachen, Germany
| | - Maren Egbert
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Long Jiao
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany; Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, PR China
| | - Roman M Eickhoff
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Daniel Heise
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Nikolaus Berndt
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany; Institute of Computer-assisted Cardiovascular Medicine, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thorsten Cramer
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Ulf Peter Neumann
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany; Department of Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Antje Egners
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Tom Florian Ulmer
- Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany; Department of Surgery, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
21
|
Sivaraman K, Pino P, Raussin G, Anchisi S, Metayer C, Dagany N, Held J, Wrenger S, Welte T, Wurm MJ, Wurm FM, Olejnicka B, Janciauskiene S. Human PBMCs Form Lipid Droplets in Response to Spike Proteins. Microorganisms 2023; 11:2683. [PMID: 38004695 PMCID: PMC10672762 DOI: 10.3390/microorganisms11112683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/22/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
Intracellular lipid droplets (LDs) can accumulate in response to inflammation, metabolic stresses, and other physiological/pathological processes. Herein, we investigated whether spike proteins of SARS-CoV-2 induce LDs in human peripheral blood mononuclear cells (PBMCs) and in pulmonary microvascular endothelial cells (HPMECs). PBMCs or HPMECs were incubated alone or with endotoxin-free recombinant variants of trimeric spike glycoproteins (Alpha, Beta, Delta, and Omicron, 12 µg/mL). Afterward, cells were stained with Oil Red O for LDs, cytokine release was determined through ELISA, and the gene expression was analyzed through real-time PCR using TaqMan assays. Our data show that spikes induce LDs in PBMCs but not in HPMECs. In line with this, in PBMCs, spike proteins lower the expression of genes involving lipid metabolism and LD formation, such as SREBF1, HMGCS1, LDLR, and CD36. On the other hand, PBMCs exposed to spikes for 6 or 18 h did not increase in IL-1β, IL-6, IL-8, MCP-1, and TNFα release or expression as compared to non-treated controls. Thus, spike-induced LD formation in PBMCs seems to not be related to cell inflammatory activation. Further detailed studies are warranted to investigate in which specific immune cells spikes induce LDs, and what are the pathophysiological mechanisms and consequences of this induction in vivo.
Collapse
Affiliation(s)
- Kokilavani Sivaraman
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, 30625 Hannover, Germany
| | - Paco Pino
- ExcellGene SA, 1970 Monthey, Switzerland
| | | | | | | | | | - Julia Held
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, 30625 Hannover, Germany
| | - Sabine Wrenger
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, 30625 Hannover, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, 30625 Hannover, Germany
| | | | - Florian M. Wurm
- ExcellGene SA, 1970 Monthey, Switzerland
- Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Beata Olejnicka
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, 30625 Hannover, Germany
| | - Sabina Janciauskiene
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
22
|
Skeby CK, Hummelgaard S, Gustafsen C, Petrillo F, Frederiksen KP, Olsen D, Kristensen T, Ivarsen P, Madsen P, Christensen EI, Nielsen R, Birn H, Glerup S, Weyer K. Proprotein convertase subtilisin/kexin type 9 targets megalin in the kidney proximal tubule and aggravates proteinuria in nephrotic syndrome. Kidney Int 2023; 104:754-768. [PMID: 37406929 DOI: 10.1016/j.kint.2023.06.024] [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: 10/06/2022] [Revised: 06/04/2023] [Accepted: 06/09/2023] [Indexed: 07/07/2023]
Abstract
Proteinuria is a prominent feature of chronic kidney disease. Interventions that reduce proteinuria slow the progression of chronic kidney disease and the associated risk of cardiovascular disease. Here, we propose a mechanistic coupling between proteinuria and proprotein convertase subtilisin/kexin type 9 (PCSK9), a regulator of cholesterol and a therapeutic target in cardiovascular disease. PCSK9 undergoes glomerular filtration and is captured by megalin, the receptor responsible for driving protein reabsorption in the proximal tubule. Accordingly, megalin-deficient mice and patients carrying megalin pathogenic variants (Donnai Barrow syndrome) were characterized by elevated urinary PCSK9 excretion. Interestingly, PCSK9 knockout mice displayed increased kidney megalin while PCSK9 overexpression resulted in its reduction. Furthermore, PCSK9 promoted trafficking of megalin to lysosomes in cultured proximal tubule cells, suggesting that PCSK9 is a negative regulator of megalin. This effect can be accelerated under disease conditions since either genetic destruction of the glomerular filtration barrier in podocin knockout mice or minimal change disease (a common cause of nephrotic syndrome) in patients resulted in enhanced tubular PCSK9 uptake and urinary PCSK9 excretion. Pharmacological PCSK9 inhibition increased kidney megalin while reducing urinary albumin excretion in nephrotic mice. Thus, glomerular damage increases filtration of PCSK9 and concomitantly megalin degradation, resulting in escalated proteinuria.
Collapse
Affiliation(s)
- Cecilie K Skeby
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Camilla Gustafsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Draupnir Bio, INCUBA Skejby, Aarhus, Denmark
| | | | | | - Ditte Olsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Draupnir Bio, INCUBA Skejby, Aarhus, Denmark
| | - Tilde Kristensen
- Department of Internal Medicine, Renal Unit, Regional Hospital Viborg, Viborg, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Per Ivarsen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Peder Madsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Draupnir Bio, INCUBA Skejby, Aarhus, Denmark
| | | | - Rikke Nielsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Henrik Birn
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Draupnir Bio, INCUBA Skejby, Aarhus, Denmark
| | - Kathrin Weyer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
23
|
Stevenson ER, Smith LC, Wilkinson ML, Lee SJ, Gow AJ. Etiology of lipid-laden macrophages in the lung. Int Immunopharmacol 2023; 123:110719. [PMID: 37595492 PMCID: PMC10734282 DOI: 10.1016/j.intimp.2023.110719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/20/2023]
Abstract
Uniquely positioned as sentinel cells constantly exposed to the environment, pulmonary macrophages are vital for the maintenance of the lung lining. These cells are responsible for the clearance of xenobiotics, pathogen detection and clearance, and homeostatic functions such as surfactant recycling. Among the spectrum of phenotypes that may be expressed by macrophages in the lung, the pulmonary lipid-laden phenotype is less commonly studied in comparison to its circulatory counterpart, the atherosclerotic lesion-associated foam cell, or the acutely activated inflammatory macrophage. Herein, we propose that lipid-laden macrophage formation in the lung is governed by lipid acquisition, storage, metabolism, and export processes. The cellular balance of these four processes is critical to the maintenance of homeostasis and the prevention of aberrant signaling that may contribute to lung pathologies. This review aims to examine mechanisms and signaling pathways that are involved in lipid-laden macrophage formation and the potential consequences of this phenotype in the lung.
Collapse
Affiliation(s)
- E R Stevenson
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States
| | - L C Smith
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States; Department of Pharmaceutical Sciences, University of Connecticut School of Pharmacy, Storrs, CT, United States
| | - M L Wilkinson
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States
| | - S J Lee
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States
| | - A J Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
24
|
Li W, Pang Y, Jin K, Wang Y, Wu Y, Luo J, Xu W, Zhang X, Xu R, Wang T, Jiao L. Membrane contact sites orchestrate cholesterol homeostasis that is central to vascular aging. WIREs Mech Dis 2023; 15:e1612. [PMID: 37156598 DOI: 10.1002/wsbm.1612] [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: 10/15/2022] [Revised: 02/12/2023] [Accepted: 04/19/2023] [Indexed: 05/10/2023]
Abstract
Chronological age causes structural and functional vascular deterioration and is a well-established risk factor for the development of cardiovascular diseases, leading to more than 40% of all deaths in the elderly. The etiology of vascular aging is complex; a significant impact arises from impaired cholesterol homeostasis. Cholesterol level is balanced through synthesis, uptake, transport, and esterification, the processes executed by multiple organelles. Moreover, organelles responsible for cholesterol homeostasis are spatially and functionally coordinated instead of isolated by forming the membrane contact sites. Membrane contact, mediated by specific protein-protein interaction, pulls opposing organelles together and creates the hybrid place for cholesterol transfer and further signaling. The membrane contact-dependent cholesterol transfer, together with the vesicular transport, maintains cholesterol homeostasis and has intimate implications in a growing list of diseases, including vascular aging-related diseases. Here, we summarized the latest advances regarding cholesterol homeostasis by highlighting the membrane contact-based regulatory mechanism. We also describe the downstream signaling under cholesterol homeostasis perturbations, prominently in cholesterol-rich conditions, stimulating age-dependent organelle dysfunction and vascular aging. Finally, we discuss potential cholesterol-targeting strategies for therapists regarding vascular aging-related diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Wenjing Li
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Yiyun Pang
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Kehan Jin
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuru Wang
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yujie Wu
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jichang Luo
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Wenlong Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Xiao Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Ran Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- Department of Interventional Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
25
|
Funes AK, Avena MV, Ibañez J, Simón L, Ituarte L, Colombo R, Roldán A, Conte MI, Monclus MÁ, Boarelli P, Fornés MW, Saez Lancellotti TE. Extra-virgin olive oil ameliorates high-fat diet-induced seminal and testicular disorders by modulating the cholesterol pathway. Andrology 2023; 11:1203-1217. [PMID: 36695747 DOI: 10.1111/andr.13398] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 11/02/2022] [Accepted: 01/02/2023] [Indexed: 01/26/2023]
Abstract
BACKGROUND Rabbits are sensitive to dietary cholesterol and rapidly develop hypercholesterolemia, leading to prominent subfertility. Sterol regulatory element-binding protein isoform 2 drives the intracellular cholesterol pathway in many tissues, including the testicles. Its abnormal regulation could be the mainly responsible for the failure of suppressing cholesterol synthesis in a cholesterol-enriched environment, ultimately leading to testicular and seminal alterations. However, extra-virgin olive oil consumption has beneficial properties that promote lowering of cholesterol levels, including the recovery of seminal parameters altered under a high-fat diet. OBJECTIVES Our goal was to investigate the effects of high-fat diet supplementation with extra-virgin olive oil at the molecular level on rabbit testes, by analyzing sterol regulatory element-binding protein isoform 2 protein and its corresponding downstream effectors. MATERIALS AND METHODS During 12 months, male rabbits were fed a control diet, high-fat diet, or 6-month high-fat diet followed by 6-month high-fat diet plus extra-virgin olive oil. Serum lipids, testosterone levels, bodyweight, and seminal parameters were tested. The mRNA and protein levels of sterol regulatory element-binding protein isoform 2, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase, and low-density lipoprotein receptor were determined by semi-quantitative polymerase chain reaction and Western blotting techniques. The expression pattern of sterol regulatory element-binding protein isoform 2 protein in the rabbit testicles was studied by indirect immunofluorescence. In addition, testicular cholesterol was detected and quantified by filipin staining and gas chromatography. RESULTS The data showed that the addition of extra-virgin olive oil to high-fat diet reduced testicular cholesterol levels and recovered the expression of sterol regulatory element-binding protein isoform 2, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase, and low-density lipoprotein receptor initially altered by the high-fat diet. DISCUSSION AND CONCLUSIONS The combination of high-fat diet with extra-virgin olive oil encourages testicular recovery by modifying the expression of the enzymes related to intracellular cholesterol management.
Collapse
Affiliation(s)
- Abi Karenina Funes
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - María Virginia Avena
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - Jorge Ibañez
- Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - Layla Simón
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - Leonor Ituarte
- Área de Física Biológica, Departamento de Morfofisiología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Regina Colombo
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - Adrián Roldán
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de Investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina
| | - María Inés Conte
- Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - María Ángeles Monclus
- Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de Investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina
| | - Paola Boarelli
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Laboratorio de Enfermedades Metabólicas (LEM), Universidad Juan Agustín Maza, Mendoza, Argentina
| | - Miguel Walter Fornés
- Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
| | - Tania Estefanía Saez Lancellotti
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (MeNu). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de Histología y Embriología (IHEM), CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza, Argentina
- Instituto de Investigaciones, Facultad de Ciencias Médicas, Universidad del Aconcagua, Mendoza, Argentina
| |
Collapse
|
26
|
Song YJ, Zhang J, Xiao J, Feng H, Xu Z, Nie P, Chang MX. Piscine Vitamin D Receptors Vdra/Vdrb in the Absence of Vitamin D Are Utilized by Grass Carp Reovirus for Promoting Viral Replication. Microbiol Spectr 2023; 11:e0128723. [PMID: 37466438 PMCID: PMC10433867 DOI: 10.1128/spectrum.01287-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/23/2023] [Indexed: 07/20/2023] Open
Abstract
The vitamin D receptor (VDR) plays a pivotal role in the biological actions of vitamin D (VitD). However, little is known about the functions of VDR in the production of viral inclusion bodies (VIBs). Using a representative strain of grass carp reovirus (GCRV) genotype I, GCRV-873, we show that GCRV-873 recruits grass carp Vdrs for promoting the production of VIBs in the absence of VitD. Inhibition of cholesterol synthesis by lovastatin impairs the production of VIBs and blocks the effects of grass carp Vdrs in promoting the production of VIBs in the absence of VitD. Furthermore, grass carp Vdrs are found to form the Vdra-Vdrb heterodimer, which is vital for 3-hydroxy-3-methylglutaryl-coenzyme A reductase (hmgcr)-dependent cholesterol synthesis and GCRV replication. Intriguingly in the presence of VitD, grass carp Vdra but not Vdrb forms the heterodimer with the retinoid X receptor beta b (Rxrbb), which induces the transcription of those genes involved in the RIG-I-like receptor (RLR) antiviral signaling pathway for inhibiting GCRV infection. Furthermore, the VitD-activated Vdra-Vdrb heterodimer attenuates the transcription of the RLR antiviral signaling pathway induced by VitD. In the presence of VitD, a balance between the Vdra-Rxrbb heterodimers as coactivators and Vdra-Vdrb heterodimers as corepressors in affecting the transcriptional regulation of the RLR antiviral signaling pathway may eventually determine the outcome of GCRV infection. Transfection with VitD can abolish the effect of grass carp Vdrs in promoting GCRV replication in a dose-dependent manner. Taken together, these findings demonstrate that GCRV utilizes host Vdrs to increase hmgcr-dependent cholesterol synthesis for promoting its replication, which can be prevented by VitD treatment. IMPORTANCE Grass carp reovirus (GCRV) is the causative agent of grass carp hemorrhagic disease, which seriously harms freshwater fish. Although many positive or negative regulators of GCRV infection have been identified in teleosts, little is known about the molecular mechanisms by which GCRV utilizes host factors to generate its infectious compartments beneficial for viral replication and infection. Here, we show that in the absence of VitD, the GCRV-873 strain utilizes host vitamin D receptors Vdra/Vdrb to increase hmgcr-dependent cholesterol synthesis for promoting the production of VIBs, which are important functional sites for aquareovirus replication and assembly. The negative regulation of Vdrs during viral infection can be prevented by VitD treatment. Thus, this present work broadens understanding of the pivotal roles of Vdrs in the interaction between the host and GCRV in the absence or presence of VitD, which might provide a rational basis for developing novel anti-GCRV strategies.
Collapse
Affiliation(s)
- Yun Jie Song
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jie Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Jun Xiao
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, China
| | - Hao Feng
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, China
| | - Zhen Xu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Pin Nie
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
- Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Ming Xian Chang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
- Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
27
|
Wu R, Prachyathipsakul T, Zhuang J, Liu H, Han Y, Liu B, Gong S, Qiu J, Wong S, Ribbe A, Medeiros J, Bhagabati J, Gao J, Wu P, Dutta R, Herrera R, Faraci S, Xiao H, Thayumanavan S. Conferring liver selectivity to a thyromimetic using a novel nanoparticle increases therapeutic efficacy in a diet-induced obesity animal model. PNAS NEXUS 2023; 2:pgad252. [PMID: 37649581 PMCID: PMC10465086 DOI: 10.1093/pnasnexus/pgad252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 07/21/2023] [Indexed: 09/01/2023]
Abstract
Optimization of metabolic regulation is a promising solution for many pathologies, including obesity, dyslipidemia, type 2 diabetes, and inflammatory liver disease. Synthetic thyroid hormone mimics-based regulation of metabolic balance in the liver showed promise but was hampered by the low biocompatibility and harmful effects on the extrahepatic axis. In this work, we show that specifically directing the thyromimetic to the liver utilizing a nanogel-based carrier substantially increased therapeutic efficacy in a diet-induced obesity mouse model, evidenced by the near-complete reversal of body weight gain, liver weight and inflammation, and cholesterol levels with no alteration in the thyroxine (T4) / thyroid stimulating hormone (TSH) axis. Mechanistically, the drug acts by binding to thyroid hormone receptor β (TRβ), a ligand-inducible transcription factor that interacts with thyroid hormone response elements and modulates target gene expression. The reverse cholesterol transport (RCT) pathway is specifically implicated in the observed therapeutic effect. Overall, the study demonstrates a unique approach to restoring metabolic regulation impacting obesity and related metabolic dysfunctions.
Collapse
Affiliation(s)
- Ruiling Wu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Theeraphop Prachyathipsakul
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jiaming Zhuang
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Hongxu Liu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Yanhui Han
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Bin Liu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Shuai Gong
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jingyi Qiu
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Siu Wong
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Alexander Ribbe
- Department of Polymer Science and Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Jewel Medeiros
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jayashree Bhagabati
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Jingjing Gao
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Peidong Wu
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Ranit Dutta
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | | | | | - Hang Xiao
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- The Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
28
|
Liu L, Kong Q, Xiang Z, Kuang X, Wang H, Zhou L, Feng S, Chen T, Ding C. Integrated Analysis of Transcriptome and Metabolome Provides Insight into Camellia oleifera Oil Alleviating Fat Accumulation in High-Fat Caenorhabditis elegans. Int J Mol Sci 2023; 24:11615. [PMID: 37511379 PMCID: PMC10380387 DOI: 10.3390/ijms241411615] [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: 05/30/2023] [Revised: 07/05/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Camellia oil (CO) is a high medicinal and nutritional value edible oil. However, its ability to alleviate fat accumulation in high-fat Caenorhabditis elegans has not been well elucidated. Therefore, this study aimed to investigate the effect of CO on fat accumulation in high-fat C. elegans via transcriptome and metabolome analysis. The results showed that CO significantly reduced fat accumulation in high-fat C. elegans by 10.34% (Oil Red O method) and 11.54% (TG content method), respectively. Furthermore, CO primarily altered the transcription levels of genes involved in longevity regulating pathway. Specifically, CO decreased lipid storage in high-fat C. elegans by inhibiting fat synthesis. In addition, CO supplementation modulated the abundance of metabolic biomarkers related to pyrimidine metabolism and riboflavin metabolism. The integrated transcriptome and metabolome analyses indicated that CO supplementation could alleviate fat accumulation in high-fat C. elegans by regulating retinol metabolism, drug metabolism-cytochrome P450, metabolism of xenobiotics by cytochrome P450, ascorbate and aldarate metabolism, and pentose and glucuronate interconversions. Overall, these findings highlight the potential health benefits of CO that could potentially be used as a functional edible oil.
Collapse
Affiliation(s)
- Li Liu
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China
| | - Qingbo Kong
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China
| | - Zhuoya Xiang
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China
- Institute of Agro-Products Processing Science and Technology, Sichuan Academy of Agricultural Sciences, Chengdu 610066, China
| | - Xuekun Kuang
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China
| | - Heng Wang
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China
| | - Lijun Zhou
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China
| | - Shiling Feng
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China
| | - Tao Chen
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China
| | - Chunbang Ding
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, China
| |
Collapse
|
29
|
White AM, Best OG, Hotinski AK, Kuss BJ, Thurgood LA. The Role of Cholesterol in Chronic Lymphocytic Leukemia Development and Pathogenesis. Metabolites 2023; 13:799. [PMID: 37512506 PMCID: PMC10385576 DOI: 10.3390/metabo13070799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
Cholesterol has many critical functions in cells. It is a key component of membranes and cell-signalling processes, and it functions as a chemical precursor in several biochemical pathways, such as Vitamin D and steroid synthesis. Cholesterol has also been implicated in the development and progression of various cancers, in which it is thought to promote cell proliferation, migration, and invasion. Chronic lymphocytic leukemia (CLL) is an example of a lipid-avid cancer that relies on lipid metabolism, rather than glycolysis, to fuel cell proliferation. However, data regarding the role of cholesterol in CLL are conflicting. Studies have shown that dyslipidaemia is more common among CLL patients than age-matched healthy controls, and that CLL patients who take cholesterol-lowering drugs, such as statins, appear to have improved survival rates. Therefore, defining the roles of cholesterol in CLL may highlight the importance of monitoring and managing hyperlipidaemia as part of the routine management of patients with CLL. In this review, we discuss the roles of cholesterol in the context of CLL by examining the literature concerning the trafficking, uptake, endogenous synthesis, and intracellular handling of this lipid. Data from clinical trials investigating various classes of cholesterol and lipid-lowering drugs in CLL are also discussed.
Collapse
Affiliation(s)
- Alana M White
- Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Oliver G Best
- Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Anya K Hotinski
- Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Bryone J Kuss
- Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Lauren A Thurgood
- Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| |
Collapse
|
30
|
Munkhsaikhan U, Kwon YI, Sahyoun AM, Galán M, Gonzalez AA, Ait-Aissa K, Abidi AH, Kassan A, Kassan M. The Beneficial Effect of Lomitapide on the Cardiovascular System in LDLr -/- Mice with Obesity. Antioxidants (Basel) 2023; 12:1287. [PMID: 37372017 PMCID: PMC10295391 DOI: 10.3390/antiox12061287] [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: 03/07/2023] [Revised: 05/26/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
OBJECTIVES Homozygous familial hypercholesteremia (HoFH) is a rare, life-threatening metabolic disease, mainly caused by a mutation in the LDL receptor. If untreated, HoFH causes premature death from acute coronary syndrome. Lomitapide is approved by the FDA as a therapy to lower lipid levels in adult patients with HoFH. Nevertheless, the beneficial effect of lomitapide in HoFH models remains to be defined. In this study, we investigated the effect of lomitapide on cardiovascular function using LDL receptor-knockout mice (LDLr-/-). METHODS Six-week-old LDLr-/- mice were fed a standard diet (SD) or a high-fat diet (HFD) for 12 weeks. Lomitapide (1 mg/Kg/Day) was given by oral gavage for the last 2 weeks in the HFD group. Body weight and composition, lipid profile, blood glucose, and atherosclerotic plaques were measured. Vascular reactivity and markers for endothelial function were determined in conductance arteries (thoracic aorta) and resistance arteries (mesenteric resistance arteries (MRA)). Cytokine levels were measured by using the Mesoscale discovery V-Plex assays. RESULTS Body weight (47.5 ± 1.5 vs. 40.3 ± 1.8 g), % of fat mass (41.6 ± 1.9% vs. 31.8 ± 1.7%), blood glucose (215.5 ± 21.9 vs. 142.3 ± 7.7 mg/dL), and lipid levels (cholesterol: 600.9 ± 23.6 vs. 451.7 ± 33.4 mg/dL; LDL/VLDL: 250.6 ± 28.9 vs. 161.1 ± 12.24 mg/dL; TG: 299.5 ± 24.1 vs. 194.1 ± 28.1 mg/dL) were significantly decreased, and the % of lean mass (56.5 ± 1.8% vs. 65.2 ± 2.1%) was significantly increased in the HFD group after lomitapide treatment. The atherosclerotic plaque area also decreased in the thoracic aorta (7.9 ± 0.5% vs. 5.7 ± 0.1%). After treatment with lomitapide, the endothelium function of the thoracic aorta (47.7 ± 6.3% vs. 80.7 ± 3.1%) and mesenteric resistance artery (66.4 ± 4.3% vs. 79.5 ± 4.6%) was improved in the group of LDLr-/- mice on HFD. This was correlated with diminished vascular endoplasmic (ER) reticulum stress, oxidative stress, and inflammation. CONCLUSIONS Treatment with lomitapide improves cardiovascular function and lipid profile and reduces body weight and inflammatory markers in LDLr-/- mice on HFD.
Collapse
Affiliation(s)
- Undral Munkhsaikhan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Bioscience Research and General Dentistry, College of Dentistry, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Young In Kwon
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Amal M. Sahyoun
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Food Science and Agriculture Chemistry, McGill University, Montreal, QC H9X 3V9, Canada
| | - María Galán
- Faculty of Health Sciences, University Rey Juan Carlos, 28922 Alcorcón, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), ISCIII, 28029 Madrid, Spain
| | - Alexis A. Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso 300, Chile
| | - Karima Ait-Aissa
- College of Dental Medicine, Lincoln Memorial University, Knoxville, TN 37923, USA
| | - Ammaar H. Abidi
- Department of Bioscience Research and General Dentistry, College of Dentistry, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
- College of Dental Medicine, Lincoln Memorial University, Knoxville, TN 37923, USA
| | - Adam Kassan
- Department of Pharmaceutical Sciences, School of Pharmacy, West Coast University, Los Angeles, CA 91606, USA
| | - Modar Kassan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- College of Dental Medicine, Lincoln Memorial University, Knoxville, TN 37923, USA
| |
Collapse
|
31
|
Liu XH, Pang X, Jin L, Pu DY, Wang ZJ, Zhang YG. Exposure to acute waterborne cadmium caused severe damage on lipid metabolism of freshwater fish, revealed by nuclear lipid droplet deposition in hepatocytes of rare minnow. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 257:106433. [PMID: 36841070 DOI: 10.1016/j.aquatox.2023.106433] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/02/2023] [Accepted: 02/16/2023] [Indexed: 05/12/2023]
Abstract
Cadmium (Cd) is a widely distributed aquatic toxic heavy metal with the potential to disrupt fish metabolism; however, more research is needed to clarify the underlying mechanisms. In the present study, rare minnows (Gobiocypris rarus) were used to detect the effects of cadmium on freshwater fish lipid metabolism and its underlying mechanism by histopathological observation, measurement of serum and liver biochemical indexes, and analysis of gene expression in terms of lipid oxidation, synthesis and transport. Here, severe damage, such as cytoplasmic lipid droplet (LD) accumulation, ectopic deposition of LDs, and the appearance of nuclear LDs (nLDs), was detected after exposure to 2.0 mg/L or higher concentrations (2.5 and 2.8 mg/L CdCl2) for 96 h. Other damage included abnormal increases in rough endoplasmic reticulum (RER) lamellae in a fingerprint or concentric circle pattern and necrosis of hepatocytes, and which was observed in the livers of fish exposed to 2.0 mg/L CdCl2.. Both hepatic and serum lipids, such as triglycerides and total cholesterol, were significantly increased after exposure to 2.0 mg/L CdCl2, as was serum lipase (LPS). Hepatic lipase and lipoprotein lipase remained unchanged, in accordance with the unchanged hepatic mRNA transcripts of PPARɑ. Furthermore, the mRNA transcripts of both SCD and SQLE were significantly decreased. Moreover, hepatic and serum low-density and high-density lipoprotein cholesterol showed significant changes, which were accompanied by a significant increase and decrease in hepatic APOAI and APOB100 mRNA levels, respectively. All the results indicate the presence of severe damage to hepatic lipid metabolism and that disrupted lipid transport may play a key role in the accumulation of hepatic LDs. In addition, the hepatic nLDs of nonmammalian vertebrates and their location across the nuclear envelope are intriguing, suggesting that large-size nLDs are a common marker for severe liver damage.
Collapse
Affiliation(s)
- Xiao-Hong Liu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, Southwest University School of Life Sciences, Chongqing 400715, China
| | - Xu Pang
- College of Fisheries, Institute of Three Gorges Ecological Fisheries of Chongqing, Southwest University, Chongqing 400715, China
| | - Li Jin
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, Southwest University School of Life Sciences, Chongqing 400715, China
| | - De-Yong Pu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, Southwest University School of Life Sciences, Chongqing 400715, China
| | - Zhi-Jian Wang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, Southwest University School of Life Sciences, Chongqing 400715, China.
| | - Yao-Guang Zhang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, Southwest University School of Life Sciences, Chongqing 400715, China.
| |
Collapse
|
32
|
Huang YW, Wang LT, Zhang M, Nie Y, Yang JB, Meng WL, Wang XJ, Sheng J. Caffeine can alleviate non-alcoholic fatty liver disease by augmenting LDLR expression via targeting EGFR. Food Funct 2023; 14:3269-3278. [PMID: 36916513 DOI: 10.1039/d2fo02701a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Increasing low-density lipoprotein receptor (LDLR) protein levels represents a key strategy for the prevention and treatment. Berberine can reportedly alleviate non-alcoholic fatty liver disease (NAFLD) by increasing the LDLR expression in an ERK1/2 signaling-dependent manner of NAFLD. Studies have shown that caffeine can inhibit fat deposition in the livers of mice; however, caffeine has not been reported to alleviate NAFLD by augmenting the LDLR expression via targeting EGFR. Here, an MTT assay, western blotting, RT-qPCR, immunohistochemistry, and surface plasmon resonance (SPR) analysis were used to investigate the role of caffeine in low-density lipoprotein cholesterol (LDL-C) clearance both in vitro and in vivo. In vitro, we found that caffeine could activate the EGFR-ERK1/2 signaling pathway in HepG2 cells, leading to increased LDLR mRNA and protein expression, and this effect could be inhibited by cetuximab. The SPR assay results have indicated that caffeine may increase the LDLR expression by directly binding to the EGFR extracellular domain and activating the EGFR-ERK1/2 signaling pathway. In vivo, caffeine markedly improved fatty liver and related blood indices in ApoE KO mice with high-fat-diet-induced NAFLD. Consistent with our in vitro results, we found that caffeine could also activate EGFR-ERK1/2 signaling and promote the LDLR expression in ApoE KO mice. In summary, caffeine can enhance the LDLR expression by directly binding to EGFR and activating the EGFR-ERK1/2 signaling pathway. EGFR signaling may represent a novel target for the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Ye-Wei Huang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China. .,College of Science, Yunnan Agricultural University, Kunming, 650201, China
| | - Li-Tian Wang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China. .,College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Meng Zhang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China. .,College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Yan Nie
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China. .,College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Jin-Bo Yang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China. .,College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Wen-Luer Meng
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China. .,College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Xuan-Jun Wang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China. .,College of Science, Yunnan Agricultural University, Kunming, 650201, China
| | - Jun Sheng
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China. .,State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Kunming, 650201, China
| |
Collapse
|
33
|
Proteomics- and Metabolomics-Based Analysis of Metabolic Changes in a Swine Model of Pulmonary Hypertension. Int J Mol Sci 2023; 24:ijms24054870. [PMID: 36902298 PMCID: PMC10003314 DOI: 10.3390/ijms24054870] [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: 12/30/2022] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 03/06/2023] Open
Abstract
Pulmonary vein stenosis (PVS) causes a rare type of pulmonary hypertension (PH) by impacting the flow and pressure within the pulmonary vasculature, resulting in endothelial dysfunction and metabolic changes. A prudent line of treatment in this type of PH would be targeted therapy to relieve the pressure and reverse the flow-related changes. We used a swine model in order to mimic PH after PVS using pulmonary vein banding (PVB) of the lower lobes for 12 weeks to mimic the hemodynamic profile associated with PH and investigated the molecular alterations that provide an impetus for the development of PH. Our current study aimed to employ unbiased proteomic and metabolomic analyses on both the upper and lower lobes of the swine lung to identify regions with metabolic alterations. We detected changes in the upper lobes for the PVB animals mainly pertaining to fatty acid metabolism, reactive oxygen species (ROS) signaling and extracellular matrix (ECM) remodeling and small, albeit, significant changes in the lower lobes for purine metabolism.
Collapse
|
34
|
Essaidi-Laziosi M, Royston L, Boda B, Pérez-Rodriguez FJ, Piuz I, Hulo N, Kaiser L, Clément S, Huang S, Constant S, Tapparel C. Altered cell function and increased replication of rhinoviruses and EV-D68 in airway epithelia of asthma patients. Front Microbiol 2023; 14:1106945. [PMID: 36937308 PMCID: PMC10014885 DOI: 10.3389/fmicb.2023.1106945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/18/2023] [Indexed: 03/05/2023] Open
Abstract
Introduction Rhinovirus (RV) infections constitute one of the main triggers of asthma exacerbations and an important burden in pediatric yard. However, the mechanisms underlying this association remain poorly understood. Methods In the present study, we compared infections of in vitro reconstituted airway epithelia originating from asthmatic versus healthy donors with representative strains of RV-A major group and minor groups, RV-C, RV-B, and the respiratory enterovirus EV-D68. Results We found that viral replication was higher in tissues derived from asthmatic donors for all tested viruses. Viral receptor expression was comparable in non-infected tissues from both groups. After infection, ICAM1 and LDLR were upregulated, while CDHR3 was downregulated. Overall, these variations were related to viral replication levels. The presence of the CDHR3 asthma susceptibility allele (rs6967330) was not associated with increased RV-C replication. Regarding the tissue response, a significantly higher interferon (IFN) induction was demonstrated in infected tissues derived from asthmatic donors, which excludes a defect in IFN-response. Unbiased transcriptomic comparison of asthmatic versus control tissues revealed significant modifications, such as alterations of cilia structure and motility, in both infected and non-infected tissues. These observations were supported by a reduced mucociliary clearance and increased mucus secretion in non-infected tissues from asthmatic donors. Discussion Altogether, we demonstrated an increased permissiveness and susceptibility to RV and respiratory EV infections in HAE derived from asthmatic patients, which was associated with a global alteration in epithelial cell functions. These results unveil the mechanisms underlying the pathogenesis of asthma exacerbation and suggest interesting therapeutic targets.
Collapse
Affiliation(s)
- Manel Essaidi-Laziosi
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Léna Royston
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - Francisco Javier Pérez-Rodriguez
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Infectious Diseases, Geneva University Hospital, Geneva, Switzerland
| | - Isabelle Piuz
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Nicolas Hulo
- Service for Biomathematical and Biostatistical Analyses, Institute of Genetics and Genomics, University of Geneva, Geneva, Switzerland
| | - Laurent Kaiser
- Division of Infectious Diseases, Geneva University Hospital, Geneva, Switzerland
| | - Sophie Clément
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Song Huang
- Epithelix Sàrl, Plan les Ouates, Geneva, Switzerland
| | | | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- *Correspondence: Caroline Tapparel,
| |
Collapse
|
35
|
Sreekumar PG, Su F, Spee C, Araujo E, Nusinowitz S, Reddy ST, Kannan R. Oxidative Stress and Lipid Accumulation Augments Cell Death in LDLR-Deficient RPE Cells and Ldlr-/- Mice. Cells 2022; 12:43. [PMID: 36611838 PMCID: PMC9818299 DOI: 10.3390/cells12010043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/14/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022] Open
Abstract
Lipid peroxidation from oxidative stress is considered a major contributor to age-related macular degeneration (AMD). The retina is abundant with circulating low-density lipoproteins (LDL), which are taken up by LDL receptor (LDLR) in the RPE and Müller cells. The purpose of this study is to investigate the role of LDLR in the NaIO3-induced model of dry AMD. Confluent primary human RPE (hRPE) and LDLR-silenced ARPE-19 cells were stressed with 150 µM tert-butyl hydroperoxide (tBH) and caspase 3/7 activation was determined. WT and Ldlr-/- mice were administered NaIO3 (20 mg/kg) intravenously. On day 7, fundus imaging, OCT, ERG, and retinal thickness were measured. Histology, TUNEL, cleaved caspase 3 and lipid accumulation were assessed. Treatment of hRPE with tBH markedly decreased LDLR expression. Caspase 3/7 activation was significantly increased in LDLR-silenced ARPE-19 cells treated with tBH. In Ldlr-/- mice, NaIO3 administration resulted in significant (a) retinal thinning, (b) compromised photoreceptor function, (c) increased percentage of cleaved caspase 3 positive and apoptotic cells, and (d) increased lipid droplet accumulation in the RPE, Bruch membrane, choroid, and sclera, compared to WT mice. Our findings imply that LDLR loss leads to lipid accumulation and impaired retinal function, which may contribute to the development of AMD.
Collapse
Affiliation(s)
| | - Feng Su
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | | | - Eduardo Araujo
- Jules Stein Eye Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Steven Nusinowitz
- Jules Stein Eye Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Srinivasa T Reddy
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Ram Kannan
- Doheny Eye Institute, Pasadena, CA 91103, USA
- Jules Stein Eye Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
36
|
Liu D, Pi J, Zhang B, Zeng H, Li C, Xiao Z, Fang F, Liu M, Deng N, Wang J. Phytosterol of lotus seed core powder alleviates hypercholesterolemia by regulating gut microbiota in high-cholesterol diet-induced C57BL/6J mice. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.102279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
|
37
|
Feng Y, Sun W, Sun F, Yin G, Liang P, Chen S, Liu X, Jiang T, Zhang F. Biological Mechanisms and Related Natural Inhibitors of CD36 in Nonalcoholic Fatty Liver. Drug Des Devel Ther 2022; 16:3829-3845. [PMID: 36388082 PMCID: PMC9642071 DOI: 10.2147/dddt.s386982] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 10/25/2022] [Indexed: 07/30/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), a spectrum of liver disorders from non-alcoholic fatty liver (NAFL) to the more severe non-alcoholic steatohepatitis (NASH), is the leading etiology of chronic liver disease and its global prevalence is increasing. Hepatic steatosis, a condition marked by an abnormal buildup of triglycerides in the liver, is the precursor to NAFLD. Differentiated cluster 36 (CD36), a scavenger receptor class B protein, is a membrane receptor that recognizes multiple lipid and non-lipid ligands. It is generally agreed that CD36 contributes significantly to hepatic steatosis by taking part in fatty acid uptake as well as triglyceride storage and secretion. While there has not been any conclusive research on how CD36 inhibitors prevent NAFLD from progressing and no clinically approved CD36 inhibitors are currently available for use in NAFLD, CD36 remains a target worthy of further investigation in NAFLD. In recent years, the potential role of natural products acting through CD36 in treating non-alcoholic fatty liver disease has attracted much attention. This paper offers an overview of the pathogenesis of CD36 in NAFLD and summarizes some of the natural compounds or extracts that are currently being investigated for modulating NAFLD via CD36 or the CD36 pathway, providing an alternative approach to the development of CD36-related drugs in NAFLD.
Collapse
Affiliation(s)
- Yanan Feng
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, People’s Republic of China
| | - Wenxiu Sun
- Department of Nursing, Taishan Vocational College of Nursing, Taian, People’s Republic of China
| | - Fengcui Sun
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, People’s Republic of China
| | - Guoliang Yin
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, People’s Republic of China
| | - Pengpeng Liang
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, People’s Republic of China
| | - Suwen Chen
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, People’s Republic of China
| | - Xiangyi Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, People’s Republic of China
| | - Tongfei Jiang
- Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Fengxia Zhang
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People’s Republic of China
| |
Collapse
|
38
|
Lund NC, Kayode Y, McReynolds MR, Clemmer DC, Hudson H, Clerc I, Hong HK, Brenchley JM, Bass J, D'Aquila RT, Taylor HE. mTOR regulation of metabolism limits LPS-induced monocyte inflammatory and procoagulant responses. Commun Biol 2022; 5:878. [PMID: 36028574 PMCID: PMC9412771 DOI: 10.1038/s42003-022-03804-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 08/05/2022] [Indexed: 11/30/2022] Open
Abstract
Translocated lipopolysaccharide (LPS) activates monocytes via TLR4 and is hypothesized to increase cardiovascular disease risk in persons living with HIV. We tested whether mTOR activity supports LPS-stimulated monocyte production of pro-inflammatory cytokines and tissue factor (TF), as it propels the inflammatory response in several immune cell types besides monocytes. However, multi-omics analyses here demonstrate that mTOR activates a metabolic pathway that limits abundance of these gene products in monocytes. Treatment of primary human monocytes with catalytic mTOR inhibitors (mTORi) increased LPS-induced polyfunctional responses, including production of IL-1β, IL-6, and the pro-coagulant, TF. NF-κB-driven transcriptional activity is enhanced with LPS stimulation after mTORi treatment to increase expression of F3 (TF). Moreover, intracellular NAD+ availability is restricted due to decreased salvage pathway synthesis. These results document mTOR-mediated restraint of the LPS-induced transcriptional response in monocytes and a metabolic mechanism informing strategies to reverse enhanced risk of coagulopathy in pro-inflammatory states.
Collapse
Affiliation(s)
- Nina C Lund
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Yetunde Kayode
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Melanie R McReynolds
- Department of Biochemistry and Molecular Biology, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Deanna C Clemmer
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Hannah Hudson
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Isabelle Clerc
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Hee-Kyung Hong
- Division of Endocrinology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Jason M Brenchley
- Barrier Immunity Section, Laboratory of Viral Disease, National Institute of Allergy and Infectious Diseases, Bethesda, MD, 20892, USA
| | - Joseph Bass
- Division of Endocrinology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Richard T D'Aquila
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| | - Harry E Taylor
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
39
|
Sato I, Yamamoto S, Kakimoto M, Fujii M, Honma K, Kumazaki S, Matsui M, Nakayama H, Kirihara S, Ran S, Usui S, Shinohata R, Kitamori K, Hirohata S, Watanabe S. Suppression of nitric oxide synthase aggravates non-alcoholic steatohepatitis and atherosclerosis in SHRSP5/Dmcr rat via acceleration of abnormal lipid metabolism. Pharmacol Rep 2022; 74:669-683. [PMID: 35819592 DOI: 10.1007/s43440-022-00380-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/20/2022] [Accepted: 06/09/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) is a progressive subtype of non-alcoholic fatty liver disease (NAFLD) that is closely related to cardiovascular disease (CVD). Nitric oxide (NO) plays a critical role in the control of various biological processes. Dysfunction of the NO signaling pathway is associated with various diseases such as atherosclerosis, vascular inflammatory disease, and diabetes. Recently, it has been reported that NO is related to lipid and cholesterol metabolism. Chronic NO synthase (NOS) inhibition accelerates NAFLD by increasing hepatic lipid deposition. However, the detailed relationship between NO and abnormal lipid and cholesterol metabolism in NAFLD/NASH has not been completely explained. We aimed to determine the effects of NOS inhibition by N omega-nitro-L-arginine methyl ester hydrochloride (L-NAME), a NOS inhibitor, on NASH and CVD via lipid and cholesterol metabolism. METHODS Stroke-prone spontaneously hypertensive rats were fed a high-fat and high-cholesterol diet for 8 weeks and administered L-NAME for the last 2 weeks. Following blood and tissue sampling, biochemical analysis, histopathological staining, quantitative RT-PCR analysis, and western blotting were performed. RESULTS L-NAME markedly increased hepatic triglyceride (TG) and cholesterol levels by promoting TG synthesis and cholesterol absorption from the diet. L-NAME increased the mRNA levels of inflammatory markers and fibrotic areas in the liver. Cholesterol secretion from the liver was promoted in rats administered L-NAME, which increased serum cholesterol. L-NAME significantly increased the level of oxidative stress marker and lipid deposition in the arteries. CONCLUSIONS NOS inhibition simultaneously aggravates NASH and atherosclerosis via hepatic lipid and cholesterol metabolism.
Collapse
Affiliation(s)
- Ikumi Sato
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Shusei Yamamoto
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
- Academic Field of Health Science, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Mai Kakimoto
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Moe Fujii
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Koki Honma
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Shota Kumazaki
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Mami Matsui
- Department of Medical Technology, Faculty of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Hinako Nakayama
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Sora Kirihara
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Shang Ran
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Shinichi Usui
- Department of Pathobiological Science and Technology, School of Health Science, Faculty of Medicine, Tottori University, 86, Nishi-machi, Yonago-shi, Tottori, 683-8503, Japan
| | - Ryoko Shinohata
- Academic Field of Health Science, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Kazuya Kitamori
- Collage of Human Life and Environment, Kinjo Gakuin University, 2-1723, Omori, Moriyama-ku, Nagoya-shi, Aichi, 463-8521, Japan
| | - Satoshi Hirohata
- Academic Field of Health Science, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan
| | - Shogo Watanabe
- Academic Field of Health Science, Okayama University, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama, 700-8558, Japan.
| |
Collapse
|
40
|
Neuronal ApoE Regulates the Cell-to-Cell Transmission of α-Synuclein. Int J Mol Sci 2022; 23:ijms23158311. [PMID: 35955451 PMCID: PMC9369063 DOI: 10.3390/ijms23158311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
The presence of protein inclusions, called Lewy bodies (LBs) and Lewy neurites (LNs), in the brain is the main feature of Parkinson’s disease (PD). Recent evidence that the prion-like propagation of α-synuclein (α-syn), as a major component of LBs and LNs, plays an important role in the progression of PD has gained much attention, although the molecular mechanism remains unclear. In this study, we evaluated whether neuronal ApoE regulates the cell-to-cell transmission of α-syn and explored its molecular mechanism using in vitro and in vivo model systems. We demonstrate that neuronal ApoE deficiency attenuates both α-syn uptake and release by downregulating LRP-1 and LDLR expression and enhancing chaperone-mediated autophagy activity, respectively, thereby contributing to α-syn propagation. In addition, we observed that α-syn propagation was attenuated in ApoE knockout mice injected with pre-formed mouse α-syn fibrils. This study will help our understanding of the molecular mechanisms underlying α-syn propagation.
Collapse
|
41
|
Mutant p53, the Mevalonate Pathway and the Tumor Microenvironment Regulate Tumor Response to Statin Therapy. Cancers (Basel) 2022; 14:cancers14143500. [PMID: 35884561 PMCID: PMC9323637 DOI: 10.3390/cancers14143500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
Tumor cells have the ability to co-opt multiple metabolic pathways, enhance glucose uptake and utilize aerobic glycolysis to promote tumorigenesis, which are characteristics constituting an emerging hallmark of cancer. Mutated tumor suppressor and proto-oncogenes are frequently responsible for enhanced metabolic pathway signaling. The link between mutant p53 and the mevalonate (MVA) pathway has been implicated in the advancement of various malignancies, with tumor cells relying heavily on increased MVA signaling to fuel their rapid growth, metastatic spread and development of therapy resistance. Statin drugs inhibit HMG-CoA reductase, the pathway’s rate-limiting enzyme, and as such, have long been studied as a potential anti-cancer therapy. However, whether statins provide additional anti-cancer properties is worthy of debate. Here, we examine retrospective, prospective and pre-clinical studies involving the use of statins in various cancer types, as well as potential issues with statins’ lack of efficacy observed in clinical trials and future considerations for upcoming clinical trials.
Collapse
|
42
|
Das M, Geetha V, Zarei M, Harohally NV, Kumar G S. Modulation of obesity associated metabolic dysfunction by novel lipophilic fraction obtained from Agaricus bisporus. Life Sci 2022; 305:120779. [DOI: 10.1016/j.lfs.2022.120779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/20/2022] [Accepted: 06/30/2022] [Indexed: 11/28/2022]
|
43
|
Arya P, Bhandari U, Sharma K, Bansal P. Anti-PCSK9 monoclonal antibody attenuates high-fat diet and zymosan-induced vascular inflammation in C57BL/6 mice by modulating TLR2/NF-ƙB signaling pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:577-585. [PMID: 35911646 PMCID: PMC9282737 DOI: 10.22038/ijbms.2022.60467.13404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 05/01/2022] [Indexed: 11/05/2022]
Abstract
Objectives Excess intake of a high-fatty diet (HFD) together with zymosan administration mediates vasculitis response which leads to impaired serum lipid levels and causes arterial stiffness. In the development of new cholesterol-lowering medications, PCSK9 inhibitor (proprotein convertase subtilisin/kexin type 9) is an emerging therapeutic. The goal of the present study was to see whether anti-PCSK9 mAb1 might prevent vasculitis in C57BL/6 mice by blocking TLR2/NF-B activation in HFD and Zymosan-induced vasculitis. Materials and Methods Protein-protein molecular docking was performed to validate the binding affinity of anti-PCSK9 mAb1 against TLR2. Under the experimental study, mice were randomly allocated to the following groups: Group I: standard mice diet (30 days) + Zymosan vehicle (sterile PBS solution of 5mg/ml on 8th day); Group II: HFD (30 days) + Zymosan ( single IP dose 80 mg/kg on day 8th); Group III: HFD+Zymosan + anti-PCSK9 mAb1 (6 mg/kg, s.c. on 10th and 20th days); Group IV: HFD+Zymosan+anti-PCSK9 mAb1 (10 mg/kg, s.c. on 10th and 20th days). Results In comparison with the low dose of anti-PCSK9 mAb1 (6 mg/kg), the high dose of anti-PCSK9 mAb1 (10 mg/kg) together with HFD and Zymosan inhibited vasculitis more effectively by decreasing aortic TLR2 and NF-B levels, reducing serum TNF- and IL-6, and up-regulating liver LDLR levels, which down-regulated serum LDL-C and improved serum lipids levels. Histopathological studies showed that anti-PCSK9 mAb1 treatment reduced plaque accumulation in the aorta of mice. Conclusion These findings indicate that anti-PCSK9 mAb1 has therapeutic potential in reducing HFD and Zymosan-induced vascular inflammation.
Collapse
Affiliation(s)
- Priyanka Arya
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi - 110062, India
| | - Uma Bhandari
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi - 110062, India,Corresponding author: Uma Bhandari. Department of Pharmacology, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi - 110062, India.
| | - Kalicharan Sharma
- Department of Pharmaceutical Chemistry, SPS, DPSRU, New Delhi-110017, India
| | - Priyanka Bansal
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi - 110062, India
| |
Collapse
|
44
|
Wei J, Huan Y, Heng Z, Zhao C, Jia L, Yu Y, Gao Y. Dynamic urine proteome changes in a rat model of simvastatin-induced skeletal muscle injury. J Proteomics 2022; 254:104477. [PMID: 34990819 DOI: 10.1016/j.jprot.2021.104477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 10/19/2022]
Abstract
Statin-associated muscle symptoms (SAMS) are the main side effects of statins. Currently, there are no effective biomarkers for accurate clinical diagnosis. Urine is not subject to homeostatic control and therefore accumulates early changes, making it an ideal biomarker source. We therefore examined urine proteome changes associated with SAMS. Here, we established a SAMS rat model by intragastric intubation with simvastatin (80 mg/kg). Biochemical analyses and hematoxylin and eosin staining were used to evaluate the degree of muscle injury. The urine proteome on days 3, 6, 9 and 14 was profiled using liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). Differential proteins on day 14 of SAMS were mainly associated with glycolysis/gluconeogenesis, pyruvate metabolism, metabolism of reactive oxygen species and apoptosis, which were associated with the pathological mechanism of SAMS. Among the 14 differential proteins on day 3, Fibrinogen gamma chain (FIBG), Osteopontin (OSTP) and C-reactive protein (CRP) were associated with muscle damage, while EH domain-containing protein 1(EHD1), Cubilin (CUBN) and Fibronectin (FINC) were associated with the pathogenic mechanisms of SAMS. Our preliminary results indicated that the urine proteome can reflect early changes in the SAMS rat model, providing the potential for monitoring drug side effects in future clinical research. SIGNIFICANCE: This study demonstrate that the early muscle damage caused by simvastatin can be reflected in urinary proteins. The urine proteome also has the potential to reflect the pharmacology and toxicology of drugs in future clinical research.
Collapse
Affiliation(s)
- Jing Wei
- Clinical Research Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China; Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing 100875, China
| | - Yuhang Huan
- Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing 100875, China; Beijing Advanced Innovation Center for Genomics (ICG), Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, China
| | - Ziqi Heng
- Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing 100875, China
| | - Chenyang Zhao
- Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing 100875, China
| | - Lulu Jia
- Clinical Research Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yuncui Yu
- Clinical Research Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Youhe Gao
- Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing 100875, China.
| |
Collapse
|
45
|
Wang Y, Wang Y, Gu J, Su T, Gu X, Feng Y. The role of RNA m6A methylation in lipid metabolism. Front Endocrinol (Lausanne) 2022; 13:866116. [PMID: 36157445 PMCID: PMC9492936 DOI: 10.3389/fendo.2022.866116] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
The m6A methylation is the most numerous modification of mRNA in mammals, coordinated by RNA m6A methyltransferases, RNA m6A demethylases, and RNA m6A binding proteins. They change the RNA m6A methylation level in their specific manner. RNA m6A modification has a significant impact on lipid metabolic regulation. The "writer" METTL3/METTL14 and the "eraser" FTO can promote the accumulation of lipids in various cells by affecting the decomposition and synthesis of lipids. The "reader" YTHDF recognizes m6A methylation sites of RNA and regulates the target genes' translation. Due to this function that regulates lipid metabolism, RNA m6A methylation plays a pivotal role in metabolic diseases and makes it a great potential target for therapy.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yujie Wang
- Department of Orthopaedics, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Jiarui Gu
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Tianhong Su
- Department of Cardiology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaosong Gu
- Department of Cardiology, the Second Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Yu Feng, ; Xiaosong Gu,
| | - Yu Feng
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Yu Feng, ; Xiaosong Gu,
| |
Collapse
|
46
|
Adimulam T, Abdul N, Chuturgoon A. HepG2 liver cells treated with fumonisin B1 in galactose supplemented media have altered expression of genes and proteins known to regulate cholesterol flux. WORLD MYCOTOXIN J 2021. [DOI: 10.3920/wmj2021.2723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Fumonisin B1 (FB1) contributes to mycotoxicosis in animals and has been associated with the incidence of some cancers in humans. The effect of FB1 on lipidomic profiles, sphingolipids and cholesterol levels have been demonstrated in experimental models, however, the events leading to altered cholesterol levels are unclear. This study investigates the molecular mechanisms that regulate the effect of FB1 on cholesterol homeostasis in galactose supplemented HepG2 liver cells. Galactose supplementation is a proven method utilised to circumvent the Crabtree effect exhibited by cancer cells, which forces cancer cells to activate the mitochondria. HepG2 cells were cultured in galactose supplemented media and treated with FB1 (IC50 = 25 μM) for 6 h. Cell viability was determined using the MTT assay. Metabolic status was evaluated using ATP luciferase assay, and cholesterol regulatory transcription factors (SIRT1, SREBP-1C, LXR, LDLR, PCSK9, and ABCA1) were investigated using western blotting and qPCR. FB1 in galactose supplemented HepG2 cells increased gene expression of SIRT1 (P<0.05), SREBP-1C, LXR, and LDLR; however, PCSK9 (P<0.05) was decreased. Furthermore, protein expression of SIRT1, LXR, and LDLR was elevated upon FB1 treatment, while SREBP-1C and PCSK9 were reduced. The data provides evidence that SIRT1 reduced the expression of PCSK9 and deacetylated LXR to prevent degradation of LDLR. This could result in a dysregulated cholesterol flux, which may contribute to FB1 mediated toxicity.
Collapse
Affiliation(s)
- T. Adimulam
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | - N.S. Abdul
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
- Applied Microbial and Health Biotechnology Institute, Cape Peninsula University of Technology, Cape Town 7535, South Africa
| | - A.A. Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| |
Collapse
|
47
|
Sun H, Chen J, Sun L, Shi B, Li J. Role of Thioredoxin-Interacting Protein in Diabetic Fatty Kidney Induced by Advanced Glycation End-Products. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:11982-11991. [PMID: 34606256 DOI: 10.1021/acs.jafc.1c03559] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Advanced glycation end-products (AGEs) have been identified as the etiological factors associated with the fatty kidney. Thioredoxin-interacting protein (TXNIP) might be a mediator involved in AGE-induced fatty kidney. This study focused on investigating how TXNIP affected the AGE-mediated renal lipid deposition. In an in vivo experiment, the db/db mice injected with the lentiviral vector encoding shRNA targeting TXNIP were given the AIN-76 basal or the high-AGE diet. TXNIP-targeting siRNA-transfected human renal proximal tubular epithelial (HK-2) cells were exposed to AGE-BSA in a study in vitro. The results showed that the silencing of TXNIP reduced tubular lipid droplets and intracellular cholesterol content, as well as upregulated Insig-1 and downregulated HMGCoAR, LDLr, nSREBP-2, and SCAP in the kidneys of the db/db mice, the high-AGE-diet-fed db/db mice, and AGE-BSA-treated HK-2 cells. Furthermore, AGE-BSA enhanced SCAP-SREBP-2 complex formation while promoting their transportation to the Golgi apparatus. However, these could be inhibited by TXNIP silencing in the HK-2 cells. The above findings indicated that TXNIP knockdown mitigated the accumulation of renal tubular lipids in diabetes through the regulation of SCAP, thereby inhibiting the SCAP-SREBP-2 signaling pathway, resulting in reduced cholesterol uptake and synthesis. Therefore, TXNIP might be a potential therapeutic target to treat a diabetic fatty kidney.
Collapse
Affiliation(s)
- Hong Sun
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Juan Chen
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Lili Sun
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Bimin Shi
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Jianzhong Li
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000 China
| |
Collapse
|
48
|
Ilias AN, Ismail IS, Hamzah H, Mohd Mohidin TB, Idris MF, Ajat M. Rebaudioside A Enhances LDL Cholesterol Uptake in HepG2 Cells via Suppression of HMGCR Expression. Rep Biochem Mol Biol 2021; 10:477-487. [PMID: 34981026 PMCID: PMC8718780 DOI: 10.52547/rbmb.10.3.477] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/08/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Rebaudioside A is one of the major diterpene glycosides found in Stevia had been reported to possess anti-hyperlipidemic effects. In this study, we explore the potential cholesterol-regulating mechanisms of Rebaudioside A in the human hepatoma (HepG2) cell line in comparison with simvastatin. METHODS Cells were incubated with Rebaudioside A at several concentrations (0-10 µM) to determine the cytotoxicity by the MTT assay. Cells were treated with selected dosage (1 and 5 µM) in further experiments. Total cellular lipid was extracted by Bligh and Dyer method and subjected to quantitative colorimetric assay. To illustrate the effect of Rebaudioside A on cellular lipid droplets and low-density lipoprotein receptors, treated cells were subjected to immunofluorescence microscopy. Finally, we investigated the expression of experimental gene patterns of cells in response to treatment. RESULTS In this study, cytotoxicity of Rebaudioside A was determined at 27.72 µM. Treatment of cells with a higher concentration of Rebaudioside A promotes better hepatocellular cholesterol internalization and ameliorates cholesterol-regulating genes such as HMGCR, LDLR, and ACAT2. CONCLUSION In conclusion, our data demonstrated that Rebaudioside A is capable to regulate cholesterol levels in HepG2 cells. Hence, we proposed that Rebaudioside A offers a potential alternative to statins for atherosclerosis therapy.
Collapse
Affiliation(s)
- Amirul Nazhan Ilias
- Department of Veterinary Preclinical Science, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM, Selangor, Malaysia.
| | - Intan Safinar Ismail
- Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400 UPM, Selangor, Malaysia.
| | - Hazilawati Hamzah
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM, elangor, Malaysia.
| | - Taznim Begam Mohd Mohidin
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Mohd Faiz Idris
- Pusat Bahasa dan Pengajian Umum, Universiti Pendidikan Sultan Idris, 35900 Tanjong Malim, Perak, Malaysia.
| | - Mokrish Ajat
- Department of Veterinary Preclinical Science, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM, Selangor, Malaysia.
| |
Collapse
|
49
|
Bucher M, Montaniel KRC, Myatt L, Weintraub S, Tavori H, Maloyan A. Dyslipidemia, insulin resistance, and impairment of placental metabolism in the offspring of obese mothers. J Dev Orig Health Dis 2021; 12:738-747. [PMID: 33185172 PMCID: PMC8606174 DOI: 10.1017/s2040174420001026] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Obesity is a chronic condition associated with dyslipidemia and insulin resistance. Here, we show that the offspring of obese mothers are dyslipidemic and insulin resistant from the outset.Maternal and cord blood and placental tissues were collected following C-section at term. Patients were grouped as being normal weight (NW, BMI = 18-24.9) or obese (OB, BMI ≥ 30), and separated by fetal sex. We measured plasma lipids, insulin, and glucose in maternal and cord blood. Insulin resistance was quantified using the HOMA-IR. Placental markers of lipid and energy metabolism and relevant metabolites were measured by western blot and metabolomics, respectively.For OB women, total cholesterol was decreased in both maternal and cord blood, while HDL was decreased only in cord blood, independent of sex. In babies born to OB women, cord blood insulin and insulin resistance were increased. Placental protein expression of the energy and lipid metabolism regulators PGC1α, and SIRT3, ERRα, CPT1α, and CPT2 decreased with maternal obesity in a sex-dependent manner (P < 0.05). Metabolomics showed lower levels of acylcarnitines C16:0, C18:2, and C20:4 in OB women's placentas, suggesting a decrease in β-oxidation. Glutamine, glutamate, alpha-ketoglutarate (αKG), and 2-hydroxyglutarate (2-HG) were increased, and the glutamine-to-glutamate ratio decreased (P < 0.05), in OB placentas, suggesting induction of glutamate into αKG conversion to maintain a normal metabolic flux.Newly-born offspring of obese mothers begin their lives dyslipidemic and insulin resistant. If not inherited genetically, such major metabolic perturbations might be explained by abnormal placental metabolism with potential long-term adverse consequences for the offspring's health and wellbeing.
Collapse
Affiliation(s)
- Matthew Bucher
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, OR, USA
- Department of OB/GYN, Oregon Health & Science University, Portland, OR, USA
| | - Kim Ramil C. Montaniel
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, OR, USA
- The Graduate Program in Biomedical Sciences (PBMS), Oregon Health & Science University, Portland, OR, USA
| | - Leslie Myatt
- Department of OB/GYN, Oregon Health & Science University, Portland, OR, USA
| | - Susan Weintraub
- Department of Biochemistry, The Metabolomics Core Facility, Institutional Mass Spectrometry Laboratory, University of Texas Health, San Antonio, TX, USA
| | - Hagai Tavori
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Alina Maloyan
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, OR, USA
- The Graduate Program in Biomedical Sciences (PBMS), Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
50
|
Shi H, Santos HO, de Souza IGO, Hoilat GJ, Martins CEC, Varkaneh HK, Alkhwildi JA, Hejji AT, Almuqayyid F, Abu-Zaid A. The Effect of Raloxifene Treatment on Lipid Profile in Elderly Individuals: A Systematic Review and Meta-analysis of Randomized Clinical Trials. Clin Ther 2021; 43:297-317. [PMID: 34462124 DOI: 10.1016/j.clinthera.2021.07.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/10/2021] [Accepted: 07/22/2021] [Indexed: 02/01/2023]
Abstract
PURPOSE To perform a systematic review and meta-analysis of randomized clinical trials (RCTs) to elucidate the effects of raloxifene on the lipid profile in elderly individuals. METHODS A systematic review and meta-analysis of RCTs was performed following the PRISMA statement. Data on triglycerides (TGs), total cholesterol (TC), HDL-C, and LDL-C were extracted. Relevant publications up to October 2020 were detected through searches in the PubMed/MEDLINE, Web of Science, Scopus, and Embase databases. Changes were reported as weighted mean differences (WMDs) and 95% CIs using random-effects models. FINDINGS Nine studies were selected, with a duration of intervention ranging from 2 and 12 months and a raloxifene dose of 60 to 120 mg/d. Studies were performed in healthy individuals and in those with disorders, such as osteoporosis, type 2 diabetes, and kidney disease required long-term hemodialysis. Overall, TG (WMD, -6.50 mg/dL; 95% CI, -34.18 to 21.20 mg/eL; P = 0.646), LDL-C (WMD, -17.86 mg/dL; 95% CI, -42.44 to 6.72 mg/dL; P = 0.154), and HDL-C (WMD, 2.35 mg/dL; 95% CI, -1.14 to 5.84 mg/dL; P = 0.187) levels did not change significantly after the administration of raloxifene. In contrast, TC levels decreased after raloxifene therapy (WMD, -6.59 mg/dL; 95% CI, -13.13 to -0.05 mg/dL; P = 0.048). IMPLICATIONS Raloxifene therapy decreased TC levels but did not alter TG, HDL-C, and LDL-C concentrations in elderly individuals. Regarding the LDL-C levels, although the finding lacked statistical significance, we believe that there was a mean reduction that deserves further clinical attention as much as TC.
Collapse
Affiliation(s)
- Hong Shi
- Department of Gerontology, Lanzhou University Second Hospital Lanzhou, Gansu Province, China
| | - Heitor O Santos
- School of Medicine, Federal University of Uberlandia, Uberlandia, Minas Gerais, Brazil
| | - Ivan G O de Souza
- Universidade Salvador, Escola de Ciências da Saúde, Salvador, Bahia, Brazil
| | - Gilles Jadd Hoilat
- Department of Internal Medicine, SUNY Upstate Medical University, Syracuse, New York
| | | | - Hamed Kord Varkaneh
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Joud Amer Alkhwildi
- College of Medicine, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia
| | - Aljawhara Talal Hejji
- College of Medicine, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia
| | | | - Ahmed Abu-Zaid
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia; Department of Pharmacology, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee.
| |
Collapse
|