1
|
Ansari A, Yadav PK, Zhou L, Prakash B, Ijaz L, Christiano A, Ahmad S, Rimbert A, Hussain MM. Casz1 and Znf101/Zfp961 differentially regulate apolipoproteins A1 and B, alter plasma lipoproteins, and reduce atherosclerosis. JCI Insight 2025; 10:e182260. [PMID: 39782688 PMCID: PMC11721306 DOI: 10.1172/jci.insight.182260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 11/19/2024] [Indexed: 01/12/2025] Open
Abstract
High apolipoprotein B-containing (apoB-containing) low-density lipoproteins (LDLs) and low apoA1-containing high-density lipoproteins (HDLs) are associated with atherosclerotic cardiovascular diseases. In search of a molecular regulator that could simultaneously and reciprocally control both LDL and HDL levels, we screened a microRNA (miR) library using human hepatoma Huh-7 cells. We identified miR-541-3p that both significantly decreases apoB and increases apoA1 expression by inducing mRNA degradation of 2 different transcription factors, Znf101 and Casz1. We found that Znf101 enhances apoB expression, while Casz1 represses apoA1 expression. The hepatic knockdown of Casz1 in mice increased plasma apoA1, HDL, and cholesterol efflux capacity. The hepatic knockdown of Zfp961, an ortholog of Znf101, reduced lipogenesis and production of triglyceride-rich lipoproteins and atherosclerosis, without causing hepatic lipid accumulation. This study identifies hepatic Znf101/Zfp961 and Casz1 as potential therapeutic targets to alter plasma lipoproteins and reduce atherosclerosis without causing liver steatosis.
Collapse
Affiliation(s)
- Abulaish Ansari
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Pradeep Kumar Yadav
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Liye Zhou
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York, USA
| | - Binu Prakash
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Laraib Ijaz
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Amanda Christiano
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Sameer Ahmad
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
| | - Antoine Rimbert
- Nantes Université, CNRS, INSERM, l’institut du thorax, F-44000 Nantes, France
| | - M. Mahmood Hussain
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, New York, USA
- VA New York Harbor Healthcare System, Brooklyn, New York, USA
| |
Collapse
|
2
|
Duan F, Wu J, Chang J, Peng H, Liu Z, Liu P, Han X, Sun T, Shang D, Yang Y, Li Z, Li P, Liu Y, Zhu Y, Lv Y, Guo X, Zhao Y, An Y. Deciphering endocrine function of adipose tissue and its significant influences in obesity-related diseases caused by its dysfunction. Differentiation 2025; 141:100832. [PMID: 39709882 DOI: 10.1016/j.diff.2024.100832] [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: 10/29/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
Current research has found that adipose tissue is not only involved in energy metabolism, but also a highly active endocrine organ that secretes various adipokines, including adiponectin, leptin, resistin and apelin, which are involved in the regulation of physiology and pathology of tissues and organs throughout the body. With the yearly increasing incidence, obesity has become a risk factor for a variety of pathological changes, including inflammation and metabolic syndrome in various system (endocrine, circulatory, locomotor and central nervous system). Thus these symptoms lead to multi-organ dysfunctions, including the heart, liver, kidneys, brain and joints. An in-depth summary of the roles of adipokines in the regulation of other tissues and organs can help to provide more effective therapeutic strategies for obesity-related diseases and explore potential therapeutic targets. Therefore, this review has retrospected the endocrine function of adipose tissue under obesity and the role of dysregulated adipokine secretion in related diseases and the underlying mechanisms, in order to provide a theoretical basis for targeting adipokine-mediated systemic dysregulation.
Collapse
Affiliation(s)
- Feiyi Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Jiaoyan Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Jiayi Chang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Haoyuan Peng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Zitao Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Pengfei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Xu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Tiantian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Dandan Shang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yutian Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Zhihao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Pengkun Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yixuan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yonghao Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Yunzhi Lv
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Xiumei Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Ying Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
3
|
Valmiki S, Bredefeld C, Hussain MM. A novel mutation, Ile344Asn, in microsomal triglyceride transfer protein abolishes binding to protein disulfide isomerase. J Lipid Res 2025; 66:100725. [PMID: 39672332 PMCID: PMC11745965 DOI: 10.1016/j.jlr.2024.100725] [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: 09/30/2024] [Revised: 11/27/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024] Open
Abstract
Microsomal triglyceride transfer protein (MTP) plays crucial roles in the assembly and secretion of apolipoprotein B-containing lipoproteins and loss of function MTP variants are associated with abetalipoproteinemia, a disease characterized by the absence of these lipoproteins. MTP is a heterodimeric protein of two subunits, MTP and protein disulfide isomerase (PDI). In this study, we report a proband with abetalipoproteinemia who was monitored annually for 10 years in her third decade and had very low plasma lipids and undetectable apoB-containing lipoproteins. Genetic testing revealed biallelic variants in the MTTP gene. She has a well-documented nonsense mutation Gly865∗ that does not interact with the PDI subunit. She also has a novel missense MTP mutation, Ile344Asn. We show that this mutation abrogates lipid transfer activity in MTP and does not support apolipoprotein B secretion. This residue is present in the central α-helical domain of MTP and the substitution of Ile with Asn at this position disrupts interactions between MTP and PDI subunits. Ile344 is away from the known MTP:PDI interacting sites identified in the crystal structure of MTP suggesting that MTP:PDI interactions are more dynamic than previously envisioned. Identification of more missense mutations will enhance our understanding of the structure-function of MTP and the role of critical residues in these interactions between the two subunits. This knowledge may guide us in developing novel treatment modalities to reduce plasma lipids and atherosclerosis.
Collapse
Affiliation(s)
- Swati Valmiki
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Cindy Bredefeld
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY, USA; Department of Medicine, NYU Grossman Long Island School of Medicine, Garden City, NY, USA
| | - M Mahmood Hussain
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY, USA.
| |
Collapse
|
4
|
Liao Q, Zhang Y, Pan T, Sun Y, Liu S, Zhang Z, Li Y, Yu L, Luo Z, Xiao Y, Qi X, Jiang T, Su S, Liu S, Qi X, Li X, Damba T, Batchuluun K, Liang Y, Wei S, Zhou L. Liver knockout of MCU leads to greater dysregulation of lipid metabolism in MAFLD. Sci Rep 2024; 14:28167. [PMID: 39548134 PMCID: PMC11568211 DOI: 10.1038/s41598-024-78935-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is a common chronic condition that poses a significant threat to human health. Mitochondrial dysfunction, particularly involving the mitochondrial Ca2+ uniporter (MCU), plays a key role in its pathogenesis. This study aimed to investigate the impact of the MCU gene on hepatic lipid metabolism in mice fed a high-fat diet. Using MCU knockout and wild-type mice, subjected to either a high-fat or normal diet for 14 weeks, we observed notable Steatosis and liver weight gain in MCU-deficient mice. Liver function markers, serum triglycerides, very low-density lipoprotein (VLDL) levels, and ApoB protein expression were all significantly elevated. Mechanistic studies revealed that MCU deletion led to mitochondrial dysfunction, increased oxidative stress. These findings highlight the critical role of the MCU gene in maintaining hepatic lipid balance and suggest its potential as a therapeutic target for managing nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Qichao Liao
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yurou Zhang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Tingli Pan
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yu Sun
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Siqi Liu
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zhiwang Zhang
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yixing Li
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Lin Yu
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zupeng Luo
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yang Xiao
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xinyi Qi
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Tianyu Jiang
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Songtao Su
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Shi Liu
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xinyu Qi
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xiangling Li
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Turtushikh Damba
- School of Pharmacy, Mongolian National University of Medical Sciences, Ulan Bator, 14200, Mongolia
| | - Khongorzul Batchuluun
- Center for Research and Development of Institute of Biomedical Sciences, Mongolian National University of Medical Sciences, Ulan Bator, 14200, Mongolia
| | - Yunxiao Liang
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Suosu Wei
- Department of Scientific Cooperation of Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.
| | - Lei Zhou
- Institute of Digestive Disease, Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China.
| |
Collapse
|
5
|
Aumont-Rodrigue G, Picard C, Labonté A, Poirier J. Apolipoprotein B gene expression and regulation in relation to Alzheimer's disease pathophysiology. J Lipid Res 2024; 65:100667. [PMID: 39395793 PMCID: PMC11602985 DOI: 10.1016/j.jlr.2024.100667] [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: 04/23/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024] Open
Abstract
Apolipoprotein B (APOB), a receptor-binding protein present in cholesterol-rich lipoproteins, has been implicated in Alzheimer's disease (AD). High levels of APOB-containing low-density lipoproteins (LDL) are linked to the pathogenesis of both early-onset familial and late-onset sporadic AD. Rare coding mutations in the APOB gene are associated with familial AD, suggesting a role for APOB-bound lipoproteins in the central nervous system. This research explores APOB gene regulation across the AD spectrum using four cohorts: BRAINEAC (elderly control brains), DBCBB (controls, AD brains), ROSMAP (controls, MCI, AD brains), and ADNI (control, MCI, AD clinical subjects). APOB protein levels, measured via mass spectrometry and ELISA, positively correlated with AD pathology indices and cognition, while APOB mRNA levels showed negative correlations. Brain APOB protein levels are also correlated with cortical Aβ levels. A common coding variant in the APOB gene locus affected its expression but didn't impact AD risk or brain cholesterol concentrations, except for 24-S-hydroxycholesterol. Polymorphisms in the CYP27A1 gene, notably rs4674344, were associated with APOB protein levels. A negative correlation was observed between brain APOB gene expression and AD biomarker levels. CSF APOB correlated with Tau pathology in presymptomatic subjects, while cortical APOB was strongly associated with cortical Aβ deposition in late-stage AD. The study discusses the potential link between blood-brain barrier dysfunction and AD symptoms in relation to APOB neurobiology. Overall, APOB's involvement in lipoprotein metabolism appears to influence AD pathology across different stages of the disease.
Collapse
Affiliation(s)
- Gabriel Aumont-Rodrigue
- Douglas Mental Health University Institute, Neurosciences divison, Montréal, Québec, Canada; Centre for the Studies on Prevention of Alzheimer's Disease, Montréal, Québec, Canada; McGill University, Psychiatry department, Montréal, Québec, Canada
| | - Cynthia Picard
- Douglas Mental Health University Institute, Neurosciences divison, Montréal, Québec, Canada; Centre for the Studies on Prevention of Alzheimer's Disease, Montréal, Québec, Canada
| | - Anne Labonté
- Douglas Mental Health University Institute, Neurosciences divison, Montréal, Québec, Canada; Centre for the Studies on Prevention of Alzheimer's Disease, Montréal, Québec, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute, Neurosciences divison, Montréal, Québec, Canada; Centre for the Studies on Prevention of Alzheimer's Disease, Montréal, Québec, Canada; McGill University, Psychiatry department, Montréal, Québec, Canada.
| |
Collapse
|
6
|
Ansari A, Yadav PK, Valmiki S, Laine A, Rimbert A, Islam S, Osman I, Najafi-Shoushtari SH, Hussain MM. MicroRNA-615-3p decreases apo B expression in human liver cells. J Lipid Res 2024; 65:100659. [PMID: 39332527 PMCID: PMC11513542 DOI: 10.1016/j.jlr.2024.100659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024] Open
Abstract
Plasma lipids are mainly carried in apolipoprotein B (apoB) containing lipoproteins. High levels of these lipoproteins are associated with several metabolic diseases and lowering their plasma levels is associated with reduced incidence of atherosclerotic cardiovascular disease. MicroRNAs (miRs) are small non-coding RNAs that reduce the protein expression of their target mRNAs and are potential therapeutic agents. Here, we identified a novel miR-615-3p that interacts with human 3'-UTR of apoB mRNA, induces post-transcriptional mRNA degradation, and reduces cellular and secreted apoB100 in human hepatoma Huh-7 cells. Reducing cellular miR-615-3p levels by CRISPR-sgRNA increased cellular and secreted apoB100 indicating endogenous miR regulates apoB expression. Overexpression of miR-615-3p along with or without palmitic acid treatment decreased cellular and media apoB and increased cellular triglyceride levels without inducing endoplasmic reticulum stress. These studies have identified miR-615-3p as a negative regulator of apoB expression in human liver-derived cells. It is likely that there are more miRs that regulate apoB-containing lipoprotein assembly and secretion. Discovery of additional miRs may uncover novel mechanisms that control lipoprotein assembly and secretion.
Collapse
Affiliation(s)
- Abulaish Ansari
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA; Department of Research, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Pradeep Kumar Yadav
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA; Department of Botany, Faculty of Science, University of Allahabad, Prayagraj, India
| | - Swati Valmiki
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Antoine Laine
- Nantes Université, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Antoine Rimbert
- Nantes Université, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Shahidul Islam
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Iman Osman
- Department of Medicine, New York University Langone Medical Center, New York, NY, USA
| | - S Hani Najafi-Shoushtari
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA; Department of Research, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - M Mahmood Hussain
- Department of Foundations of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY, USA; Research Department, New York Harbor Healthcare System, Brooklyn, NY, USA.
| |
Collapse
|
7
|
Zhang L, Wang X, Chen XW. The biogenesis and transport of triglyceride-rich lipoproteins. Trends Endocrinol Metab 2024:S1043-2760(24)00196-6. [PMID: 39164120 DOI: 10.1016/j.tem.2024.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/22/2024]
Abstract
Triglyceride-rich lipoproteins (TRLs) play essential roles in human health and disease by transporting bulk lipids into the circulation. This review summarizes the fundamental mechanisms and diverse factors governing lipoprotein production, secretion, and regulation. Emphasizing the broader implications for human health, we outline the intricate landscape of lipoprotein research and highlight the potential coordination between the biogenesis and transport of TRLs in physiology, particularly the unexpected coupling of metabolic enzymes and transport machineries. Challenges and opportunities in lipoprotein biology with respect to inherited diseases and viral infections are also discussed. Further characterization of the biogenesis and transport of TRLs will advance both basic research in lipid biology and translational medicine for metabolic diseases.
Collapse
Affiliation(s)
- Linqi Zhang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China
| | - Xiao Wang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China.
| | - Xiao-Wei Chen
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China; Peking University (PKU)-Tsinghua University (THU) Joint Center for Life Sciences, Peking University, Beijing 100871, PR China.
| |
Collapse
|
8
|
Shi L, Li G, Hou N, Tu L, Li J, Luo J, Hu S. APOB and CCL17 as mediators in the protective effect of SGLT2 inhibition against myocardial infarction: Insights from proteome-wide mendelian randomization. Eur J Pharmacol 2024; 976:176619. [PMID: 38679119 DOI: 10.1016/j.ejphar.2024.176619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/16/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
AIMS Sodium-glucose cotransporter 2 (SGLT2) inhibitors offer a novel therapeutic avenue for myocardial infarction (MI). However, the exact nature of this relationship and the underlying mechanisms are not fully understood. METHODS Utilizing a two-sample Mendelian Randomization (MR) analysis, we elucidated the causal effects stemming from the inhibition of SGLT2 on MI. Then, The pool of 4907 circulating proteins within the plasma proteome were utilized to explore the mediators of SGLT2 inhibitors on MI. Protein-protein network and enrichment analysis were conducted to clarify the potential mechanism. Finally, employing MR analysis and meta-analysis techniques, we systematically assessed the causal associations between SGLT2 inhibition and coronary heart diseases (CHD). RESULTS SGLT2 inhibition (per 1 SD decrement in HbA1c) was associated with reduced risk of MI (odds ratio [OR] = 0.462, [95% CI 0.222, 0.958], P = 0.038). Among 4907 circulating proteins, we identified APOB and CCL17 which were related to both SGLT2 inhibition and MI. Mediation analysis showed evidence of the indirect effect of SGLT2 inhibition on MI through APOB (β = -0.557, 95%CI [-1.098, -0.155]) with a mediated proportion of 72%, and CCL17 (β = -0.176, 95%CI [-0.332, -0.056]) with a mediated proportion of 17%. The meta-analysis result showed that SGLT2 inhibition was associated with a lower risk of CHD. CONCLUSION Based on proteome-wide mendelian randomization, APOB and CCL17 were seen as mediators in the protective effect of SGLT2 inhibition against myocardial infarction.
Collapse
Affiliation(s)
- Lili Shi
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Gen Li
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ningxin Hou
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ling Tu
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Jun Li
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jinlan Luo
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China.
| | - Shuiqing Hu
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China; Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
9
|
Borén J, Taskinen MR, Packard CJ. Biosynthesis and Metabolism of ApoB-Containing Lipoproteins. Annu Rev Nutr 2024; 44:179-204. [PMID: 38635875 DOI: 10.1146/annurev-nutr-062222-020716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Recent advances in human genetics, together with a substantial body of epidemiological, preclinical and clinical trial evidence, strongly support a causal relationship between triglyceride-rich lipoproteins (TRLs) and atherosclerotic cardiovascular disease. Consequently, the secretion and metabolism of TRLs have a significant impact on cardiovascular health. This knowledge underscores the importance of understanding the molecular mechanisms and regulation of very-low-density lipoprotein (VLDL) and chylomicron biogenesis. Fortunately, there has been a resurgence of interest in the intracellular assembly, trafficking, degradation, and secretion of VLDL, leading to many ground-breaking molecular insights. Furthermore, the identification of molecular control mechanisms related to triglyceride metabolism has greatly advanced our understanding of the complex metabolism of TRLs. In this review, we explore recent advances in the assembly, secretion, and metabolism of TRLs. We also discuss available treatment strategies for hypertriglyceridemia.
Collapse
Affiliation(s)
- Jan Borén
- Wallenberg Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden;
| | - Marja-Riitta Taskinen
- Research Programs Unit, Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
10
|
Ting KK. John Yudkin's hypothesis: sugar is a major dietary culprit in the development of cardiovascular disease. Front Nutr 2024; 11:1407108. [PMID: 39027662 PMCID: PMC11257042 DOI: 10.3389/fnut.2024.1407108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024] Open
Abstract
To date, the risk of developing atherosclerosis has extended beyond Western countries and now affecting individuals from various ethnic backgrounds and age groups. Traditional risk factors of atherosclerosis, such as hypercholesterolemia, has been better controlled than before due to highly effective and inexpensive therapies at lowering plasma cholesterol levels. However, the role of reducing dietary cholesterol intake, as a public healthy strategy, in preventing the occurrence of cardiovascular mortalities has been recently challenged. Indeed, despite our continuous decline of dietary cholesterol intake within the last 50 years, the incidence of cardiovascular mortalities has continued to rise, thus raising the possibility that other dietary factors, such as fructose-containing sugars, are the major culprit. In the 1970s, John Yudkin first proposed that sugar was the predominant dietary factor that underlies the majority of cardiovascular mortalities, yet his hypothesis was dismissed. However, over the last 25 years substantial scientific evidence has been accumulated to support Yudkin's hypothesis. The objectives of this review are to highlight Yudkin's significant contribution to nutritional science by reviewing his hypothesis and summarizing the recent advances in our understanding of fructose metabolism. The metabolic consequences of fructose metabolism, such as fructose-induced uricemia, insulin resistance, lipoprotein hyperproduction and chronic inflammation, and how they are linked to atherosclerosis as risk factors will be discussed. Finally, the review will explore areas that warrant future research and raise important considerations that we need to evaluate when designing future studies.
Collapse
Affiliation(s)
- Kenneth K.Y. Ting
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
11
|
Sun H. Manganese ions acts as a messenger to regulate serum lipid levels. Metabol Open 2024; 22:100268. [PMID: 39011160 PMCID: PMC11247199 DOI: 10.1016/j.metop.2023.100268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 07/17/2024] Open
Affiliation(s)
- Honglin Sun
- Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100020, China
| |
Collapse
|
12
|
Xiao L, Qi L, Fu R, Nie Q, Zhang X, Luo W. A large-scale comparison of the meat quality characteristics of different chicken breeds in South China. Poult Sci 2024; 103:103740. [PMID: 38701629 PMCID: PMC11087722 DOI: 10.1016/j.psj.2024.103740] [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: 01/19/2024] [Revised: 03/26/2024] [Accepted: 04/03/2024] [Indexed: 05/05/2024] Open
Abstract
Meat quality traits are essential for producing high-quality broilers, but the genetic improvement has been limited by the complexity of measurement methods and the numerous traits involved. To systematically understand the meat quality characteristics of different broiler breeds, this study collected data on slaughter performance, skin color, fat deposition, and meat quality traits of 434 broilers from 12 different breeds in South China. The results showed that there was no significant difference in the live weight and slaughter weight of various broiler breeds at their respective market ages. Commercial broiler breeds such as Xiaobai and Huangma chickens had higher breast muscle and leg muscle rates. The skin and abdominal fat of Huangma chickens cultivated in the consumer market in South China exhibited significantly higher levels of yellowness compared to other varieties. Concerning fat traits, we observed that Wenchang chickens exhibited a strong ability to fat deposition, while the younger breeds showed lower fat deposition. Additionally, there were significant positive correlations found among different traits, including traits related to weight, traits related to fat, and skin color of different parts. Hierarchical clustering analysis revealed that fast-growing and large broiler Xiaobai chickens formed a distinct cluster based on carcass characteristics, skin color, and meat quality traits. Principal component analysis (PCA) was used to extract multiple principal components as substitutes for complex meat quality indicators, establishing a chicken meat quality evaluation model to differentiate between different breeds of chickens. At the same time, we identified 46, 22, and 20 SNP loci and their adjacent genes that were significantly associated with muscle mass traits, fat deposition, and skin color through genome-wide association studies (GWAS). The above results are helpful for systematically understanding the differences and characteristics of meat quality traits among different breeds.
Collapse
Affiliation(s)
- Liangchao Xiao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Lin Qi
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Rong Fu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Qinghua Nie
- State Key Laboratory of Livestock and Poultry Breeding, and Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China; Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Xiquan Zhang
- State Key Laboratory of Livestock and Poultry Breeding, and Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China; Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Wen Luo
- State Key Laboratory of Livestock and Poultry Breeding, and Lingnan Guangdong Laboratory of Agriculture, South China Agricultural University, Guangzhou 510642, China; Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
13
|
Liu J, Ginsberg HN, Reyes-Soffer G. Basic and translational evidence supporting the role of TM6SF2 in VLDL metabolism. Curr Opin Lipidol 2024; 35:157-161. [PMID: 38465912 PMCID: PMC11168781 DOI: 10.1097/mol.0000000000000930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
PURPOSE OF REVIEW Transmembrane 6 superfamily member 2 ( TM6SF2 ) gene was identified through exome-wide studies in 2014. A genetic variant from glutamic acid to lysine substitution at amino acid position 167 (NM_001001524.3:c.499G> A) (p.Gln167Lys/p.E167K, rs58542926) was discovered (p.E167K) to be highly associated with increased hepatic fat content and reduced levels of plasma triglycerides and LDL cholesterol. In this review, we focus on the discovery of TM6SF2 and its role in VLDL secretion pathways. Human data suggest TM6SF2 is linked to hepatic steatosis and cardiovascular disease (CVD), hence understanding its metabolic pathways is of high scientific interest. RECENT FINDINGS Since its discovery, completed research studies in cell, rodent and human models have defined the role of TM6SF2 and its links to human disease. TM6SF2 resides in the endoplasmic reticulum (ER) and the ER-Golgi interface and helps with the lipidation of nascent VLDL, the main carrier of triglycerides from the liver to the periphery. Consistent results from cells and rodents indicated that the secretion of triglycerides is reduced in carriers of the p.E167K variant or when hepatic TM6SF2 is deleted. However, data for secretion of APOB, the main protein of VLDL particles responsible for triglycerides transport, are inconsistent. SUMMARY The identification of genetic variants that are highly associated with human disease presentation should be followed by the validation and investigation into the pathways that regulate disease mechanisms. In this review, we highlight the role of TM6SF2 and its role in processing of liver triglycerides.
Collapse
Affiliation(s)
- Jing Liu
- Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | | | | |
Collapse
|
14
|
Wang Y, Li X, Huang R, Chen XW, Wang X. Apolipoprotein B Secretion Assay from Primary Hepatocytes. Bio Protoc 2024; 14:e4982. [PMID: 38737509 PMCID: PMC11082783 DOI: 10.21769/bioprotoc.4982] [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: 01/08/2024] [Revised: 03/27/2024] [Accepted: 04/01/2024] [Indexed: 05/14/2024] Open
Abstract
Apolipoprotein B (APOB) is the primary structural protein of atherogenic lipoproteins, which drive atherogenesis and thereby lead to deadly cardiovascular diseases (CVDs). Plasma levels of APOB-containing lipoproteins are tightly modulated by LDL receptor-mediated endocytic trafficking and cargo receptor-initiated exocytic route; the latter is much less well understood. This protocol aims to present an uncomplicated yet effective method for detecting APOB/lipoprotein secretion. We perform primary mouse hepatocyte isolation and culture coupled with well-established techniques such as immunoblotting for highly sensitive, specific, and semi-quantitative analysis of the lipoprotein secretion process. Its inherent simplicity facilitates ease of operation, rendering it a valuable tool widely utilized to explore the intricate landscape of cellular lipid metabolism and unravel the mechanistic complexities underlying lipoprotein-related diseases. Key features • A pipeline for the isolation and subsequent culture of mouse primary hepatocytes. • A procedure for tracking the secretion of APOB-containing lipoproteins. • A rapid and sensitive assay for detecting the APOB level based on immunoblotting.
Collapse
Affiliation(s)
- Yawei Wang
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- PKU-THU Joint Center for Life Sciences, Peking University, Beijing, China
| | - Xin Li
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Runze Huang
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Xiao-Wei Chen
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- PKU-THU Joint Center for Life Sciences, Peking University, Beijing, China
- College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Xiao Wang
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- PKU-THU Joint Center for Life Sciences, Peking University, Beijing, China
- College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
| |
Collapse
|
15
|
Nagaoka M, Sakai Y, Nakajima M, Fukami T. Role of carboxylesterase and arylacetamide deacetylase in drug metabolism, physiology, and pathology. Biochem Pharmacol 2024; 223:116128. [PMID: 38492781 DOI: 10.1016/j.bcp.2024.116128] [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: 12/01/2023] [Revised: 01/20/2024] [Accepted: 03/12/2024] [Indexed: 03/18/2024]
Abstract
Carboxylesterases (CES1 and CES2) and arylacetamide deacetylase (AADAC), which are expressed primarily in the liver and/or gastrointestinal tract, hydrolyze drugs containing ester and amide bonds in their chemical structure. These enzymes often catalyze the conversion of prodrugs, including the COVID-19 drugs remdesivir and molnupiravir, to their pharmacologically active forms. Information on the substrate specificity and inhibitory properties of these enzymes, which would be useful for drug development and toxicity avoidance, has accumulated. Recently,in vitroandin vivostudies have shown that these enzymes are involved not only in drug hydrolysis but also in lipid metabolism. CES1 and CES2 are capable of hydrolyzing triacylglycerol, and the deletion of their orthologous genes in mice has been associated with impaired lipid metabolism and hepatic steatosis. Adeno-associated virus-mediated human CES overexpression decreases hepatic triacylglycerol levels and increases fatty acid oxidation in mice. It has also been shown that overexpression of CES enzymes or AADAC in cultured cells suppresses the intracellular accumulation of triacylglycerol. Recent reports indicate that AADAC can be up- or downregulated in tumors of various organs, and its varied expression is associated with poor prognosis in patients with cancer. Thus, CES and AADAC not only determine drug efficacy and toxicity but are also involved in pathophysiology. This review summarizes recent findings on the roles of CES and AADAC in drug metabolism, physiology, and pathology.
Collapse
Affiliation(s)
- Mai Nagaoka
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yoshiyuki Sakai
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
16
|
Laamanen SE, Eloranta AM, Haapala EA, Sallinen T, Schwab U, Lakka TA. Associations of diet quality and food consumption with serum biomarkers for lipid and amino acid metabolism in Finnish children: the PANIC study. Eur J Nutr 2024; 63:623-637. [PMID: 38127151 PMCID: PMC10899368 DOI: 10.1007/s00394-023-03293-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 11/28/2023] [Indexed: 12/23/2023]
Abstract
PURPOSE To investigate the associations of overall diet quality and dietary factors with serum biomarkers for lipid and amino acid metabolism in a general population of children. METHODS We studied 194 girls and 209 boys aged 6-8 years participating in the Physical Activity and Nutrition in Children study. Food consumption was assessed by 4-day food records and diet quality was quantified by the Finnish Children Healthy Eating Index (FCHEI). Fasting serum fatty acids, amino acids, apolipoproteins, as well as lipoprotein particle sizes were analyzed with high-throughput nuclear magnetic resonance spectroscopy. Data were analyzed using linear regression adjusted for age, sex, and body fat percentage. RESULTS FCHEI was directly associated with the ratio of polyunsaturated (PUFA) to saturated fatty acids (SFA) (PUFA/SFA), the ratio of PUFA to monounsaturated fatty acids (MUFA) (PUFA/MUFA), the ratio of PUFA to total fatty acids (FA) (PUFA%), the ratio of omega-3-fatty acids to total FA (omega-3 FA%), and inversely associated with the ratio of MUFA to total FA (MUFA%), alanine, glycine, histidine and very-low density lipoprotein (VLDL) particle size. Consumption of vegetable oils and vegetable-oil-based margarine (≥ 60% fat) was directly associated with PUFA/SFA, PUFA/MUFA, PUFA%, the ratio of omega-6 FA to total FA (omega-6 FA%), and inversely associated with SFA, MUFA, SFA to total FA (SFA%), MUFA%, alanine and VLDL particle size. Consumption of high-fiber grain products directly associated with PUFA/SFA, PUFA/MUFA, omega-3 FA%, omega-6 FA%, PUFA% and inversely associated with SFA and SFA%. Fish consumption directly related to omega-3 FA and omega-3 FA%. Consumption of sugary products was directly associated with histidine and VLDL particle size. Vegetable, fruit, and berry consumption had direct associations with VLDL particle size and the ratio of apolipoprotein B to apolipoprotein A1. Consumption of low fat (< 1%) milk was directly associated with phenylalanine. A higher consumption of high-fat (≥ 1%) milk was associated with lower serum MUFA/SFA and higher SFA%. Sausage consumption was directly related to SFA% and histidine. Red meat consumption was inversely associated with glycine. CONCLUSIONS Better diet quality, higher in intake of dietary sources of unsaturated fat and fiber, and lower in sugary product intake were associated with more favorable levels of serum biomarkers for lipid and amino acid metabolism independent of adiposity. TRIAL REGISTRATION ClinicalTrials.gov: NCT01803776, registered March 3, 2013.
Collapse
Affiliation(s)
- Suvi E Laamanen
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland.
| | - Aino-Maija Eloranta
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Eero A Haapala
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Taisa Sallinen
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ursula Schwab
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Timo A Lakka
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| |
Collapse
|
17
|
Hernandez-Ono A, Zhao YP, Murray JW, Östlund C, Lee MJ, Shi A, Dauer WT, Worman HJ, Ginsberg HN, Shin JY. Dynamic regulation of hepatic lipid metabolism by torsinA and its activators. JCI Insight 2024; 9:e175328. [PMID: 38194265 PMCID: PMC10967386 DOI: 10.1172/jci.insight.175328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024] Open
Abstract
Depletion of torsinA from hepatocytes leads to reduced liver triglyceride secretion and marked hepatic steatosis. TorsinA is an atypical ATPase that lacks intrinsic activity unless it is bound to its activator, lamina-associated polypeptide 1 (LAP1) or luminal domain-like LAP1 (LULL1). We previously demonstrated that depletion of LAP1 from hepatocytes has more modest effects on liver triglyceride secretion and steatosis development than depletion of torsinA. We now show that depletion of LULL1 alone does not significantly decrease triglyceride secretion or cause steatosis. However, simultaneous depletion of both LAP1 and LULL1 leads to defective triglyceride secretion and marked steatosis similar to that observed with depletion of torsinA. Depletion of both LAP1 and torsinA from hepatocytes generated phenotypes similar to those observed with only torsinA depletion, implying that the 2 proteins act in the same pathway in liver lipid metabolism. Our results demonstrate that torsinA and its activators dynamically regulate hepatic lipid metabolism.
Collapse
Affiliation(s)
| | | | - John W. Murray
- Department of Medicine
- Columbia Center for Human Development, and
| | - Cecilia Östlund
- Department of Medicine
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Michael J. Lee
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Angsi Shi
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - William T. Dauer
- Peter O’Donnell Jr. Brain Institute
- Department of Neurology, and
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Howard J. Worman
- Department of Medicine
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | | | | |
Collapse
|
18
|
van Zwol W, van de Sluis B, Ginsberg HN, Kuivenhoven JA. VLDL Biogenesis and Secretion: It Takes a Village. Circ Res 2024; 134:226-244. [PMID: 38236950 PMCID: PMC11284300 DOI: 10.1161/circresaha.123.323284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/21/2023] [Indexed: 01/23/2024]
Abstract
The production and secretion of VLDLs (very-low-density lipoproteins) by hepatocytes has a direct impact on liver fat content, as well as the concentrations of cholesterol and triglycerides in the circulation and thus affects both liver and cardiovascular health, respectively. Importantly, insulin resistance, excess caloric intake, and lack of physical activity are associated with overproduction of VLDL, hepatic steatosis, and increased plasma levels of atherogenic lipoproteins. Cholesterol and triglycerides in remnant particles generated by VLDL lipolysis are risk factors for atherosclerotic cardiovascular disease and have garnered increasing attention over the last few decades. Presently, however, increased risk of atherosclerosis is not the only concern when considering today's cardiometabolic patients, as they often also experience hepatic steatosis, a prevalent disorder that can progress to steatohepatitis and cirrhosis. This duality of metabolic risk highlights the importance of understanding the molecular regulation of the biogenesis of VLDL, the lipoprotein that transports triglycerides and cholesterol out of the liver. Fortunately, there has been a resurgence of interest in the intracellular assembly, trafficking, degradation, and secretion of VLDL by hepatocytes, which has led to many exciting new molecular insights that are the topic of this review. Increasing our understanding of the biology of this pathway will aid to the identification of novel therapeutic targets to improve both the cardiovascular and the hepatic health of cardiometabolic patients. This review focuses, for the first time, on this duality.
Collapse
Affiliation(s)
- Willemien van Zwol
- Department of Paediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Bart van de Sluis
- Department of Paediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Henry. N. Ginsberg
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Jan Albert Kuivenhoven
- Department of Paediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
19
|
Wang Y, Feng X, Zhou W, Huang R, Hu Y, Hui H, Tian J, Wang X, Chen XW. Manganese therapy for dyslipidemia and plaque reversal in murine models. LIFE METABOLISM 2023; 2:load040. [PMID: 39872857 PMCID: PMC11749556 DOI: 10.1093/lifemeta/load040] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/09/2023] [Accepted: 10/14/2023] [Indexed: 01/30/2025]
Abstract
Precise control of circulating lipid levels is vital in both health and disease. We recently uncovered that bulk lipids, transported by lipoproteins, enter the circulation initially via the coat protein complex II (COPII) in a condensation-dependent manner. Divalent manganese, acting as a signaling messenger, selectively controls COPII condensation to regulate lipid homeostasis in vivo. Here, we present evidence for a manganese-based therapy in murine models of hypolipidemia and hyperlipidemia, aided by advanced in vivo multimodal imaging of atherosclerosis. Dietary titration of manganese supply enables tailored control of circulating lipid levels in whole animals, with no apparent toxicity. Strikingly, elevating the manganese signal through diets could not only effectively treat pathological hyperlipidemia but also further achieve significant reversal of atherosclerotic plaques. Hence, the study provides critical proof-of-principle for a novel therapy for deadly cardiovascular diseases with a potentially broad impact.
Collapse
Affiliation(s)
- Yawei Wang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, China
- PKU-THU Joint Center for Life Sciences, Peking University, Beijing 100871, China
| | - Xin Feng
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100080, China
| | - Wenjing Zhou
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, China
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Runze Huang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, China
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Yating Hu
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, China
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Hui Hui
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100080, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China
| | - Xiao Wang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, China
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Xiao-Wei Chen
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, China
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- PKU-THU Joint Center for Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
20
|
Clay R, Siddiqi S, Siddiqi SA. α-Tocopherol reduces VLDL secretion through modulation of intracellular ER-to-Golgi transport of VLDL. Can J Physiol Pharmacol 2023; 101:554-564. [PMID: 37683292 PMCID: PMC11418172 DOI: 10.1139/cjpp-2023-0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
Avoiding hepatic steatosis is crucial for preventing liver dysfunction, and one mechanism by which this is accomplished is through synchronization of the rate of very low density lipoprotein (VLDL) synthesis with its secretion. Endoplasmic reticulum (ER)-to-Golgi transport of nascent VLDL is the rate-limiting step in its secretion and is mediated by the VLDL transport vesicle (VTV). Recent in vivo studies have indicated that α-tocopherol (α-T) supplementation can reverse steatosis in nonalcoholic fatty liver disease, but its effects on hepatic lipoprotein metabolism are poorly understood. Here, we investigated the impact of α-T on hepatic VLDL synthesis, secretion, and intracellular ER-to-Golgi VLDL trafficking using an in vitro model. Pulse-chase assays using [3H]-oleic acid and 100 µmol/L α-T demonstrated a disruption of early VLDL synthesis, resulting in enhanced apolipoprotein B-100 expression, decreased expression in markers for VTV budding, ER-to-Golgi VLDL transport, and reduced VLDL secretion. Additionally, an in vitro VTV budding assay indicated a significant decrease in VTV production and VTV-Golgi fusion. Confocal imaging of lipid droplet (LD) localization revealed a decrease in overall LD retention, diminished presence of ER-associated LDs, and an increase in Golgi-level LD retention. We conclude that α-T disrupts ER-to-Golgi VLDL transport by modulating the expression of specific proteins and thus reduces VLDL secretion.
Collapse
Affiliation(s)
- Ryan Clay
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Shaila Siddiqi
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Shadab A Siddiqi
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| |
Collapse
|
21
|
Wang X, Huang R, Wang Y, Zhou W, Hu Y, Yao Y, Cheng K, Li X, Xu B, Zhang J, Xu Y, Zeng F, Zhu Y, Chen XW. Manganese regulation of COPII condensation controls circulating lipid homeostasis. Nat Cell Biol 2023; 25:1650-1663. [PMID: 37884645 DOI: 10.1038/s41556-023-01260-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/18/2023] [Indexed: 10/28/2023]
Abstract
Precise control of circulating lipids is instrumental in health and disease. Bulk lipids, carried by specialized lipoproteins, are secreted into the circulation, initially via the coat protein complex II (COPII). How the universal COPII machinery accommodates the abundant yet unconventional lipoproteins remains unclear, let alone its therapeutic translation. Here we report that COPII uses manganese-tuning, self-constrained condensation to selectively drive lipoprotein delivery and set lipid homeostasis in vivo. Serendipitously, adenovirus hijacks the condensation-based transport mechanism, thus enabling the identification of cytosolic manganese as an unexpected control signal. Manganese directly binds the inner COPII coat and enhances its condensation, thereby shifting the assembly-versus-dynamics balance of the transport machinery. Manganese can be mobilized from mitochondria stores to signal COPII, and selectively controls lipoprotein secretion with a distinctive, bell-shaped function. Consequently, dietary titration of manganese enables tailored lipid management that counters pathological dyslipidaemia and atherosclerosis, implicating a condensation-targeting strategy with broad therapeutic potential for cardio-metabolic health.
Collapse
Affiliation(s)
- Xiao Wang
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China.
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China.
- PKU-THU Joint Center for Life Sciences, Peking University, Beijing, China.
| | - Runze Huang
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Yawei Wang
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- PKU-THU Joint Center for Life Sciences, Peking University, Beijing, China
| | - Wenjing Zhou
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Yating Hu
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Yuanhang Yao
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Kunlun Cheng
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Xin Li
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Bolin Xu
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Jie Zhang
- Department of Clinical Research Center, Dazhou Hospital, Dazhou, Sichuan, China
| | - Yaowen Xu
- Department of Clinical Research Center, Dazhou Hospital, Dazhou, Sichuan, China
| | - Fanxin Zeng
- Department of Clinical Research Center, Dazhou Hospital, Dazhou, Sichuan, China
| | - Yuangang Zhu
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Xiao-Wei Chen
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China.
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China.
- PKU-THU Joint Center for Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
22
|
Kim JW, Lee JY, Oh M, Lee EW. An integrated view of lipid metabolism in ferroptosis revisited via lipidomic analysis. Exp Mol Med 2023; 55:1620-1631. [PMID: 37612411 PMCID: PMC10474074 DOI: 10.1038/s12276-023-01077-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 08/25/2023] Open
Abstract
Ferroptosis is a form of regulated cell death characterized by iron-dependent lipid peroxidation. This process contributes to cellular and tissue damage in various human diseases, such as cardiovascular diseases, neurodegeneration, liver disease, and cancer. Although polyunsaturated fatty acids (PUFAs) in membrane phospholipids are preferentially oxidized, saturated/monounsaturated fatty acids (SFAs/MUFAs) also influence lipid peroxidation and ferroptosis. In this review, we first explain how cells differentially synthesize SFA/MUFAs and PUFAs and how they control fatty acid pools via fatty acid uptake and β-oxidation, impacting ferroptosis. Furthermore, we discuss how fatty acids are stored in different lipids, such as diacyl or ether phospholipids with different head groups; triglycerides; and cholesterols. Moreover, we explain how these fatty acids are released from these molecules. In summary, we provide an integrated view of the diverse and dynamic metabolic processes in the context of ferroptosis by revisiting lipidomic studies. Thus, this review contributes to the development of therapeutic strategies for ferroptosis-related diseases.
Collapse
Affiliation(s)
- Jong Woo Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon, 34141, Korea
| | - Ji-Yoon Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
| | - Mihee Oh
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon, 34141, Korea.
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Korea.
| |
Collapse
|
23
|
Hernandez-Ono A, Zhao YP, Murray JW, Östlund C, Lee MJ, Shi A, Dauer WT, Worman HJ, Ginsberg HN, Shin JY. Functional interaction of torsinA and its activators in liver lipid metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.21.545957. [PMID: 37547008 PMCID: PMC10401926 DOI: 10.1101/2023.06.21.545957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
TorsinA is an atypical ATPase that lacks intrinsic activity unless it is bound to its activators lamina-associated polypeptide 1 (LAP1) in the perinuclear space or luminal domain-like LAP1 (LULL1) throughout the endoplasmic reticulum. However, the interaction of torsinA with LAP1 and LULL1 has not yet been shown to modulate a defined physiological process in mammals in vivo . We previously demonstrated that depletion of torsinA from mouse hepatocytes leads to reduced liver triglyceride secretion and marked steatosis, whereas depletion of LAP1 had more modest similar effects. We now show that depletion of LULL1 alone does not significantly decrease liver triglyceride secretion or cause steatosis. However, simultaneous depletion of both LAP1 and LULL1 from hepatocytes leads to defective triglyceride secretion and marked steatosis similar to that observed with depletion of torsinA. Our results demonstrate that torsinA and its activators dynamically regulate a physiological process in mammals in vivo .
Collapse
|
24
|
Alfheeaid HA, Barakat H, Althwab SA, Musa KH, Malkova D. Nutritional and Physicochemical Characteristics of Innovative High Energy and Protein Fruit- and Date-Based Bars. Foods 2023; 12:2777. [PMID: 37509869 PMCID: PMC10379009 DOI: 10.3390/foods12142777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
With the increasing global nutritional bar market, developing and formulating innovative high-energy and protein bars to compensate for nutrients using date fruits is beneficial for health-conscious individuals. The current research was undertaken to study the composition and physicochemical characteristics of innovative high-energy and high-protein bars using two combinations of Sukkari dates or fruit mixtures as a base. Fifty percent of either Sukkari date paste or dried fruit mixture (25% raisin, 12.5% fig, and 12.5% apricot) combined with other different ingredients was used to produce a date-based bar (DBB) or fruit-based bar (FBB). Proximate composition, sugar content, amino and fatty acid profiles, minerals and vitamins, phytochemicals, antioxidant activity, and visual color parameters of the DBB and the FBB were determined and statistically compared. Proximate analysis revealed higher moisture and fat content in the FBB than the DBB, while ash and crude fiber were higher in the DBB than the FBB. The protein content in the DBB and the FBB was not statistically different. Both prepared bars exuded around 376-378 kcal 100 g-1 fresh weight. Sugar profile analysis of the DBB and the FBB showed dependable changes based on date or fruit content. Fructose, glucose, and maltose contents were higher in the FBB than in the DBB, while sucrose content was higher in the DBB than in the FBB. The DBB showed significantly higher content in Ca, Cu, Fe, Zn, Mn, and Se and significantly lower content in Mg, K, and Na than the FBB, with no variation in phosphorus content. The DBB and the FBB contained both essential (EAA) and non-essential (NEAA) amino acids. The DBB scored higher Lysine, Methionine, Histidine, Threonine, Phenylalanine, Isoleucine, and Cystine contents than the FBB, while the FBB scored only higher Leucine and Valine contents than the DBB. Seventeen saturated fatty acids were identified in the DBB and the FBB, with Palmitic acid (C16:0) as the predominant fatty acid. Oleic acid (C18:1n9c) was predominant among seven determined monounsaturated fatty acids. Linoleic fatty acid (C18:2n6c) was predominant among eight identified polyunsaturated fatty acids. In addition, α-Linolenic (C18:3n3) was detected in a considerable amount. However, in both the DBB and the FBB, the content and distribution of fatty acids were not remarkably changed. Regarding phytochemicals and bioactive compounds, the FBB was significantly higher in total phenolic content (TPC), total flavonoids (TF), and total flavonols (TFL) contents and scavenging activity against DPPH and ABTS free radicals than the DBB. The DBB and the FBB showed positive a* values, indicating a reddish color. The b* values were 27.81 and 28.54 for the DBB and the FBB, respectively. The DBB is affected by the lower L* value and higher browning index (BI) to make its color brownish. Sensory evaluation data showed that panelists significantly preferred the DBB over the FBB. In conclusion, processing and comparing these bars indicated that using Sukkari dates is a nutrient-dense, convenient, economical, and better sugar alternative that helps combat the calorie content. Thus, scaling up the use of dates instead of fruits in producing high-energy and protein bars commercially is highly recommended.
Collapse
Affiliation(s)
- Hani A. Alfheeaid
- Department of Food Science and Human Nutrition, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah 51452, Saudi Arabia; (H.A.A.); (S.A.A.); (K.H.M.)
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK;
| | - Hassan Barakat
- Department of Food Science and Human Nutrition, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah 51452, Saudi Arabia; (H.A.A.); (S.A.A.); (K.H.M.)
- Food Technology Department, Faculty of Agriculture, Benha University, Moshtohor 13736, Egypt
| | - Sami A. Althwab
- Department of Food Science and Human Nutrition, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah 51452, Saudi Arabia; (H.A.A.); (S.A.A.); (K.H.M.)
| | - Khalid Hamid Musa
- Department of Food Science and Human Nutrition, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah 51452, Saudi Arabia; (H.A.A.); (S.A.A.); (K.H.M.)
| | - Dalia Malkova
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK;
| |
Collapse
|
25
|
Liu JT, Doueiry C, Jiang YL, Blaszkiewicz J, Lamprecht MP, Heslop JA, Peterson YK, Carten JD, Traktman P, Yuan Y, Khetani SR, Twal WO, Duncan SA. A human iPSC-derived hepatocyte screen identifies compounds that inhibit production of Apolipoprotein B. Commun Biol 2023; 6:452. [PMID: 37095219 PMCID: PMC10125972 DOI: 10.1038/s42003-023-04739-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 03/21/2023] [Indexed: 04/26/2023] Open
Abstract
Familial hypercholesterolemia (FH) patients suffer from excessively high levels of Low Density Lipoprotein Cholesterol (LDL-C), which can cause severe cardiovascular disease. Statins, bile acid sequestrants, PCSK9 inhibitors, and cholesterol absorption inhibitors are all inefficient at treating FH patients with homozygous LDLR gene mutations (hoFH). Drugs approved for hoFH treatment control lipoprotein production by regulating steady-state Apolipoprotein B (apoB) levels. Unfortunately, these drugs have side effects including accumulation of liver triglycerides, hepatic steatosis, and elevated liver enzyme levels. To identify safer compounds, we used an iPSC-derived hepatocyte platform to screen a structurally representative set of 10,000 small molecules from a proprietary library of 130,000 compounds. The screen revealed molecules that could reduce the secretion of apoB from cultured hepatocytes and from humanized livers in mice. These small molecules are highly effective, do not cause abnormal lipid accumulation, and share a chemical structure that is distinct from any known cholesterol lowering drug.
Collapse
Affiliation(s)
- Jui-Tung Liu
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Caren Doueiry
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Yu-Lin Jiang
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Josef Blaszkiewicz
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Mary Paige Lamprecht
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - James A Heslop
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Yuri K Peterson
- Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Juliana Debrito Carten
- Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Paula Traktman
- Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Yang Yuan
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Salman R Khetani
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | | | - Stephen A Duncan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA.
- Grùthan Biosciences LLC, Hollywood, SC, 29449, USA.
| |
Collapse
|
26
|
Ginting RP, Lee JM, Lee MW. The Influence of Ambient Temperature on Adipose Tissue Homeostasis, Metabolic Diseases and Cancers. Cells 2023; 12:cells12060881. [PMID: 36980222 PMCID: PMC10047443 DOI: 10.3390/cells12060881] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 03/14/2023] Open
Abstract
Adipose tissue is a recognized energy storage organ during excessive energy intake and an endocrine and thermoregulator, which interacts with other tissues to regulate systemic metabolism. Adipose tissue dysfunction is observed in most obese mouse models and humans. However, most studies using mouse models were conducted at room temperature (RT), where mice were chronically exposed to mild cold. In this condition, energy use is prioritized for thermogenesis to maintain body temperature in mice. It also leads to the activation of the sympathetic nervous system, followed by the activation of β-adrenergic signaling. As humans live primarily in their thermoneutral (TN) zone, RT housing for mice limits the interpretation of disease studies from mouse models to humans. Therefore, housing mice in their TN zone (~28–30 °C) can be considered to mimic humans physiologically. However, factors such as temperature ranges and TN pre-acclimatization periods should be examined to obtain reliable results. In this review, we discuss how adipose tissue responds to housing temperature and the outcomes of the TN zone in metabolic disease studies. This review highlights the critical role of TN housing in mouse models for studying adipose tissue function and human metabolic diseases.
Collapse
Affiliation(s)
- Rehna Paula Ginting
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Ji-Min Lee
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Min-Woo Lee
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Republic of Korea
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Republic of Korea
- Correspondence: ; Tel.: +82-41-413-5029
| |
Collapse
|
27
|
Awadh AA. The Role of Cytosolic Lipid Droplets in Hepatitis C Virus Replication, Assembly, and Release. BIOMED RESEARCH INTERNATIONAL 2023; 2023:5156601. [PMID: 37090186 PMCID: PMC10121354 DOI: 10.1155/2023/5156601] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 03/02/2023] [Accepted: 03/09/2023] [Indexed: 04/25/2023]
Abstract
The hepatitis C virus (HCV) causes chronic hepatitis by establishing a persistent infection. Patients with chronic hepatitis frequently develop hepatic cirrhosis, which can lead to liver cancer-the progressive liver damage results from the host's immune response to the unresolved infection. The HCV replication process, including the entry, replication, assembly, and release stages, while the virus circulates in the bloodstream, it is intricately linked to the host's lipid metabolism, including the dynamic of the cytosolic lipid droplets (cLDs). This review article depicts how this interaction regulates viral cell tropism and aids immune evasion by coining viral particle characteristics. cLDs are intracellular organelles that store most of the cytoplasmic components of neutral lipids and are assumed to play an increasingly important role in the pathophysiology of lipid metabolism and host-virus interactions. cLDs are involved in the replication of several clinically significant viruses, where viruses alter the lipidomic profiles of host cells to improve viral life cycles. cLDs are involved in almost every phase of the HCV life cycle. Indeed, pharmacological modulators of cholesterol synthesis and intracellular trafficking, lipoprotein maturation, and lipid signaling molecules inhibit the assembly of HCV virions. Likewise, small-molecule inhibitors of cLD-regulating proteins inhibit HCV replication. Thus, addressing the molecular architecture of HCV replication will aid in elucidating its pathogenesis and devising preventive interventions that impede persistent infection and prevent disease progression. This is possible via repurposing the available therapeutic agents that alter cLDs metabolism. This review highlights the role of cLD in HCV replication.
Collapse
Affiliation(s)
- Abdullah A. Awadh
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah 21423, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah 21423, Saudi Arabia
| |
Collapse
|
28
|
The Expression of Myoz1 and ApoB is Positively Correlated with Meat Quality of Broiler Chicken. Vet Med Int 2022; 2022:3266076. [PMID: 36624802 PMCID: PMC9825206 DOI: 10.1155/2022/3266076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 01/01/2023] Open
Abstract
Broiler chicken (Gallus gallus) is a source of animal protein with a high nutritional content. The purpose of this study was to evaluate the quality of broiler chicken meat (Gallus gallus) by analyzing its nutritional value, genetic profile, and protein level. The chicken meat samples were obtained from four different districts in Malang city, Indonesia. We analysed the proximate composition of chicken meat to detect its nutrition content. Furthermore, we have examined the sequence of the Myoz1 gene as well as the level of ApoB proteins in the same meat. The nutritional analysis of chicken meat showed that in the four locations different levels of protein, ash, water, and lipids were observed. The Myoz1 gene of femur chicken broilers from the second and third districts has five and twenty-one substitution mutations, respectively. The ApoB expression level in locations 2 and 3 was higher than that in the other districts. In conclusion, Myoz1 and ApoB levels were correlated with the nutritional content and quality of broiler chicken meat.
Collapse
|
29
|
Li X, Zhao F, Fu C, Yang Y, Xu Q, Hao Y, Shi X, Chen D, Bi X, Gong Z, Wu S, Zhang H. Early- and whole-life exposures to florfenicol disrupts lipid metabolism and induces obesogenic effects in zebrafish (Danio rerio). CHEMOSPHERE 2022; 308:136429. [PMID: 36115475 DOI: 10.1016/j.chemosphere.2022.136429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/13/2022] [Accepted: 09/09/2022] [Indexed: 06/15/2023]
Abstract
Florfenicol (FF), a widely used veterinary antibiotic, has been frequently detected in both aquatic environments and human body fluids. As a result, there is a growing concern on its health risks. Previous studies have revealed various toxicities of FF on animals, while there are relatively limited researches on its metabolic toxicity. Herein, by employing zebrafish as an in vivo model, endpoints at multiple levels of biological organization were measured to investigate the metabolic toxicity, especially disturbances on lipid metabolism, of this emerging pollutant. Our results indicated that early-life exposure (from 2 h past fertilization (hpf) to 15 days past fertilization (dpf)) to FF significantly increased body mass index (BMI) values, staining areas of visceral lipids, and triacylglycerol (TAG) and total cholesterol (TC) contents of larvae. Further, by analyzing expression patterns of genes encoding key proteins regulating lipid metabolism, our data suggested that promoted intestinal absorption and hepatic de novo synthesis of lipids, suppressed TAG decomposition, and inhibited FFA oxidation all contributed to TAG accumulation in larvae. Following whole-life exposure (from 2 hpf to 120 dpf), BMI values, TAG and TC contents all increased significantly in males, and significant increases of hepatic TAG levels were also observed in females. Moreover, FF exposure interfered with lipid homeostasis of males and females in a gender-specific pattern. Our study revealed the obesogenic effects of FF at environmentally relevant concentrations (1, 10, and 100 μg/L) and therefore will benefit assessment of its health risks. Additionally, our results showed that FF exposure caused a more pronounced obesogenic effect in zebrafish larvae than adults, as suggested by significant increases of all endpoints at individual, tissular, and molecular levels in larvae. Therefore, our study also advances the application of zebrafish larval model in assessing metabolic toxicity of chemicals, due to the higher susceptibility of larvae than adults.
Collapse
Affiliation(s)
- Xinhui Li
- School of Environmental and Municipal Engineering, Qingdao University of Technology, 11 Fushun Road, Qingdao 266033, PR China
| | - Fei Zhao
- School of Environmental and Municipal Engineering, Qingdao University of Technology, 11 Fushun Road, Qingdao 266033, PR China.
| | - Chen Fu
- Chengdu Academy of Environmental Sciences, Chengdu 610072, PR China
| | - Yanyu Yang
- School of Environmental and Municipal Engineering, Qingdao University of Technology, 11 Fushun Road, Qingdao 266033, PR China
| | - Qianru Xu
- School of Environmental and Municipal Engineering, Qingdao University of Technology, 11 Fushun Road, Qingdao 266033, PR China
| | - Yinfei Hao
- School of Environmental and Municipal Engineering, Qingdao University of Technology, 11 Fushun Road, Qingdao 266033, PR China
| | - Xueqing Shi
- School of Environmental and Municipal Engineering, Qingdao University of Technology, 11 Fushun Road, Qingdao 266033, PR China
| | - Dong Chen
- School of Environmental and Municipal Engineering, Qingdao University of Technology, 11 Fushun Road, Qingdao 266033, PR China
| | - Xuejun Bi
- School of Environmental and Municipal Engineering, Qingdao University of Technology, 11 Fushun Road, Qingdao 266033, PR China
| | - Zhilin Gong
- School of Environmental and Municipal Engineering, Qingdao University of Technology, 11 Fushun Road, Qingdao 266033, PR China
| | - Shujian Wu
- School of Environmental and Municipal Engineering, Qingdao University of Technology, 11 Fushun Road, Qingdao 266033, PR China
| | - Haifeng Zhang
- School of Environmental and Municipal Engineering, Qingdao University of Technology, 11 Fushun Road, Qingdao 266033, PR China
| |
Collapse
|
30
|
Zhou J, Zhang N, Aldhahrani A, Soliman MM, Zhang L, Zhou F. Puerarin ameliorates nonalcoholic fatty liver in rats by regulating hepatic lipid accumulation, oxidative stress, and inflammation. Front Immunol 2022; 13:956688. [PMID: 35958617 PMCID: PMC9359096 DOI: 10.3389/fimmu.2022.956688] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/27/2022] [Indexed: 12/22/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become one of the public health problems globally. The occurrence of NAFLD is usually accompanied by a series of chronic metabolic diseases, with a prevalence rate is 25.24% among adults worldwide. Therefore, NAFLD seriously affects the quality of life in patients and causes a large economic burden. It has been reported that puerarin has the function of lowering the serum lipids, but due to the complexity of NAFLD, the specific mechanism of action has not been clarified. The aim of this study was to evaluate the preventive or ameliorating effects of two doses of puerarin (0.11% and 0.22% in diet) on high-fat and high-fructose diet (HFFD)-induced NAFLD in rats. The rats were fed with HFFD-mixed puerarin for 20 weeks. The results showed that puerarin ameliorated the levels of lipids in the serum and liver. Further exploration of the mechanism found that puerarin ameliorated hepatic lipid accumulation in NAFLD rats by reducing the expression of Srebf1, Chrebp, Acaca, Scd1, Fasn, Acacb, Cd36, Fatp5, Degs1, Plin2, and Apob100 and upregulating the expression of Mttp, Cpt1a, and Pnpla2. At the same time, after administration of puerarin, the levels of antioxidant markers (superoxide dismutase, glutathione peroxidase, and catalase) were significantly increased in the serum and liver, and the contents of serum and hepatic inflammatory factors (interleukin-18, interleukins-1β, and tumor necrosis factor α) were clearly decreased. In addition, puerarin could ameliorate the liver function. Overall, puerarin ameliorated HFFD-induced NAFLD by modulating liver lipid accumulation, liver function, oxidative stress, and inflammation.
Collapse
Affiliation(s)
- Jingxuan Zhou
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Nanhai Zhang
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Adil Aldhahrani
- Clinical Laboratory Sciences Department, Turabah University College, Taif University, Taif, Saudi Arabia
| | - Mohamed Mohamed Soliman
- Clinical Laboratory Sciences Department, Turabah University College, Taif University, Taif, Saudi Arabia
| | - Liebing Zhang
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Feng Zhou
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- *Correspondence: Feng Zhou,
| |
Collapse
|
31
|
Formulation and Characterization of O/W Nanoemulsions of Hemp Seed Oil for Protection from Steatohepatitis: Analysis of Hepatic Free Fatty Acids and Oxidation Markers. Pharmaceuticals (Basel) 2022; 15:ph15070864. [PMID: 35890162 PMCID: PMC9316199 DOI: 10.3390/ph15070864] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 12/04/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a common type of metabolic liver disease which is characterized by fatty changes associated with hepatocyte injury, lobular inflammation, and/or liver fibrosis. Nanoemulsions are kinetically stable colloidal systems characterized by small droplet size. Hemp seed oil is a natural oil derived from Cannabis sativa seeds. The current study was designed to formulate nanoemulsion preparations of hemp seed oil with promising enhanced biological activity against high fat (HF) diet induced NASH in rats. Four nanoemulsion formulas (NEFs) were formulated based on high-pressure homogenization technique and evaluated for droplet size, zeta potential (ZP), polydispersity index (PDI), electrical conductivity, pH, and viscosity, as well as the preparation stability. The best NEF was selected to perform an in vivo rat study; selection was based on the smallest droplet size and highest physical stability. Results showed that NEF#4 showed the best physiochemical characters among the other preparations. Twenty male rats were assigned to four groups as follows: normal, NASH control, NASH + hemp seed oil and NASH + hemp seed oil NEF4. The rats were tested for body weight (BWt) change, insulin resistance (IR) and hepatic pathology. The hemp seed NEF#4 protected against NASH progression in rats and decreased the % of BWt gain compared to the original Hemp seed oil. NEF#4 of Hemp seed oil showed greater protective activity against experimental NASH and IR in rats. Hence, we can consider the nanoemulsion preparations as a useful tool for enhancing the biological action of the hemp seed oil, and further studies are warranted for application of this technique for preparing natural oils aiming at enhancing their activities.
Collapse
|
32
|
Borén J, Taskinen MR, Björnson E, Packard CJ. Metabolism of triglyceride-rich lipoproteins in health and dyslipidaemia. Nat Rev Cardiol 2022; 19:577-592. [PMID: 35318466 DOI: 10.1038/s41569-022-00676-y] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/02/2022] [Indexed: 02/07/2023]
Abstract
Accumulating evidence points to the causal role of triglyceride-rich lipoproteins and their cholesterol-enriched remnants in atherogenesis. Genetic studies in particular have not only revealed a relationship between plasma triglyceride levels and the risk of atherosclerotic cardiovascular disease, but have also identified key proteins responsible for the regulation of triglyceride transport. Kinetic studies in humans using stable isotope tracers have been especially useful in delineating the function of these proteins and revealing the hitherto unappreciated complexity of triglyceride-rich lipoprotein metabolism. Given that triglyceride is an essential energy source for mammals, triglyceride transport is regulated by numerous mechanisms that balance availability with the energy demands of the body. Ongoing investigations are focused on determining the consequences of dysregulation as a result of either dietary imprudence or genetic variation that increases the risk of atherosclerosis and pancreatitis. The identification of molecular control mechanisms involved in triglyceride metabolism has laid the groundwork for a 'precision-medicine' approach to therapy. Novel pharmacological agents under development have specific molecular targets within a regulatory framework, and their deployment heralds a new era in lipid-lowering-mediated prevention of disease. In this Review, we outline what is known about the dysregulation of triglyceride transport in human hypertriglyceridaemia.
Collapse
Affiliation(s)
- Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Marja-Riitta Taskinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Elias Björnson
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
33
|
The role of RNA binding proteins in hepatocellular carcinoma. Adv Drug Deliv Rev 2022; 182:114114. [PMID: 35063534 DOI: 10.1016/j.addr.2022.114114] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/02/2021] [Accepted: 01/12/2022] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of overall cancer deaths worldwide with limited therapeutic options. Due to the heterogeneity of HCC pathogenesis, the molecular mechanisms underlying HCC development are not fully understood. Emerging evidence indicates that RNA-binding proteins (RBPs) play a vital role throughout hepatocarcinogenesis. Thus, a deeper understanding of how RBPs contribute to HCC progression will provide new tools for early diagnosis and prognosis of this devastating disease. In this review, we summarize the tumor suppressive and oncogenic roles of RBPs and their roles in hepatocarcinogenesis. The diagnostic and therapeutic potential of RBPs in HCC, including their limitations, are also discussed.
Collapse
|
34
|
Abstract
Apolipoproteins are important structural components of plasma lipoproteins that influence vascular biology and atherosclerotic disease pathophysiology by regulating lipoprotein metabolism. Clinically important apolipoproteins related to lipid metabolism and atherogenesis include apolipoprotein B-100, apolipoprotein B-48, apolipoprotein A-I, apolipoprotein C-II, apolipoprotein C-III, apolipoprotein E and apolipoprotein(a). Apolipoprotein B-100 is the major structural component of VLDL, IDL, LDL and lipoprotein(a). Apolipoprotein B-48 is a truncated isoform of apolipoprotein B-100 that forms the backbone of chylomicrons. Apolipoprotein A-I provides the scaffolding for lipidation of HDL and has an important role in reverse cholesterol transport. Apolipoproteins C-II, apolipoprotein C-III and apolipoprotein E are involved in triglyceride-rich lipoprotein metabolism. Apolipoprotein(a) covalently binds to apolipoprotein B-100 to form lipoprotein(a). In this Review, we discuss the mechanisms by which these apolipoproteins regulate lipoprotein metabolism and thereby influence vascular biology and atherosclerotic disease. Advances in the understanding of apolipoprotein biology and their translation into therapeutic agents to reduce the risk of cardiovascular disease are also highlighted.
Collapse
|
35
|
Conlon DM, Schneider CV, Ko YA, Rodrigues A, Guo K, Hand NJ, Rader DJ. Sortilin restricts secretion of apolipoprotein B-100 by hepatocytes under stressed but not basal conditions. J Clin Invest 2022; 132:144334. [PMID: 35113816 PMCID: PMC8920325 DOI: 10.1172/jci144334] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/02/2022] [Indexed: 12/02/2022] Open
Abstract
Genetic variants at the SORT1 locus in humans, which cause increased SORT1 expression in the liver, are significantly associated with reduced plasma levels of LDL cholesterol and apolipoprotein B (apoB). However, the role of hepatic sortilin remains controversial, as genetic deletion of sortilin in mice has resulted in variable and conflicting effects on apoB secretion. Here, we found that Sort1-KO mice on a chow diet and several Sort1-deficient hepatocyte lines displayed no difference in apoB secretion. When these models were challenged with high-fat diet or ER stress, the loss of Sort1 expression resulted in a significant increase in apoB-100 secretion. Sort1-overexpression studies yielded reciprocal results. Importantly, carriers of SORT1 variant with diabetes had larger decreases in plasma apoB, TG, and VLDL and LDL particle number as compared with people without diabetes with the same variants. We conclude that, under basal nonstressed conditions, loss of sortilin has little effect on hepatocyte apoB secretion, whereas, in the setting of lipid loading or ER stress, sortilin deficiency leads to increased apoB secretion. These results are consistent with the directionality of effect in human genetics studies and suggest that, under stress conditions, hepatic sortilin directs apoB toward lysosomal degradation rather than secretion, potentially serving as a quality control step in the apoB secretion pathway in hepatocytes.
Collapse
Affiliation(s)
- Donna M Conlon
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Carolin V Schneider
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Yi-An Ko
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Amrith Rodrigues
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Kathy Guo
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Nicholas J Hand
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Daniel J Rader
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| |
Collapse
|
36
|
Ghosh SS, Wang J, Ghosh S. Measurement of In Vivo VLDL and Chylomicron Secretion. Methods Mol Biol 2022; 2455:63-71. [PMID: 35212986 DOI: 10.1007/978-1-0716-2128-8_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Intestinal lipid absorption as well as secretion of absorbed lipids as chylomicrons by the enterocytes is a direct measure of the availability of dietary lipids. Measurement of this parameter is central to the understanding of the influence of diet on plasma lipids, specifically when modulation of intestinal lipid absorption by targeted interventions is being examined. In the post-prandial state, very low-density lipoprotein (VLDL) secreted from the liver represent the major source of plasma lipids and rate of VLDL secretion reports on hepatic lipid homeostasis. Here, we describe the methods to specifically measure secretion of chylomicron and VLDL in vivo. Tight regulation of dietary lipid absorption (chylomicron secretion) and hepatic secretion of VLDL underlies the development of dyslipidemia preceding hepatic lipid accumulation seen in non-alcoholic fatty liver disease (NAFLD) and subsequent progression to non-alcoholic steatohepatitis (NASH) underscoring the importance of measurement of lipoprotein secretion in vivo.
Collapse
Affiliation(s)
- Siddhartha S Ghosh
- Department of Internal Medicine, Virginia Commonwealth University Medical Center, Richmond, VA, USA
| | - Jing Wang
- Department of Internal Medicine, Virginia Commonwealth University Medical Center, Richmond, VA, USA
| | - Shobha Ghosh
- Department of Internal Medicine, Virginia Commonwealth University Medical Center, Richmond, VA, USA.
- Hunter Holmes McGuire VA Medical Center, Richmond, VA, USA.
| |
Collapse
|
37
|
Stahel P, Xiao C, Nahmias A, Tian L, Lewis GF. Multi-organ Coordination of Lipoprotein Secretion by Hormones, Nutrients and Neural Networks. Endocr Rev 2021; 42:815-838. [PMID: 33743013 PMCID: PMC8599201 DOI: 10.1210/endrev/bnab008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Indexed: 12/15/2022]
Abstract
Plasma triglyceride-rich lipoproteins (TRL), particularly atherogenic remnant lipoproteins, contribute to atherosclerotic cardiovascular disease. Hypertriglyceridemia may arise in part from hypersecretion of TRLs by the liver and intestine. Here we focus on the complex network of hormonal, nutritional, and neuronal interorgan communication that regulates secretion of TRLs and provide our perspective on the relative importance of these factors. Hormones and peptides originating from the pancreas (insulin, glucagon), gut [glucagon-like peptide 1 (GLP-1) and 2 (GLP-2), ghrelin, cholecystokinin (CCK), peptide YY], adipose tissue (leptin, adiponectin) and brain (GLP-1) modulate TRL secretion by receptor-mediated responses and indirectly via neural networks. In addition, the gut microbiome and bile acids influence lipoprotein secretion in humans and animal models. Several nutritional factors modulate hepatic lipoprotein secretion through effects on the central nervous system. Vagal afferent signaling from the gut to the brain and efferent signals from the brain to the liver and gut are modulated by hormonal and nutritional factors to influence TRL secretion. Some of these factors have been extensively studied and shown to have robust regulatory effects whereas others are "emerging" regulators, whose significance remains to be determined. The quantitative importance of these factors relative to one another and relative to the key regulatory role of lipid availability remains largely unknown. Our understanding of the complex interorgan regulation of TRL secretion is rapidly evolving to appreciate the extensive hormonal, nutritional, and neural signals emanating not only from gut and liver but also from the brain, pancreas, and adipose tissue.
Collapse
Affiliation(s)
- Priska Stahel
- Division of Endocrinology and Metabolism, Departments of Medicine and Physiology, Banting & Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| | - Changting Xiao
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Avital Nahmias
- Division of Endocrinology and Metabolism, Departments of Medicine and Physiology, Banting & Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| | - Lili Tian
- Division of Endocrinology and Metabolism, Departments of Medicine and Physiology, Banting & Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| | - Gary Franklin Lewis
- Division of Endocrinology and Metabolism, Departments of Medicine and Physiology, Banting & Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
38
|
Apolipoprotein B and Cardiovascular Disease: Biomarker and Potential Therapeutic Target. Metabolites 2021; 11:metabo11100690. [PMID: 34677405 PMCID: PMC8540246 DOI: 10.3390/metabo11100690] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 12/19/2022] Open
Abstract
Apolipoprotein (apo) B, the critical structural protein of the atherogenic lipoproteins, has two major isoforms: apoB48 and apoB100. ApoB48 is found in chylomicrons and chylomicron remnants with one apoB48 molecule per chylomicron particle. Similarly, a single apoB100 molecule is contained per particle of very-low-density lipoprotein (VLDL), intermediate density lipoprotein, LDL and lipoprotein(a). This unique one apoB per particle ratio makes plasma apoB concentration a direct measure of the number of circulating atherogenic lipoproteins. ApoB levels indicate the atherogenic particle concentration independent of the particle cholesterol content, which is variable. While LDL, the major cholesterol-carrying serum lipoprotein, is the primary therapeutic target for management and prevention of atherosclerotic cardiovascular disease, there is strong evidence that apoB is a more accurate indicator of cardiovascular risk than either total cholesterol or LDL cholesterol. This review examines multiple aspects of apoB structure and function, with a focus on the controversy over use of apoB as a therapeutic target in clinical practice. Ongoing coronary artery disease residual risk, despite lipid-lowering treatment, has left patients and clinicians with unsatisfactory options for monitoring cardiovascular health. At the present time, the substitution of apoB for LDL-C in cardiovascular disease prevention guidelines has been deemed unjustified, but discussions continue.
Collapse
|
39
|
Chen P, Li Y, Xiao L. Berberine ameliorates nonalcoholic fatty liver disease by decreasing the liver lipid content via reversing the abnormal expression of MTTP and LDLR. Exp Ther Med 2021; 22:1109. [PMID: 34504563 PMCID: PMC8383777 DOI: 10.3892/etm.2021.10543] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 02/15/2021] [Indexed: 12/14/2022] Open
Abstract
The global incidence of nonalcoholic fatty liver disease (NAFLD) is increasing. The present study explored the effect and mechanism of berberine (BBR) on NAFLD in rats. Thirty-five Sprague-Dawley rats were randomly divided into the control and NAFLD groups, which were fed a normal diet or high-fat diet, respectively, for 8 weeks. Hematoxylin and eosin staining was performed on liver tissues and establishment of the NAFLD model was confirmed by microscopy. NAFLD rats were subsequently randomly subdivided and treated with saline or BBR for 8 weeks. The liver wet weight of rats in each group was measured, the liver tissue structure was observed by microscopy, and alanine aminotransferase (ALT), aspartate aminotransferase (AST), total cholesterol (TC), triglyceride (TG), fasting blood glucose (FBG), low-density lipoprotein (LDL) and high-density lipoprotein (HDL) levels were detected using a semi-automatic biochemical detector. Reverse transcription-quantitative PCR and western blotting were performed to determine the mRNA and protein expression levels of microsomal triglyceride transfer protein (MTTP), apolipoprotein B and low-density lipoprotein receptor (LDLR). Compared with the control group, the liver wet weight of the NAFLD rats was higher; the liver showed obvious fatty degeneration and liver TG levels increased significantly, as did serum levels of ALT, AST, TC, TG, FBG, HDL and LDL, while expression of MTTP and LDLR significantly decreased. Compared with the saline-treated NAFLD rats, the BBR-treated rats had reduced liver wet weight, improved liver steatosis and a significant decrease in liver TG levels, while ALT, AST, TC, TG, and LDL serum levels significantly decreased and MTTP levels were significantly upregulated. In conclusion, BBR treatment ameliorated the fatty liver induced by a high-fat diet in rats. Furthermore, BBR reversed the abnormal expression of MTTP and LDLR in rats with high-fat diet induced-NAFLD. The present findings suggest that fatty liver could be improved by BBR administration, via reversing the abnormal expression of MTTP and LDLR and inhibiting lipid synthesis.
Collapse
Affiliation(s)
- Ping Chen
- Department of Pharmacy, Affiliated Hospital of Shandong Medical College, Linyi, Shandong 276000, P.R. China
| | - Yusheng Li
- Department of Pharmacy, Linyi Maternal and Child Health Care Hospital, Linyi, Shandong 276000, P.R. China
| | - Li Xiao
- Department of Pharmacy, Linyi Maternal and Child Health Care Hospital, Linyi, Shandong 276000, P.R. China
| |
Collapse
|
40
|
Fang W, Chen Q, Cui K, Chen Q, Li X, Xu N, Mai K, Ai Q. Lipid overload impairs hepatic VLDL secretion via oxidative stress-mediated PKCδ-HNF4α-MTP pathway in large yellow croaker (Larimichthys crocea). Free Radic Biol Med 2021; 172:213-225. [PMID: 34116177 DOI: 10.1016/j.freeradbiomed.2021.06.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/30/2021] [Accepted: 06/02/2021] [Indexed: 02/08/2023]
Abstract
Lipid overload-induced hepatic steatosis is a major public health problem worldwide. However, the potential molecular mechanism is not completely understood. Herein, we found that high-fat diet (HFD) or oleic acid (OA) treatment induced oxidative stress which prevented the entry of hepatocyte nuclear factor 4 alpha (HNF4α) into the nucleus by activating protein kinase C delta (PKCδ) in vivo and in vitro in large yellow croaker (Larimichthys crocea). This reduced the level of microsomal triglyceride transfer protein (MTP) transcription, resulting in the impaired secretion of very-low-density lipoprotein (VLDL) and the abnormal accumulation of triglyceride (TG) in hepatocytes. Meanwhile, the detrimental effects induced by lipid overload could be partly alleviated by pretreating hepatocytes with Go6983 (PKCδ inhibitor) or N-acetylcysteine (NAC, reactive oxygen species (ROS) scavenger). In conclusion, for the first time, we revealed that lipid overload impaired hepatic VLDL secretion via oxidative stress-mediated PKCδ-HNF4α-MTP pathway in fish. This study may provide critical insights into potential intervention strategies against lipid overload-induced hepatic steatosis of fish and human beings.
Collapse
Affiliation(s)
- Wei Fang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China
| | - Qiuchi Chen
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China
| | - Kun Cui
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China
| | - Qiang Chen
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China
| | - Xueshan Li
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China
| | - Ning Xu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, 266237, Qingdao, Shandong, People's Republic of China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) and Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, 266003, Qingdao, Shandong, People's Republic of China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, 266237, Qingdao, Shandong, People's Republic of China.
| |
Collapse
|
41
|
Huang D, Xu B, Liu L, Wu L, Zhu Y, Ghanbarpour A, Wang Y, Chen FJ, Lyu J, Hu Y, Kang Y, Zhou W, Wang X, Ding W, Li X, Jiang Z, Chen J, Zhang X, Zhou H, Li JZ, Guo C, Zheng W, Zhang X, Li P, Melia T, Reinisch K, Chen XW. TMEM41B acts as an ER scramblase required for lipoprotein biogenesis and lipid homeostasis. Cell Metab 2021; 33:1655-1670.e8. [PMID: 34015269 DOI: 10.1016/j.cmet.2021.05.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/06/2021] [Accepted: 05/05/2021] [Indexed: 02/06/2023]
Abstract
How amphipathic phospholipids are shuttled between the membrane bilayer remains an essential but elusive process, particularly at the endoplasmic reticulum (ER). One prominent phospholipid shuttling process concerns the biogenesis of APOB-containing lipoproteins within the ER lumen, which may require bulk trans-bilayer movement of phospholipids from the cytoplasmic leaflet of the ER bilayer. Here, we show that TMEM41B, present in the lipoprotein export machinery, encodes a previously conceptualized ER lipid scramblase mediating trans-bilayer shuttling of bulk phospholipids. Loss of hepatic TMEM41B eliminates plasma lipids, due to complete absence of mature lipoproteins within the ER, but paradoxically also activates lipid production. Mechanistically, scramblase deficiency triggers unique ER morphological changes and unsuppressed activation of SREBPs, which potently promotes lipid synthesis despite stalled secretion. Together, this response induces full-blown nonalcoholic hepatosteatosis in the TMEM41B-deficient mice within weeks. Collectively, our data uncovered a fundamental mechanism safe-guarding ER function and integrity, dysfunction of which disrupts lipid homeostasis.
Collapse
Affiliation(s)
- Dong Huang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Bolin Xu
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Lu Liu
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Lingzhi Wu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Yuangang Zhu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Alireza Ghanbarpour
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Yawei Wang
- Center for Life Sciences, Peking University, Beijing 100871, China
| | - Feng-Jung Chen
- Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China
| | - Jia Lyu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Yating Hu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Yunlu Kang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Wenjing Zhou
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Xiao Wang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Wanqiu Ding
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Xin Li
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Zhaodi Jiang
- National Institute of Biological Sciences, Tsinghua University, Beijing 100086, China
| | - Jizheng Chen
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510503, China
| | - Xu Zhang
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, The Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongwen Zhou
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - John Zhong Li
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, The Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunguang Guo
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Wen Zheng
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Xiuqin Zhang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Peng Li
- Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China; School of Life Sciences, Tsinghua University, Beijing 100086, China
| | - Thomas Melia
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Karin Reinisch
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Xiao-Wei Chen
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China; Center for Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
42
|
Olanzapine leads to nonalcoholic fatty liver disease through the apolipoprotein A5 pathway. Biomed Pharmacother 2021; 141:111803. [PMID: 34146854 DOI: 10.1016/j.biopha.2021.111803] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/17/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
The antipsychotic drug olanzapine was reported to induce nonalcoholic fatty liver disease (NAFLD), whereas the underlying mechanism remains incompletely understood. This study investigated whether apolipoprotein A5 (apoA5) and sortilin, two interactive factors involved in NAFLD pathogenesis, are implicated in olanzapine-induced NAFLD. In our study, at week 8, olanzapine treatment successfully induced hepatic steatosis in female C57 BL/6 J mice, which was independent of body weight gain. Likewise, olanzapine effectively mediated hepatocyte steatosis in HepG2 cells characterized by substantially elevated intracellular lipid droplets. Increased plasma triglyceride concentration and decreased plasma apoA5 levels were observed in mice treated with 8-week olanzapine. Surprisingly, olanzapine markedly enhanced hepatic apoA5 protein levels in mice, without a significant effect on rodent hepatic ApoA5 mRNA. Our in vitro study showed that olanzapine reduced apoA5 protein levels in the medium and enhanced apoA5 protein levels in hepatocytes, whereas this drug exerted no effect on hepatocyte APOA5 mRNA. By transfecting APOA5 siRNA into HepG2 cells, it was demonstrated that APOA5 knockdown effectively reversed olanzapine-induced hepatocyte steatosis in vitro. In addition, olanzapine drastically increased sortilin mRNA and protein levels in vivo and in vitro. Interestingly, SORT1 knockdown reduced intracellular apoA5 protein levels and increased medium apoA5 protein levels in vitro, without affecting intracellular APOA5 mRNA levels. Furthermore, SORT1 knockdown greatly ameliorated hepatocyte steatosis in vitro. This study provides the first evidence that sortilin inhibits the hepatic apoA5 secretion that is attributable to olanzapine-induced NAFLD, which provides new insight into effective strategies against NAFLD for patients with schizophrenia administered olanzapine.
Collapse
|
43
|
Abstract
Chylomicrons and very-low-density lipoproteins (VLDLs) are large, complex cargos that may require specific chaperones for efficient transport from the ER to Golgi. In this issue of Cell Metabolism, Wang et al. (2020) identify SURF4, in coordination with SAR1B, as an essential player in COPII transport of VLDLs from ER to Golgi, suggesting that SURF4 may be a target for approaches aimed at reducing secretion of triglyceride-rich, atherogenic lipoproteins from the liver.
Collapse
Affiliation(s)
- Henry N Ginsberg
- Department of Medicine, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY, USA.
| |
Collapse
|
44
|
Receptor-Mediated ER Export of Lipoproteins Controls Lipid Homeostasis in Mice and Humans. Cell Metab 2021; 33:350-366.e7. [PMID: 33186557 DOI: 10.1016/j.cmet.2020.10.020] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 08/24/2020] [Accepted: 10/20/2020] [Indexed: 12/14/2022]
Abstract
Efficient delivery of specific cargos in vivo poses a major challenge to the secretory pathway, which shuttles products encoded by ∼30% of the genome. Newly synthesized protein and lipid cargos embark on the secretory pathway via COPII-coated vesicles, assembled by the GTPase SAR1 on the endoplasmic reticulum (ER), but how lipid-carrying lipoproteins are distinguished from the general protein cargos in the ER and selectively secreted has not been clear. Here, we show that this process is quantitatively governed by the GTPase SAR1B and SURF4, a high-efficiency cargo receptor. While both genes are implicated in lipid regulation in humans, hepatic inactivation of either mouse Sar1b or Surf4 selectively depletes plasma lipids to near-zero and protects the mice from atherosclerosis. These findings show that the pairing between SURF4 and SAR1B synergistically operates a specialized, dosage-sensitive transport program for circulating lipids, while further suggesting a potential translation to treat atherosclerosis and related cardio-metabolic diseases.
Collapse
|
45
|
Hua H, Zhang Y, Zhao F, Chen K, Wu T, Liu Q, Huang S, Zhang A, Jia Z. Celastrol inhibits intestinal lipid absorption by reprofiling the gut microbiota to attenuate high-fat diet-induced obesity. iScience 2021; 24:102077. [PMID: 33598642 PMCID: PMC7868996 DOI: 10.1016/j.isci.2021.102077] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/22/2020] [Accepted: 01/14/2021] [Indexed: 12/18/2022] Open
Abstract
Celastrol, a compound extracted from traditional Chinese medicine, has been reported as a potent anti-obesity agent with controversial mechanisms. Here both C57BL/6J and leptin-deficient (ob/ob) mice fed a high-fat diet (HFD) displayed body weight loss after celastrol therapy, opposing the previous viewpoint that celastrol improves obesity by sensitizing leptin signaling. More importantly, celastrol downregulated lipid transporters in the intestine, increased lipid excretion in feces, and reduced body weight gain in HFD mice. Meanwhile, analysis of gut microbiota revealed that celastrol altered the gut microbiota composition in HFD-fed mice, and modulating gut microbiota by antibiotics or fecal microbiota transplantation blocked the celastrol effect on intestinal lipid transport and body weight gain, suggesting a critical role of the gut microbiota composition in mediating the anti-obesity role of celastrol under HFD. Together, the findings revealed that celastrol reduces intestinal lipid absorption to antagonize obesity by resetting the gut microbiota profile under HFD feeding. Celastrol reduced intestinal lipid transporters and lipids absorption Celastrol reset gut microbiota profile to modulate intestinal lipid transport Celastrol attenuated obesity in leptin-deficient (ob/ob) mice fed high fat diet
Collapse
Affiliation(s)
- Hu Hua
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, P. R. of China
| | - Yue Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, P. R. of China
| | - Fei Zhao
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China
| | - Ke Chen
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, China.,Department of Child Health Care, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, China
| | - Tong Wu
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, China.,Department of Child Health Care, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, China
| | - Qianqi Liu
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, China.,Department of Child Health Care, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, China
| | - Songming Huang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, P. R. of China
| | - Aihua Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, P. R. of China
| | - Zhanjun Jia
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road #72, Nanjing 210008, P. R. of China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing 210029, P. R. of China
| |
Collapse
|
46
|
Soares TS, Moraes-Souza RQ, Carneiro TB, Araujo-Silva VC, Schavinski AZ, Gratão TB, Damasceno DC, Volpato GT. Maternal-fetal outcomes of exercise applied in rats with mild hyperglycemia after embryonic implantation. Birth Defects Res 2020; 113:287-298. [PMID: 33058545 DOI: 10.1002/bdr2.1818] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/21/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Exercise is commonly recommended to control hyperglycemia, including during pregnancy. We conducted this study to understand the potential benefits and risks of exercise during pregnancy of women with diabetes. Specifically, we evaluated the effects of swimming on a diabetic rat during pregnancy and assayed maternal-fetal parameters. METHODS Diabetes was induced in the female newborn from Wistar rats by the streptozotocin administration on first postnatal day. At 110 days of life, after confirm mild symptoms of diabetes, the rats were mated and randomly distributed into four experimental groups (minimum of 13 animals/group): Control (C)-nondiabetic animals without swimming; Control and Exercise (CEx)-nondiabetic animals submitted to swimming; Mild Diabetic (MD)-diabetic animals without swimming; Mild Diabetic and Exercise (MDEx)-diabetic animals submitted to swimming. The swimming program was performed from day 7 to 21 of pregnancy. Maternal parameters were evaluated during the pregnancy period. On day 21 of pregnancy, the rats were sacrificed and maternal and fetal parameters analyzed. RESULTS There are no alterations in body weight, food consumption, water intake, and reproductive outcomes among the groups. The swimming program did not normalize maternal glycemia and other biochemical biomarkers. The diabetes and exercise combination increased organ weight. The fetuses born to these exercising diabetic rats had reduced fetal weight and increased skeletal anomalies (mainly incomplete ossification of sternebra). CONCLUSION The intense swimming exercise imposed on female rats during pregnancy impaired maternal metabolic repercussions, contributing to intrauterine growth restriction and fetal skeletal anomalies.
Collapse
Affiliation(s)
- Thaigra S Soares
- Laboratory of General Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, University Center of Araguaia, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso, Brazil.,Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Univ Estadual Paulista_Unesp, Botucatu, São Paulo State, Brazil
| | - Rafaianne Q Moraes-Souza
- Laboratory of General Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, University Center of Araguaia, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso, Brazil.,Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Univ Estadual Paulista_Unesp, Botucatu, São Paulo State, Brazil
| | - Thalita B Carneiro
- Laboratory of General Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, University Center of Araguaia, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso, Brazil
| | - Vanessa C Araujo-Silva
- Laboratory of General Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, University Center of Araguaia, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso, Brazil
| | - Aline Z Schavinski
- Laboratory of General Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, University Center of Araguaia, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso, Brazil
| | - Thamires B Gratão
- Laboratory of General Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, University Center of Araguaia, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso, Brazil
| | - Débora C Damasceno
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Univ Estadual Paulista_Unesp, Botucatu, São Paulo State, Brazil
| | - Gustavo T Volpato
- Laboratory of General Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, University Center of Araguaia, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso, Brazil
| |
Collapse
|
47
|
Hu S, Gao S, Zhu J, Gan X, Chen X, He H, Liang L, Hu B, Hu J, Liu H, Han C, Kang B, Xia L, Wang J. Differential actions of diacylglycerol acyltransferase (DGAT) 1 and 2 in regulating lipid metabolism and progesterone secretion of goose granulosa cells. J Steroid Biochem Mol Biol 2020; 202:105721. [PMID: 32565248 DOI: 10.1016/j.jsbmb.2020.105721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/23/2020] [Accepted: 06/14/2020] [Indexed: 12/28/2022]
Abstract
Accumulating evidence shows that granulosa cells within both mammalian and avian ovaries have the ability to synthesize fatty acids through de novo lipogenesis and to accumulate triglycerides essential for oocyte and ovarian development. However, very little is known about the exact roles of key genes involved in the lipid metabolic pathway in granulosa cells. The goal of this study was to investigate the differential actions of diacylglycerol acyltransferase (DGAT) 1 and 2, which are recognized as the rate-limiting enzymes catalyzing the last step of triglyceride biosynthesis, in regulating lipid metabolism and steroidogenesis in granulosa cells of goose follicles at different developmental stages. It was observed that the mRNAs encoding DGAT1 and DGAT2 were ubiquitous in all examined granulosa cell layers but exhibited distinct expression profiles during follicle development. Notably, the mRNA levels of DGAT1, DGAT2, FSHR, LHR, STAR, CYP11A1, and 3βHSD remained almost constant in all except for 1-2 follicles within the 8-10 mm cohort, followed by an acute increase/decrease in the F5 follicles. At the cellular level, siRNA-mediated downregulation of DGAT1 or DGAT2 did not change the amount of lipids accumulated in both undifferentiated- and differentiated granulosa cells, while overexpression of DGAT2 promoted lipid accumulation and expression of lipogenic-related genes in these cells. Meanwhile, we found that interfering DGAT2 had no effect but interfering DGAT1 or overexpressing DGAT2 stimulated progesterone secretion in undifferentiated granulosa cells; in contrast, interference or overexpression of DGAT1/2 failed to change progesterone levels in differentiated granulosa cells but differently modulated expression of steroidogenic-related genes. Therefore, it could be concluded that DGAT1 is less efficient than DGAT2 in promoting lipid accumulation in both undifferentiated- and differentiated granulosa cells and that DGAT1 negatively while DGAT2 positively regulates progesterone production in undifferentiated granulosa cells.
Collapse
Affiliation(s)
- Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shanyan Gao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiaran Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiang Gan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xi Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Liang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Bo Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Hehe Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Chunchun Han
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Bo Kang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lu Xia
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
48
|
Pathophysiology of Type 2 Diabetes Mellitus. Int J Mol Sci 2020; 21:ijms21176275. [PMID: 32872570 PMCID: PMC7503727 DOI: 10.3390/ijms21176275] [Citation(s) in RCA: 1166] [Impact Index Per Article: 233.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023] Open
Abstract
Type 2 Diabetes Mellitus (T2DM), one of the most common metabolic disorders, is caused by a combination of two primary factors: defective insulin secretion by pancreatic β-cells and the inability of insulin-sensitive tissues to respond appropriately to insulin. Because insulin release and activity are essential processes for glucose homeostasis, the molecular mechanisms involved in the synthesis and release of insulin, as well as in its detection are tightly regulated. Defects in any of the mechanisms involved in these processes can lead to a metabolic imbalance responsible for the development of the disease. This review analyzes the key aspects of T2DM, as well as the molecular mechanisms and pathways implicated in insulin metabolism leading to T2DM and insulin resistance. For that purpose, we summarize the data gathered up until now, focusing especially on insulin synthesis, insulin release, insulin sensing and on the downstream effects on individual insulin-sensitive organs. The review also covers the pathological conditions perpetuating T2DM such as nutritional factors, physical activity, gut dysbiosis and metabolic memory. Additionally, because T2DM is associated with accelerated atherosclerosis development, we review here some of the molecular mechanisms that link T2DM and insulin resistance (IR) as well as cardiovascular risk as one of the most important complications in T2DM.
Collapse
|
49
|
Ubellacker JM, Tasdogan A, Ramesh V, Shen B, Mitchell EC, Martin-Sandoval MS, Gu Z, McCormick ML, Durham AB, Spitz DR, Zhao Z, Mathews TP, Morrison SJ. Lymph protects metastasizing melanoma cells from ferroptosis. Nature 2020; 585:113-118. [PMID: 32814895 PMCID: PMC7484468 DOI: 10.1038/s41586-020-2623-z] [Citation(s) in RCA: 553] [Impact Index Per Article: 110.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 07/08/2020] [Indexed: 02/07/2023]
Abstract
Cancer cells, including melanoma, often metastasize regionally through lymphatics before metastasizing systemically through the blood1–4; however, the reason for this is unclear. Here we show that melanoma cells in lymph experience less oxidative stress and form more metastases than melanoma cells in blood. Immunocompromised mice with patient-derived melanomas and immunocompetent mice with mouse melanomas had more melanoma cells per microliter of tumor-draining lymph than tumor-draining blood. Cells metastasizing through blood, but not lymph, became dependent on the ferroptosis inhibitor GPX4. Cells pre-treated with chemical ferroptosis inhibitors formed more metastases than untreated cells after intravenous, but not intralymphatic, injection. We observed multiple differences between lymph fluid and blood plasma that may contribute to decreased oxidative stress and ferroptosis in lymph, including higher levels of glutathione and oleic acid, and less free iron, in lymph. Oleic acid protected melanoma cells from ferroptosis in an Acsl3-dependent manner and increased their capacity to form metastatic tumors. Melanoma cells from lymph nodes were more resistant to ferroptosis and formed more metastases after intravenous injection than melanoma cells from subcutaneous tumors. Exposure to the lymphatic environment thus protects melanoma cells from ferroptosis and increases their ability to survive during subsequent metastasis through the blood. Lymph protects melanoma cells from the oxidative stress and ferroptotic cell death that occurs in the blood during metastasis.
Collapse
Affiliation(s)
- Jessalyn M Ubellacker
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alpaslan Tasdogan
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vijayashree Ramesh
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bo Shen
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Evann C Mitchell
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Misty S Martin-Sandoval
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhimin Gu
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael L McCormick
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Alison B Durham
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Zhiyu Zhao
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas P Mathews
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sean J Morrison
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
50
|
Tan SY, Little HC, Sarver DC, Watkins PA, Wong GW. CTRP12 inhibits triglyceride synthesis and export in hepatocytes by suppressing HNF-4α and DGAT2 expression. FEBS Lett 2020; 594:3227-3239. [PMID: 32749667 DOI: 10.1002/1873-3468.13895] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/21/2020] [Accepted: 07/25/2020] [Indexed: 12/15/2022]
Abstract
C1q/TNF-related protein 12 (CTRP12) is an antidiabetic adipokine whose circulating levels are reduced in obesity and diabetes. Although partial and complete loss-of-function mouse models suggest a role for CTRP12 in modulating lipid metabolism and adiposity, its effect on cellular lipid metabolism remains poorly defined. Here, we demonstrate a direct action of CTRP12 in regulating lipid synthesis and secretion. In hepatoma cells and primary mouse hepatocytes, CTRP12 treatment inhibits triglyceride synthesis by suppressing glycerophosphate acyltransferase (GPAT) and diacylglycerol acyltransferase (DGAT) expression. CTRP12 treatment also downregulates the expression of hepatocyte nuclear factor-4α (HNF-4α) and its target gene microsomal triglyceride transfer protein (MTTP), leading to reduced very-low-density lipoprotein (VLDL)-triglyceride export from hepatocytes. Consistent with the in vitro findings, overexpressing CTRP12 lowers fasting and postprandial serum triglyceride levels in mice. These results underscore the important function of CTRP12 in lipid metabolism in hepatocytes.
Collapse
Affiliation(s)
- Stefanie Y Tan
- Department of Physiology and Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Pfizer, 1 Portland St., Cambridge, MA, 02139, USA
| | - Hannah C Little
- Department of Physiology and Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dylan C Sarver
- Department of Physiology and Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul A Watkins
- Department of Neurology and Biological Chemistry, Johns Hopkins University School of Medicine, and Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
| | - G William Wong
- Department of Physiology and Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|