1
|
Zhang W, Xu Y, Fang Y, Li M, Li D, Guo H, Li H, He J, Miao L. Ubiquitination in lipid metabolism reprogramming: implications for pediatric solid tumors. Front Immunol 2025; 16:1554311. [PMID: 40370434 PMCID: PMC12075147 DOI: 10.3389/fimmu.2025.1554311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
Pediatric solid tumors represent a significant subset of childhood cancers, accounting for approximately 60% of new diagnoses. Despite advancements in therapeutic strategies, survival rates remain markedly disparate between high-income and resource-limited settings, underscoring the urgent need for novel and effective treatments. Lipid metabolic reprogramming is a fundamental hallmark of cancer, driving tumor progression, therapeutic resistance, and immune evasion through enhanced fatty acid uptake, increased de novo lipid synthesis, and activated fatty acid β-oxidation (FAO). Ubiquitination, a dynamic post-translational modification mediated by the ubiquitin-proteasome system (UPS), plays a crucial role in regulating lipid metabolism by modulating the stability and activity of key metabolic enzymes and transporters involved in cholesterol and fatty acid pathways. This review comprehensively examines the complex interplay between ubiquitination and lipid metabolic reprogramming in pediatric solid tumors. It delineates the mechanisms by which ubiquitination influences cholesterol biosynthesis, uptake, efflux, and fatty acid synthesis and oxidation, thereby facilitating tumor growth and survival. Furthermore, the review identifies potential UPS-mediated therapeutic targets and explores the feasibility of integrating ubiquitination-based strategies with existing treatments. By targeting the UPS to disrupt lipid metabolism pathways, novel therapeutic avenues may emerge to enhance treatment efficacy and overcome resistance in pediatric oncology. This synthesis of current knowledge aims to provide a foundation for the development of innovative, precision medicine approaches to improve clinical outcomes for children afflicted with solid tumors.
Collapse
Affiliation(s)
- Weixin Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Yile Xu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Yingjin Fang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Precision Medicine for Pancreatic Cancer, Shanghai, China
| | - Meng Li
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Di Li
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Huiqin Guo
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Hang Li
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Lei Miao
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Shao WQ, Li YT, Zhou X, Zhang SG, Fan MH, Zhang D, Chen ZM, Yi CH, Wang SH, Zhu WW, Lu M, Chen JS, Lin J, Zhou Y. Cholesterol suppresses AMFR-mediated PDL1 ubiquitination and degradation in HCC. Mol Cell Biochem 2025; 480:1807-1818. [PMID: 39231894 PMCID: PMC11842428 DOI: 10.1007/s11010-024-05106-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
The degradation of proteasomes or lysosomes is emerging as a principal determinant of programmed death ligand 1 (PDL1) expression, which affects the efficacy of immunotherapy in various malignancies. Intracellular cholesterol plays a central role in maintaining the expression of membrane receptors; however, the specific effect of cholesterol on PDL1 expression in cancer cells remains poorly understood. Cholesterol starvation and stimulation were used to modulate the cellular cholesterol levels. Immunohistochemistry and western blotting were used to analyze the protein levels in the samples and cells. Quantitative real-time PCR, co-immunoprecipitation, and confocal co-localization assays were used for mechanistic investigation. A xenograft tumor model was constructed to verify these results in vivo. Our results showed that cholesterol suppressed the ubiquitination and degradation of PDL1 in hepatocellular carcinoma (HCC) cells. Further mechanistic studies revealed that the autocrine motility factor receptor (AMFR) is an E3 ligase that mediated the ubiquitination and degradation of PDL1, which was regulated by the cholesterol/p38 mitogenic activated protein kinase axis. Moreover, lowering cholesterol levels using statins improved the efficacy of programmed death 1 (PD1) inhibition in vivo. Our findings indicate that cholesterol serves as a signal to inhibit AMFR-mediated ubiquitination and degradation of PDL1 and suggest that lowering cholesterol by statins may be a promising combination strategy to improve the efficiency of PD1 inhibition in HCC.
Collapse
Affiliation(s)
- Wei-Qing Shao
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
| | - Yi-Tong Li
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
| | - Xu Zhou
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
| | - Sheng-Guo Zhang
- Department of Infection, The Third Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325000, China
| | - Ming-Hao Fan
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
| | - Dong Zhang
- Department of Infection, The Third Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325000, China
| | - Zhen-Mei Chen
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
| | - Chen-He Yi
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
| | - Sheng-Hao Wang
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
| | - Wen-Wei Zhu
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China
| | - Ming Lu
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ji-Song Chen
- Depatment of Hepatobiliary Surgery, Taizhou Fourth People's Hospital, Jiangsu, 214527, China
| | - Jing Lin
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, 200040, China.
| | - Yu Zhou
- Department of Infection, The Third Affiliated Hospital of Wenzhou Medical University, Zhejiang, 325000, China.
| |
Collapse
|
3
|
Yi SA, Sepic S, Schulman BA, Ordureau A, An H. mTORC1-CTLH E3 ligase regulates the degradation of HMG-CoA synthase 1 through the Pro/N-degron pathway. Mol Cell 2024; 84:2166-2184.e9. [PMID: 38788716 PMCID: PMC11186538 DOI: 10.1016/j.molcel.2024.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/15/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024]
Abstract
Mammalian target of rapamycin (mTOR) senses changes in nutrient status and stimulates the autophagic process to recycle amino acids. However, the impact of nutrient stress on protein degradation beyond autophagic turnover is incompletely understood. We report that several metabolic enzymes are proteasomal targets regulated by mTOR activity based on comparative proteome degradation analysis. In particular, 3-hydroxy-3-methylglutaryl (HMG)-coenzyme A (CoA) synthase 1 (HMGCS1), the initial enzyme in the mevalonate pathway, exhibits the most significant half-life adaptation. Degradation of HMGCS1 is regulated by the C-terminal to LisH (CTLH) E3 ligase through the Pro/N-degron motif. HMGCS1 is ubiquitylated on two C-terminal lysines during mTORC1 inhibition, and efficient degradation of HMGCS1 in cells requires a muskelin adaptor. Importantly, modulating HMGCS1 abundance has a dose-dependent impact on cell proliferation, which is restored by adding a mevalonate intermediate. Overall, our unbiased degradomics study provides new insights into mTORC1 function in cellular metabolism: mTORC1 regulates the stability of limiting metabolic enzymes through the ubiquitin system.
Collapse
Affiliation(s)
- Sang Ah Yi
- Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sara Sepic
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany; Technical University of Munich, School of Natural Sciences, Munich, Germany
| | - Brenda A Schulman
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany; Technical University of Munich, School of Natural Sciences, Munich, Germany; Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Alban Ordureau
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Heeseon An
- Chemical Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Tri-Institutional PhD Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
4
|
Fenton NM, Qian L, Scott NA, Paine EG, Sharpe LJ, Brown AJ. SC5D is the sixth enzyme in cholesterol biosynthesis targeted by the E3 ubiquitin ligase MARCHF6. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159482. [PMID: 38508300 DOI: 10.1016/j.bbalip.2024.159482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/17/2024] [Indexed: 03/22/2024]
Affiliation(s)
- Nicole M Fenton
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Lydia Qian
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Nicola A Scott
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Eloise G Paine
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Laura J Sharpe
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Andrew J Brown
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
5
|
Zuo FW, Liu ZY, Wang MW, Du JY, Ding PZ, Zhang HR, Tang W, Sun Y, Wang XJ, Zhang Y, Xie YS, Wu JC, Liu M, Wang ZY, Yi F. CCDC92 promotes podocyte injury by regulating PA28α/ABCA1/cholesterol efflux axis in type 2 diabetic mice. Acta Pharmacol Sin 2024; 45:1019-1031. [PMID: 38228909 PMCID: PMC11053164 DOI: 10.1038/s41401-023-01213-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/07/2023] [Indexed: 01/18/2024]
Abstract
Podocyte lipotoxicity mediated by impaired cellular cholesterol efflux plays a crucial role in the development of diabetic kidney disease (DKD), and the identification of potential therapeutic targets that regulate podocyte cholesterol homeostasis has clinical significance. Coiled-coil domain containing 92 (CCDC92) is a novel molecule related to metabolic disorders and insulin resistance. However, whether the expression level of CCDC92 is changed in kidney parenchymal cells and the role of CCDC92 in podocytes remain unclear. In this study, we found that Ccdc92 was significantly induced in glomeruli from type 2 diabetic mice, especially in podocytes. Importantly, upregulation of Ccdc92 in glomeruli was positively correlated with an increased urine albumin-to-creatinine ratio (UACR) and podocyte loss. Functionally, podocyte-specific deletion of Ccdc92 attenuated proteinuria, glomerular expansion and podocyte injury in mice with DKD. We further demonstrated that Ccdc92 contributed to lipid accumulation by inhibiting cholesterol efflux, finally promoting podocyte injury. Mechanistically, Ccdc92 promoted the degradation of ABCA1 by regulating PA28α-mediated proteasome activity and then reduced cholesterol efflux. Thus, our studies indicate that Ccdc92 contributes to podocyte injury by regulating the PA28α/ABCA1/cholesterol efflux axis in DKD.
Collapse
Affiliation(s)
- Fu-Wen Zuo
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Zhi-Yong Liu
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Ming-Wei Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Jun-Yao Du
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Peng-Zhong Ding
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Hao-Ran Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Wei Tang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Yu Sun
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Xiao-Jie Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Yan Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Yu-Sheng Xie
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Ji-Chao Wu
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Min Liu
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China.
| | - Zi-Ying Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China.
| | - Fan Yi
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China.
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, 250012, China.
| |
Collapse
|
6
|
Loix M, Zelcer N, Bogie JFJ, Hendriks JJA. The ubiquitous role of ubiquitination in lipid metabolism. Trends Cell Biol 2024; 34:416-429. [PMID: 37770289 DOI: 10.1016/j.tcb.2023.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/17/2023] [Accepted: 09/01/2023] [Indexed: 09/30/2023]
Abstract
Lipids are essential molecules that play key roles in cell physiology by serving as structural components, for storage of energy, and in signal transduction. Hence, efficient regulation and maintenance of lipid homeostasis are crucial for normal cellular and tissue function. In the past decade, increasing research has shown the importance of ubiquitination in regulating the stability of key players in different aspects of lipid metabolism. This review describes recent insights into the regulation of lipid metabolism by ubiquitin signaling, discusses how ubiquitination can be targeted in diseases characterized by lipid dysregulation, and identifies areas that require further research.
Collapse
Affiliation(s)
- Melanie Loix
- Biomedical Research Institute, School of Life Sciences, Hasselt University, Diepenbeek, Belgium; University MS Center Hasselt, Pelt, Belgium
| | - Noam Zelcer
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences and Gastroenterology and Metabolism, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeroen F J Bogie
- Biomedical Research Institute, School of Life Sciences, Hasselt University, Diepenbeek, Belgium; University MS Center Hasselt, Pelt, Belgium
| | - Jerome J A Hendriks
- Biomedical Research Institute, School of Life Sciences, Hasselt University, Diepenbeek, Belgium; University MS Center Hasselt, Pelt, Belgium.
| |
Collapse
|
7
|
Wang H, Shu L, Lv C, Liu N, Long Y, Peng X, Ling H, Tao T, Tang J, Cheng Y, Liu S, Xiao D, Tao Y. BRCC36 Deubiquitinates HMGCR to Regulate the Interplay Between Ferroptosis and Pyroptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304263. [PMID: 38178583 PMCID: PMC10953584 DOI: 10.1002/advs.202304263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 12/01/2023] [Indexed: 01/06/2024]
Abstract
Various forms of programmed cell death (PCD) exhibit distinct characteristics depending on their specific molecular mechanisms, and there are interactions among these different forms. Ferroptosis, which is related to autophagy and apoptosis, has an unknown potential interaction with pyroptosis. This study revealed a mutually antagonistic relationship between ferroptosis and pyroptosis, with 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) playing a key role in their interaction. It is found that HMGCR predominantly localized to mitochondria during ferroptosis but shifted to the endoplasmic reticulum following treatment with a pyroptosis inducer. Furthermore, this study demonstrated that BRCC36 (BRCA1/BRCA2-containing complex subunit 36) deubiquitinated HMGCR in a manner dependent on deubiquitinating enzyme (DUB) activity, and inhibited ferroptosis and promoted pyroptosis. Moreover, as an oncogene in hepatocellular carcinoma (HCC), BRCC36 promoted cancer cell proliferation, migration, invasion, and tumor growth. Thiolutin, an inhibitor of BRCC36, effectively suppressed the interaction between BRCC36 and HMGCR, leading to the inhibition of HCC growth. Therefore, targeting BRCC36 can offer a novel and promising therapeutic strategy for HCC treatment. In conclusion, these findings provide new theoretical evidence for further characterizing tumor heterogeneity and offer new molecular targets for the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Haiyan Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion (Central South UniversityMinistry of Education)Department of PathologyXiangya HospitalCentral South UniversityHunan410078China
- Academy of Biomedical EngineeringKunming Medical UniversityKunming650500China
- NHC Key Laboratory of CarcinogenesisCancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunan410078China
| | - Long Shu
- Key Laboratory of Carcinogenesis and Cancer Invasion (Central South UniversityMinistry of Education)Department of PathologyXiangya HospitalCentral South UniversityHunan410078China
- NHC Key Laboratory of CarcinogenesisCancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunan410078China
| | - Cairui Lv
- Key Laboratory of Carcinogenesis and Cancer Invasion (Central South UniversityMinistry of Education)Department of PathologyXiangya HospitalCentral South UniversityHunan410078China
- NHC Key Laboratory of CarcinogenesisCancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunan410078China
| | - Na Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion (Central South UniversityMinistry of Education)Department of PathologyXiangya HospitalCentral South UniversityHunan410078China
- NHC Key Laboratory of CarcinogenesisCancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunan410078China
| | - Yao Long
- Key Laboratory of Carcinogenesis and Cancer Invasion (Central South UniversityMinistry of Education)Department of PathologyXiangya HospitalCentral South UniversityHunan410078China
- NHC Key Laboratory of CarcinogenesisCancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunan410078China
| | - Xintong Peng
- Key Laboratory of Carcinogenesis and Cancer Invasion (Central South UniversityMinistry of Education)Department of PathologyXiangya HospitalCentral South UniversityHunan410078China
- NHC Key Laboratory of CarcinogenesisCancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunan410078China
| | - Huli Ling
- Key Laboratory of Carcinogenesis and Cancer Invasion (Central South UniversityMinistry of Education)Department of PathologyXiangya HospitalCentral South UniversityHunan410078China
- NHC Key Laboratory of CarcinogenesisCancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunan410078China
| | - Tania Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion (Central South UniversityMinistry of Education)Department of PathologyXiangya HospitalCentral South UniversityHunan410078China
- Hunan Key Laboratory of Early Diagnosis and Precision TherapyDepartment of Thoracic SurgerySecond Xiangya HospitalCentral South UniversityChangsha410011China
| | - Jun Tang
- Key Laboratory of Carcinogenesis and Cancer Invasion (Central South UniversityMinistry of Education)Department of PathologyXiangya HospitalCentral South UniversityHunan410078China
- NHC Key Laboratory of CarcinogenesisCancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunan410078China
| | - Yan Cheng
- Department of PharmacyThe Second Xiangya HospitalCentral South UniversityChangsha410011China
| | - Shuang Liu
- Department of OncologyInstitute of Medical SciencesNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Desheng Xiao
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion (Central South UniversityMinistry of Education)Department of PathologyXiangya HospitalCentral South UniversityHunan410078China
- NHC Key Laboratory of CarcinogenesisCancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunan410078China
- Hunan Key Laboratory of Early Diagnosis and Precision TherapyDepartment of Thoracic SurgerySecond Xiangya HospitalCentral South UniversityChangsha410011China
- Hunan Key Laboratory of Cancer MetabolismHunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaHunan410031China
| |
Collapse
|
8
|
Zhou X, Wu X, Wang R, Han L, Li H, Zhao W. Mechanisms of 3-Hydroxyl 3-Methylglutaryl CoA Reductase in Alzheimer's Disease. Int J Mol Sci 2023; 25:170. [PMID: 38203341 PMCID: PMC10778631 DOI: 10.3390/ijms25010170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide and has a high incidence in the elderly. Unfortunately, there is no effective therapy for AD owing to its complicated pathogenesis. However, the development of lipid-lowering anti-inflammatory drugs has heralded a new era in the treatment of Alzheimer's disease. Several studies in recent years have shown that lipid metabolic dysregulation and neuroinflammation are associated with the pathogenesis of AD. 3-Hydroxyl 3-methylglutaryl CoA reductase (HMGCR) is a rate-limiting enzyme in cholesterol synthesis that plays a key role in cholesterol metabolism. HMGCR inhibitors, known as statins, have changed from being solely lipid-lowering agents to neuroprotective compounds because of their effects on lipid levels and inflammation. In this review, we first summarize the main regulatory mechanism of HMGCR affecting cholesterol biosynthesis. We also discuss the pathogenesis of AD induced by HMGCR, including disordered lipid metabolism, oxidative stress, inflammation, microglial proliferation, and amyloid-β (Aβ) deposition. Subsequently, we explain the possibility of HMGCR as a potential target for AD treatment. Statins-based AD treatment is an ascent field and currently quite controversial; therefore, we also elaborate on the current application prospects and limitations of statins in AD treatment.
Collapse
Affiliation(s)
- Xun Zhou
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
- Department of Endocrinology, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China;
| | - Xiaolang Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
| | - Rui Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
| | - Lu Han
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
| | - Huilin Li
- Department of Endocrinology, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China;
| | - Wei Zhao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
| |
Collapse
|
9
|
Gawden-Bone CM, Lehner PJ, Volkmar N. As a matter of fat: Emerging roles of lipid-sensitive E3 ubiquitin ligases. Bioessays 2023; 45:e2300139. [PMID: 37890275 DOI: 10.1002/bies.202300139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023]
Abstract
The dynamic structure and composition of lipid membranes need to be tightly regulated to control the vast array of cellular processes from cell and organelle morphology to protein-protein interactions and signal transduction pathways. To maintain membrane integrity, sense-and-response systems monitor and adjust membrane lipid composition to the ever-changing cellular environment, but only a relatively small number of control systems have been described. Here, we explore the emerging role of the ubiquitin-proteasome system in monitoring and maintaining membrane lipid composition. We focus on the ER-resident RNF145 E3 ubiquitin ligase, its role in regulating adiponectin receptor 2 (ADIPOR2), its lipid hydrolase substrate, and the broader implications for understanding the homeostatic processes that fine-tune cellular membrane composition.
Collapse
Affiliation(s)
- Christian M Gawden-Bone
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Paul J Lehner
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Norbert Volkmar
- Institute for Molecular Systems Biology (IMSB), ETH Zürich, Zürich, Switzerland
| |
Collapse
|
10
|
Vatandaslar H, Garzia A, Meyer C, Godbersen S, Brandt LTL, Griesbach E, Chao JA, Tuschl T, Stoffel M. In vivo PAR-CLIP (viP-CLIP) of liver TIAL1 unveils targets regulating cholesterol synthesis and secretion. Nat Commun 2023; 14:3386. [PMID: 37296170 PMCID: PMC10256721 DOI: 10.1038/s41467-023-39135-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
System-wide cross-linking and immunoprecipitation (CLIP) approaches have unveiled regulatory mechanisms of RNA-binding proteins (RBPs) mainly in cultured cells due to limitations in the cross-linking efficiency of tissues. Here, we describe viP-CLIP (in vivo PAR-CLIP), a method capable of identifying RBP targets in mammalian tissues, thereby facilitating the functional analysis of RBP-regulatory networks in vivo. We applied viP-CLIP to mouse livers and identified Insig2 and ApoB as prominent TIAL1 target transcripts, indicating an important role of TIAL1 in cholesterol synthesis and secretion. The functional relevance of these targets was confirmed by showing that TIAL1 influences their translation in hepatocytes. Mutant Tial1 mice exhibit altered cholesterol synthesis, APOB secretion and plasma cholesterol levels. Our results demonstrate that viP-CLIP can identify physiologically relevant RBP targets by finding a factor implicated in the negative feedback regulation of cholesterol biosynthesis.
Collapse
Affiliation(s)
- Hasan Vatandaslar
- Institute of Molecular Health Sciences, ETH Zurich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Aitor Garzia
- Laboratory of RNA Molecular Biology, The Rockefeller University, 1230 York Avenue, New York, NY, 10021, USA
| | - Cindy Meyer
- Laboratory of RNA Molecular Biology, The Rockefeller University, 1230 York Avenue, New York, NY, 10021, USA
| | - Svenja Godbersen
- Institute of Molecular Health Sciences, ETH Zurich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Laura T L Brandt
- Institute of Molecular Health Sciences, ETH Zurich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland
| | - Esther Griesbach
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058, Basel, Switzerland
| | - Jeffrey A Chao
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058, Basel, Switzerland
| | - Thomas Tuschl
- Laboratory of RNA Molecular Biology, The Rockefeller University, 1230 York Avenue, New York, NY, 10021, USA
| | - Markus Stoffel
- Institute of Molecular Health Sciences, ETH Zurich, Otto-Stern-Weg 7, 8093, Zürich, Switzerland.
- Medical Faculty, University of Zürich, 8091, Zürich, Switzerland.
| |
Collapse
|
11
|
Chen Y, Xu Y, Zhao H, Zhou Y, Zhang J, Lei J, Wu L, Zhou M, Wang J, Yang S, Zhang X, Yan G, Li Y. Myeloid-derived suppressor cells deficient in cholesterol biosynthesis promote tumor immune evasion. Cancer Lett 2023; 564:216208. [PMID: 37150500 DOI: 10.1016/j.canlet.2023.216208] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/09/2023]
Abstract
Cancer immunotherapy targeting myeloid-derived suppressor cells (MDSCs) is one of the most promising anticancer strategies. Metabolic reprogramming is vital for MDSC activation, however, the regulatory mechanisms of cholesterol metabolic reprogramming in MDSCs remains largely unexplored. Using the receptor-interacting protein kinase 3 (RIPK3)-deficient MDSC model, a previously established tumor-infiltrating MDSC-like model, we found that the cholesterol accumulation was significantly decreased in these cells. Moreover, the phosphorylated AKT-mTORC1 signaling was reduced, and downstream SREBP2-HMGCR-mediated cholesterol synthesis was blunted. Interestingly, cholesterol deficiency profoundly elevated the immunosuppressive activity of MDSCs. Mechanistically, cholesterol elimination induced nuclear accumulation of LXRβ, thereby promoting LXRβ-RXRα heterodimer binding of a novel composite element in the promoter of Arg1. Furthermore, itraconazole enhanced the immunosuppressive activity of MDSCs to boost tumor growth by suppressing the RIPK3-AKT-mTORC1 pathway and impeding cholesterol synthesis. Our findings demonstrate that RIPK3 deficiency leads to cholesterol abrogation in MDSCs, which facilitates tumor-infiltrating MDSC activation, and highlight the therapeutic potential of targeting cholesterol synthesis to overcome tumor immune evasion.
Collapse
Affiliation(s)
- Yu Chen
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Yanquan Xu
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Huakan Zhao
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Yu Zhou
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Jiangang Zhang
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Juan Lei
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Lei Wu
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Mingyue Zhou
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Jingchun Wang
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Shuai Yang
- Department of Pathology, The 958th Hospital, Southwest Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xiao Zhang
- Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Army Medical University, Shigatse, Tibet Autonomous Region, 857000, China
| | - Guifang Yan
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| | - Yongsheng Li
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China; Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
12
|
Fenton NM, Nguyen TB, Sharpe LJ, Brown AJ. Refining sugar's involvement in cholesterol synthesis. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159266. [PMID: 36528253 DOI: 10.1016/j.bbalip.2022.159266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/03/2022] [Accepted: 12/06/2022] [Indexed: 12/15/2022]
Abstract
Glucose metabolism and cholesterol synthesis are often regarded in isolation. Increasing evidence not only links these pathways but also suggests glucose catabolism regulates cholesterol synthesis. Uptake of glucose increases cholesterol production. However, the precise mechanism by which this occurs is not fully understood and is likely to involve many aspects of cellular pathways participating in energy sensing, cholesterol regulation, and synthesis. Here, we review some interesting links between cholesterol synthesis and glucose metabolism. Given glucose breakdown produces energy (both via glycolysis and its products through oxidative phosphorylation), and considering cholesterol synthesis is an energetically demanding process, it would seem logical that glucose metabolism impacts cholesterol synthesis. The energy sensing kinase AMPK carefully monitors energy supply to induce or suppress cholesterol synthesis as needed. Akt, activated by the insulin signalling cascade, regulates key transcription factors involved in lipid metabolism. The insulin signalling pathway also activates machinery involved in the deubiquitination of a key cholesterol synthesis enzyme. Moreover, glucose metabolites, acetyl-CoA, and GlcNAc are substrates for protein acetylation and N-glycosylation, respectively, and can stabilise proteins involved in cholesterol synthesis. As glucose and cholesterol dysregulation are both associated with numerous diseases, understanding the mechanisms of how glucose metabolism and cholesterol synthesis intersect may offer new avenues for therapeutics that make use of these findings.
Collapse
Affiliation(s)
- Nicole M Fenton
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Tina B Nguyen
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Laura J Sharpe
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Andrew J Brown
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
13
|
Wang Z, Wang M, Zhang M, Xu K, Zhang X, Xie Y, Zhang Y, Chang C, Li X, Sun A, He F. High-affinity SOAT1 ligands remodeled cholesterol metabolism program to inhibit tumor growth. BMC Med 2022; 20:292. [PMID: 35941608 PMCID: PMC9361549 DOI: 10.1186/s12916-022-02436-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 06/13/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Although cholesterol metabolism is a common pathway for the development of antitumor drugs, there are no specific targets and drugs for clinical use. Here, based on our previous study of sterol O-acyltransferase 1 (SOAT1) in hepatocelluar carcinoma, we sought to screen an effective targeted drug for precise treatment of hepatocelluar carcinoma and, from the perspective of cholesterol metabolism, clarify the relationship between cholesterol regulation and tumorigenesis and development. METHODS In this study, we developed a virtual screening integrated affinity screening technology for target protein drug screening. A series of in vitro and in vivo experiments were used for drug activity verification. Multi-omics analysis and flow cytometry analysis were used to explore antitumor mechanisms. Comparative analysis of proteome and transcriptome combined with survival follow-up information of patients reveals the clinical therapeutic potential of screened drugs. RESULTS We screened three compounds, nilotinib, ABT-737, and evacetrapib, that exhibited optimal binding with SOAT1. In particular, nilotinib displayed a high affinity for SOAT1 protein and significantly inhibited tumor activity both in vitro and in vivo. Multi-omics analysis and flow cytometry analysis indicated that SOAT1-targeting compounds reprogrammed the cholesterol metabolism in tumors and enhanced CD8+ T cells and neutrophils to suppress tumor growth. CONCLUSIONS Taken together, we reported several high-affinity SOAT1 ligands and demonstrated their clinical potential in the precision therapy of liver cancer, and also reveal the potential antitumor mechanism of SOAT1-targeting compounds.
Collapse
Affiliation(s)
- Zhihua Wang
- grid.419611.a0000 0004 0457 9072State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, 102206 China
- grid.506261.60000 0001 0706 7839Research Unit of Proteomics Dirven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing, 102206 China
| | - Miaomiao Wang
- grid.452422.70000 0004 0604 7301Shandong First Medical University, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014 China
| | - Mengxin Zhang
- grid.419611.a0000 0004 0457 9072State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, 102206 China
| | - Kaikun Xu
- grid.419611.a0000 0004 0457 9072State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, 102206 China
| | - Xinshuai Zhang
- grid.419611.a0000 0004 0457 9072State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, 102206 China
| | - Yi Xie
- grid.419611.a0000 0004 0457 9072State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, 102206 China
- grid.12527.330000 0001 0662 3178Department of Pharmacology and Pharmaceutical Sciences, School of Medicine, Tsinghua University, Beijing, 100083 China
| | - Yiming Zhang
- grid.419611.a0000 0004 0457 9072State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, 102206 China
| | - Cheng Chang
- grid.419611.a0000 0004 0457 9072State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, 102206 China
| | - Xiaolu Li
- grid.452422.70000 0004 0604 7301Shandong First Medical University, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014 China
| | - Aihua Sun
- grid.419611.a0000 0004 0457 9072State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, 102206 China
- grid.506261.60000 0001 0706 7839Research Unit of Proteomics Dirven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing, 102206 China
| | - Fuchu He
- grid.419611.a0000 0004 0457 9072State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, 102206 China
- grid.506261.60000 0001 0706 7839Research Unit of Proteomics Dirven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing, 102206 China
| |
Collapse
|
14
|
Juhl AD, Wüstner D. Pathways and Mechanisms of Cellular Cholesterol Efflux-Insight From Imaging. Front Cell Dev Biol 2022; 10:834408. [PMID: 35300409 PMCID: PMC8920967 DOI: 10.3389/fcell.2022.834408] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/04/2022] [Indexed: 12/24/2022] Open
Abstract
Cholesterol is an essential molecule in cellular membranes, but too much cholesterol can be toxic. Therefore, mammalian cells have developed complex mechanisms to remove excess cholesterol. In this review article, we discuss what is known about such efflux pathways including a discussion of reverse cholesterol transport and formation of high-density lipoprotein, the function of ABC transporters and other sterol efflux proteins, and we highlight their role in human diseases. Attention is paid to the biophysical principles governing efflux of sterols from cells. We also discuss recent evidence for cholesterol efflux by the release of exosomes, microvesicles, and migrasomes. The role of the endo-lysosomal network, lipophagy, and selected lysosomal transporters, such as Niemann Pick type C proteins in cholesterol export from cells is elucidated. Since oxysterols are important regulators of cellular cholesterol efflux, their formation, trafficking, and secretion are described briefly. In addition to discussing results obtained with traditional biochemical methods, focus is on studies that use established and novel bioimaging approaches to obtain insight into cholesterol efflux pathways, including fluorescence and electron microscopy, atomic force microscopy, X-ray tomography as well as mass spectrometry imaging.
Collapse
Affiliation(s)
| | - Daniel Wüstner
- Department of Biochemistry and Molecular Biology, PhyLife, Physical Life Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
15
|
Goedeke L, Canfrán-Duque A, Rotllan N, Chaube B, Thompson BM, Lee RG, Cline GW, McDonald JG, Shulman GI, Lasunción MA, Suárez Y, Fernández-Hernando C. MMAB promotes negative feedback control of cholesterol homeostasis. Nat Commun 2021; 12:6448. [PMID: 34750386 PMCID: PMC8575900 DOI: 10.1038/s41467-021-26787-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/14/2021] [Indexed: 12/21/2022] Open
Abstract
Intricate regulatory networks govern the net balance of cholesterol biosynthesis, uptake and efflux; however, the mechanisms surrounding cholesterol homeostasis remain incompletely understood. Here, we develop an integrative genomic strategy to detect regulators of LDLR activity and identify 250 genes whose knockdown affects LDL-cholesterol uptake and whose expression is modulated by intracellular cholesterol levels in human hepatic cells. From these hits, we focus on MMAB, an enzyme which catalyzes the conversion of vitamin B12 to adenosylcobalamin, and whose expression has previously been linked with altered levels of circulating cholesterol in humans. We demonstrate that hepatic levels of MMAB are modulated by dietary and cellular cholesterol levels through SREBP2, the master transcriptional regulator of cholesterol homeostasis. Knockdown of MMAB decreases intracellular cholesterol levels and augments SREBP2-mediated gene expression and LDL-cholesterol uptake in human and mouse hepatic cell lines. Reductions in total sterol content were attributed to increased intracellular levels of propionic and methylmalonic acid and subsequent inhibition of HMGCR activity and cholesterol biosynthesis. Moreover, mice treated with antisense inhibitors of MMAB display a significant reduction in hepatic HMGCR activity, hepatic sterol content and increased expression of SREBP2-mediated genes. Collectively, these findings reveal an unexpected role for the adenosylcobalamin pathway in regulating LDLR expression and identify MMAB as an additional control point by which cholesterol biosynthesis is regulated by its end product.
Collapse
Affiliation(s)
- Leigh Goedeke
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Alberto Canfrán-Duque
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Noemi Rotllan
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Balkrishna Chaube
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Bonne M Thompson
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Richard G Lee
- Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Carlsbad, CA, 92010, USA
| | - Gary W Cline
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Jeffrey G McDonald
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Miguel A Lasunción
- Servicio de Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRyCIS) and CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Yajaira Suárez
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Pathology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Pathology, Yale School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
16
|
Ho WY, Chang JC, Lim K, Cazenave-Gassiot A, Nguyen AT, Foo JC, Muralidharan S, Viera-Ortiz A, Ong SJM, Hor JH, Agrawal I, Hoon S, Arogundade OA, Rodriguez MJ, Lim SM, Kim SH, Ravits J, Ng SY, Wenk MR, Lee EB, Tucker-Kellogg G, Ling SC. TDP-43 mediates SREBF2-regulated gene expression required for oligodendrocyte myelination. J Cell Biol 2021; 220:212536. [PMID: 34347016 PMCID: PMC8348376 DOI: 10.1083/jcb.201910213] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/16/2020] [Accepted: 05/28/2021] [Indexed: 12/12/2022] Open
Abstract
Cholesterol metabolism operates autonomously within the central nervous system (CNS), where the majority of cholesterol resides in myelin. We demonstrate that TDP-43, the pathological signature protein for amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), influences cholesterol metabolism in oligodendrocytes. TDP-43 binds directly to mRNA of SREBF2, the master transcription regulator for cholesterol metabolism, and multiple mRNAs encoding proteins responsible for cholesterol biosynthesis and uptake, including HMGCR, HMGCS1, and LDLR. TDP-43 depletion leads to reduced SREBF2 and LDLR expression, and cholesterol levels in vitro and in vivo. TDP-43-mediated changes in cholesterol levels can be restored by reintroducing SREBF2 or LDLR. Additionally, cholesterol supplementation rescues demyelination caused by TDP-43 deletion. Furthermore, oligodendrocytes harboring TDP-43 pathology from FTD patients show reduced HMGCR and HMGCS1, and coaggregation of LDLR and TDP-43. Collectively, our results indicate that TDP-43 plays a role in cholesterol homeostasis in oligodendrocytes, and cholesterol dysmetabolism may be implicated in TDP-43 proteinopathies-related diseases.
Collapse
Affiliation(s)
- Wan Yun Ho
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jer-Cherng Chang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kenneth Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Computational Biology Programme, Faculty of Science, National University of Singapore, Singapore
| | - Amaury Cazenave-Gassiot
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore
| | - Aivi T Nguyen
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | - Juat Chin Foo
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore
| | - Sneha Muralidharan
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore
| | - Ashley Viera-Ortiz
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sarah J M Ong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jin Hui Hor
- Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore
| | - Ira Agrawal
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shawn Hoon
- Molecular Engineering Laboratory, A*STAR Research Entities, Singapore
| | | | - Maria J Rodriguez
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Su Min Lim
- Department of Neurology, and Biomedical Research Institute, Hanyang University College of Medicine, Seoul, South Korea.,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Seung Hyun Kim
- Department of Neurology, and Biomedical Research Institute, Hanyang University College of Medicine, Seoul, South Korea
| | - John Ravits
- Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Shi-Yan Ng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Institute of Molecular and Cell Biology, A*STAR Research Entities, Singapore
| | - Markus R Wenk
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore
| | - Edward B Lee
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA
| | - Greg Tucker-Kellogg
- Computational Biology Programme, Faculty of Science, National University of Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore
| | - Shuo-Chien Ling
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Program in Neuroscience and Behavior Disorders, Duke-National University of Singapore Medical School, Singapore
| |
Collapse
|
17
|
Dong Y, Li X, Liu Y, Gao J, Tao J. The molecular targets of taurine confer anti-hyperlipidemic effects. Life Sci 2021; 278:119579. [PMID: 33961852 DOI: 10.1016/j.lfs.2021.119579] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/16/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022]
Abstract
Hyperlipidemia, an independent risk factor for atherosclerosis, is regarded as a lipid metabolism disorder associated with elevated plasma triglyceride and/or cholesterol. Genetic factors and unhealthy lifestyles, such as excess caloric intake and physical inactivity, can result in hyperlipidemia. Taurine, a sulfur-containing non-essential amino acid, is abundant in marine foods and has been associated with wide-ranging beneficial physiological effects, with special reference to regulating aberrant lipid metabolism. Its anti-hyperlipidemic mechanism is complex, which is related to many enzymes in the process of fat anabolism and catabolism (e.g., HMGCR, CYP7A1, LDLR, FXR, FAS and ACC). Anti-inflammatory and antioxidant molecular targets, lipid autophagy, metabolic reprogramming and gut microbiota will also be reviewed.
Collapse
Affiliation(s)
- Yuanyuan Dong
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, China; Division of Life Sciences and Medicine, University of Science and Technology of China, No. 17 LuJiang Road, Hefei 230001, Anhui, China
| | - Xiaoling Li
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, China; Division of Life Sciences and Medicine, University of Science and Technology of China, No. 17 LuJiang Road, Hefei 230001, Anhui, China
| | - Yaling Liu
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, China; Division of Life Sciences and Medicine, University of Science and Technology of China, No. 17 LuJiang Road, Hefei 230001, Anhui, China
| | - Jie Gao
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, China; Division of Life Sciences and Medicine, University of Science and Technology of China, No. 17 LuJiang Road, Hefei 230001, Anhui, China
| | - Jinhui Tao
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, China; Division of Life Sciences and Medicine, University of Science and Technology of China, No. 17 LuJiang Road, Hefei 230001, Anhui, China.
| |
Collapse
|
18
|
Ming J, Wei X, Han M, Adi D, Abuzhalihan J, Wang YT, Yang YN, Li XM, Xie X, Fu ZY, Gai MT, Ma YT. Genetic variation of RNF145 gene and blood lipid levels in Xinjiang population, China. Sci Rep 2021; 11:5969. [PMID: 33727652 PMCID: PMC7966793 DOI: 10.1038/s41598-021-85503-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/24/2021] [Indexed: 01/24/2023] Open
Abstract
Dyslipidemia is one of the main risk factors for coronary heart disease (CHD). The E3 ubiquitin ligase which is encoded by the ring finger protein 145 (RNF145) gene is very important in the mediation of cholesterol synthesis and effectively treats hypercholesterolemia. Thus, the purpose of the present research is to investigate the connection between the polymorphism of the RNF145 gene and cholesterol levels in the populations in Xinjiang, China. A total of 1396 participants (Male: 628, Female: 768) were included in this study for genetic analysis of RNF145 gene, and we used the modified multiple connection detection response (iMLDR) technology to label two SNPs (rs17056583, rs12188266) of RNF145 genotyping. The relationship between the genotypes and the lipid profiles was analyzed with general linear model analysis after adjusting confounding variables. Through the analysis of the two SNPs in RNF145 gene, we discovered that both rs17056583 and rs12188266 were related to total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) concentrations (All P < 0.001). In addition, the association of rs17056583 and rs12188266 with lipid profiles concentrations is still statistically significant after multivariate adjustment of sex, age, smoking, obesity, drinking, diabetes, hypertension and lipid profiles. Meanwhile, we also found that rs17056583 was associated with high triglycerides concentrations before and after adjustment (All P < 0.001). Our study shows that both rs17056583 and rs12188266 SNPs of RNP145 gene are related to TC and LDL-C concentrations in Xinjiang population.
Collapse
Affiliation(s)
- Jing Ming
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Xian Wei
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Min Han
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Dilare Adi
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Jialin Abuzhalihan
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Yong-Tao Wang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Yi-Ning Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Xiao-Mei Li
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Xiang Xie
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Zhen-Yan Fu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China.,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Min-Tao Gai
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China
| | - Yi-Tong Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People's Republic of China. .,Xinjiang Key Laboratory of Cardiovascular Disease Research, Urumqi, 830054, People's Republic of China.
| |
Collapse
|
19
|
The Degron Architecture of Squalene Monooxygenase and How Specific Lipids Calibrate Levels of This Key Cholesterol Synthesis Enzyme. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 32979157 DOI: 10.1007/5584_2020_583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register]
Abstract
Cholesterol synthesis is a fundamental process that contributes to cellular cholesterol homeostasis. Cells execute transcriptional and post-translational mechanisms to control the abundance of enzymes of the cholesterol synthesis pathway, consequently affecting cholesterol production. One such highly tuned enzyme is squalene monooxygenase (SM), which catalyzes a rate-limiting step in the pathway. A well-characterized mechanism is the cholesterol-mediated degradation of SM. Notably, lipids (cholesterol, plasmalogens, squalene, and unsaturated fatty acids) can act as cellular signals that either promote or reduce SM degradation. The N-terminal region of SM consists of the shortest known cholesterol-responsive degron, characterized by atypical membrane anchoring structures, namely a re-entrant loop and an amphipathic helix. SM also undergoes non-canonical ubiquitination on serine, a relatively uncommon attachment site for ubiquitination. The structure of the catalytic domain of SM has been solved, providing insights into the catalytic mechanisms and modes of inhibition by well-known SM inhibitors, some of which have been effective in lowering cholesterol levels in animal models. Certain human cancers have been linked to dysregulation of SM levels and activity, further emphasizing the relevance of SM in health and disease.
Collapse
|
20
|
Wangeline MA, Hampton RY. An autonomous, but INSIG-modulated, role for the sterol sensing domain in mallostery-regulated ERAD of yeast HMG-CoA reductase. J Biol Chem 2020; 296:100063. [PMID: 33184059 PMCID: PMC7948459 DOI: 10.1074/jbc.ra120.015910] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/01/2020] [Accepted: 11/12/2020] [Indexed: 01/23/2023] Open
Abstract
HMG-CoA reductase (HMGR) undergoes feedback-regulated degradation as part of sterol pathway control. Degradation of the yeast HMGR isozyme Hmg2 is controlled by the sterol pathway intermediate GGPP, which causes misfolding of Hmg2, leading to degradation by the HRD pathway; we call this process mallostery. We evaluated the role of the Hmg2 sterol sensing domain (SSD) in mallostery, as well as the involvement of the highly conserved INSIG proteins. We show that the Hmg2 SSD is critical for regulated degradation of Hmg2 and required for mallosteric misfolding of GGPP as studied by in vitro limited proteolysis. The Hmg2 SSD functions independently of conserved yeast INSIG proteins, but its function was modulated by INSIG, thus imposing a second layer of control on Hmg2 regulation. Mutant analyses indicated that SSD-mediated mallostery occurred prior to and independent of HRD-dependent ubiquitination. GGPP-dependent misfolding was still extant but occurred at a much slower rate in the absence of a functional SSD, indicating that the SSD facilitates a physiologically useful rate of GGPP response and implying that the SSD is not a binding site for GGPP. Nonfunctional SSD mutants allowed us to test the importance of Hmg2 quaternary structure in mallostery: a nonresponsive Hmg2 SSD mutant strongly suppressed regulation of a coexpressed, normal Hmg2. Finally, we have found that GGPP-regulated misfolding occurred in detergent-solubilized Hmg2, a feature that will allow next-level analysis of the mechanism of this novel tactic of ligand-regulated misfolding.
Collapse
Affiliation(s)
- Margaret A Wangeline
- Division of Biological Sciences, the Section of Cell and Developmental Biology, UCSD, La Jolla, California, USA
| | - Randolph Y Hampton
- Division of Biological Sciences, the Section of Cell and Developmental Biology, UCSD, La Jolla, California, USA.
| |
Collapse
|
21
|
Oommen D, Kizhakkedath P, Jawabri AA, Varghese DS, Ali BR. Proteostasis Regulation in the Endoplasmic Reticulum: An Emerging Theme in the Molecular Pathology and Therapeutic Management of Familial Hypercholesterolemia. Front Genet 2020; 11:570355. [PMID: 33173538 PMCID: PMC7538668 DOI: 10.3389/fgene.2020.570355] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 08/31/2020] [Indexed: 02/05/2023] Open
Abstract
Familial hypercholesterolemia (FH) is an autosomal genetic disease characterized by high serum low-density lipoprotein (LDL) content leading to premature coronary artery disease. The main genetic and molecular causes of FH are mutations in low-density lipoprotein receptor gene (LDLR) resulting in the non-clearance of LDL from the blood by hepatocytes and consequently the formation of plaques. LDLR is synthesized and glycosylated in the endoplasmic reticulum (ER) and then transported to the plasma membrane via Golgi. It is estimated that more than 50% of reported FH-causing mutations in LDLR result in misfolded proteins that are transport-defective and hence retained in ER. ER accumulation of misfolded proteins causes ER-stress and activates unfolded protein response (UPR). UPR aids protein folding, blocks further protein synthesis, and eliminates misfolded proteins via ER-associated degradation (ERAD) to alleviate ER stress. Various studies demonstrated that ER-retained LDLR mutants are subjected to ERAD. Interestingly, chemical chaperones and genetic or pharmacological inhibition of ERAD have been reported to rescue the transport defective mutant LDLR alleles from ERAD and restore their ER-Golgi transport resulting in the expression of functional plasma membrane LDLR. This suggests the possibility of pharmacological modulation of proteostasis in the ER as a therapeutic strategy for FH. In this review, we picture a detailed analysis of UPR and the ERAD processes activated by ER-retained LDLR mutants associated with FH. In addition, we discuss and critically evaluate the potential role of chemical chaperones and ERAD modulators in the therapeutic management of FH.
Collapse
Affiliation(s)
- Deepu Oommen
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Praseetha Kizhakkedath
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Aseel A. Jawabri
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Divya Saro Varghese
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Bassam R. Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
- Zayed Center for Health Sciences, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| |
Collapse
|
22
|
The MARCH6-SQLE Axis Controls Endothelial Cholesterol Homeostasis and Angiogenic Sprouting. Cell Rep 2020; 32:107944. [DOI: 10.1016/j.celrep.2020.107944] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/23/2020] [Accepted: 07/01/2020] [Indexed: 12/17/2022] Open
|
23
|
Aguilar-Ballester M, Herrero-Cervera A, Vinué Á, Martínez-Hervás S, González-Navarro H. Impact of Cholesterol Metabolism in Immune Cell Function and Atherosclerosis. Nutrients 2020; 12:nu12072021. [PMID: 32645995 PMCID: PMC7400846 DOI: 10.3390/nu12072021] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 06/29/2020] [Accepted: 07/02/2020] [Indexed: 12/24/2022] Open
Abstract
Cholesterol, the most important sterol in mammals, helps maintain plasma membrane fluidity and is a precursor of bile acids, oxysterols, and steroid hormones. Cholesterol in the body is obtained from the diet or can be de novo synthetized. Cholesterol homeostasis is mainly regulated by the liver, where cholesterol is packed in lipoproteins for transport through a tightly regulated process. Changes in circulating lipoprotein cholesterol levels lead to atherosclerosis development, which is initiated by an accumulation of modified lipoproteins in the subendothelial space; this induces significant changes in immune cell differentiation and function. Beyond lesions, cholesterol levels also play important roles in immune cells such as monocyte priming, neutrophil activation, hematopoietic stem cell mobilization, and enhanced T cell production. In addition, changes in cholesterol intracellular metabolic enzymes or transporters in immune cells affect their signaling and phenotype differentiation, which can impact on atherosclerosis development. In this review, we describe the main regulatory pathways and mechanisms of cholesterol metabolism and how these affect immune cell generation, proliferation, activation, and signaling in the context of atherosclerosis.
Collapse
Affiliation(s)
- María Aguilar-Ballester
- INCLIVA Institute of Health Research, 46010 Valencia, Spain; (M.A.-B.); (A.H.-C.); (Á.V.); (S.M.-H.)
| | - Andrea Herrero-Cervera
- INCLIVA Institute of Health Research, 46010 Valencia, Spain; (M.A.-B.); (A.H.-C.); (Á.V.); (S.M.-H.)
| | - Ángela Vinué
- INCLIVA Institute of Health Research, 46010 Valencia, Spain; (M.A.-B.); (A.H.-C.); (Á.V.); (S.M.-H.)
| | - Sergio Martínez-Hervás
- INCLIVA Institute of Health Research, 46010 Valencia, Spain; (M.A.-B.); (A.H.-C.); (Á.V.); (S.M.-H.)
- Endocrinology and Nutrition Department Clinic Hospital and Department of Medicine, University of Valencia, 46010 Valencia, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Herminia González-Navarro
- INCLIVA Institute of Health Research, 46010 Valencia, Spain; (M.A.-B.); (A.H.-C.); (Á.V.); (S.M.-H.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Department of Didactics of Experimental and Social Sciences, University of Valencia, 46010 Valencia, Spain
- Correspondence: ; Tel.: +34-963864403; Fax: +34-963987860
| |
Collapse
|
24
|
Tong X, Zhang D, Shabandri O, Oh J, Jin E, Stamper K, Yang M, Zhao Z, Yin L. DDB1 E3 ligase controls dietary fructose-induced ChREBPα stabilization and liver steatosis via CRY1. Metabolism 2020; 107:154222. [PMID: 32246987 PMCID: PMC7282961 DOI: 10.1016/j.metabol.2020.154222] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/25/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022]
Abstract
Fructose over-consumption contributes to the development of liver steatosis in part by stimulating ChREBPα-driven de novo lipogenesis. However, the mechanisms by which fructose activates ChREBP pathway remain largely undefined. Here we performed affinity purification of ChREBPα followed by mass spectrometry and identified DDB1 as a novel interaction protein of ChREBPα in the presence of fructose. Depletion and overexpression of Ddb1 showed opposite effects on the ChREBPα stability in hepatocytes. We next tested the impact of hepatic Ddb1 deficiency on the fructose-induced ChREBP pathway. After 3-week high-fructose diet feeding, both Ddb1 liver-specific knockout and AAV-TBG-Cre-injected Ddb1flox/flox mice showed significantly reduced ChREBPα, lipogenic enzymes, as well as triglycerides in the liver. Mechanistically, DDB1 stabilizes ChREBPα through CRY1, a known ubiquitination target of DDB1 E3 ligase. Finally, overexpression of a degradation-resistant CRY1 mutant (CRY1-585KA) reduces ChREBPα and its target genes in the mouse liver following high-fructose diet feeding. Our data revealed DDB1 as an intracellular sensor of fructose intake to promote hepatic de novo lipogenesis and liver steatosis by stabilizing ChREBPα in a CRY1-dependent manner.
Collapse
Affiliation(s)
- Xin Tong
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, 1137 Catherine Street, Med Sci II 7712, Ann Arbor, MI 48109, USA
| | - Deqiang Zhang
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, 1137 Catherine Street, Med Sci II 7712, Ann Arbor, MI 48109, USA
| | - Omar Shabandri
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, 1137 Catherine Street, Med Sci II 7712, Ann Arbor, MI 48109, USA
| | - Joon Oh
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, 1137 Catherine Street, Med Sci II 7712, Ann Arbor, MI 48109, USA
| | - Ethan Jin
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, 1137 Catherine Street, Med Sci II 7712, Ann Arbor, MI 48109, USA
| | - Kenneth Stamper
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, 1137 Catherine Street, Med Sci II 7712, Ann Arbor, MI 48109, USA
| | - Meichan Yang
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, 1137 Catherine Street, Med Sci II 7712, Ann Arbor, MI 48109, USA; Department of Infectious Diseases, The Second Xianya Hospital, Central South University, Changsha City 410083, Hunan Province, PR China
| | - Zifeng Zhao
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, 1137 Catherine Street, Med Sci II 7712, Ann Arbor, MI 48109, USA; Department of Pharmacology of Chinese Materia, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing City 211198, PR China
| | - Lei Yin
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, 1137 Catherine Street, Med Sci II 7712, Ann Arbor, MI 48109, USA.
| |
Collapse
|
25
|
Talamillo A, Ajuria L, Grillo M, Barroso-Gomila O, Mayor U, Barrio R. SUMOylation in the control of cholesterol homeostasis. Open Biol 2020; 10:200054. [PMID: 32370667 PMCID: PMC7276529 DOI: 10.1098/rsob.200054] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
SUMOylation—protein modification by the small ubiquitin-related modifier (SUMO)—affects several cellular processes by modulating the activity, stability, interactions or subcellular localization of a variety of substrates. SUMO modification is involved in most cellular processes required for the maintenance of metabolic homeostasis. Cholesterol is one of the main lipids required to preserve the correct cellular function, contributing to the composition of the plasma membrane and participating in transmembrane receptor signalling. Besides these functions, cholesterol is required for the synthesis of steroid hormones, bile acids, oxysterols and vitamin D. Cholesterol levels need to be tightly regulated: in excess, it is toxic to the cell, and the disruption of its homeostasis is associated with various disorders like atherosclerosis and cardiovascular diseases. This review focuses on the role of SUMO in the regulation of proteins involved in the metabolism of cholesterol.
Collapse
Affiliation(s)
- Ana Talamillo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain
| | - Leiore Ajuria
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain
| | - Marco Grillo
- Institut de Génomique Fonctionnelle de Lyon (IGFL), École Normale Supérieure de Lyon, Lyon, France.,Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Orhi Barroso-Gomila
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain
| | - Ugo Mayor
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Bizkaia, Spain
| | - Rosa Barrio
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain
| |
Collapse
|
26
|
Li MX, Yang Y, Zhao Q, Wu Y, Song L, Yang H, He M, Gao H, Song BL, Luo J, Rao Y. Degradation versus Inhibition: Development of Proteolysis-Targeting Chimeras for Overcoming Statin-Induced Compensatory Upregulation of 3-Hydroxy-3-methylglutaryl Coenzyme A Reductase. J Med Chem 2020; 63:4908-4928. [DOI: 10.1021/acs.jmedchem.0c00339] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Mei-Xin Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, P.R. China
| | - Yiqing Yang
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, P.R. China
| | - Qiuye Zhao
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, P.R. China
| | - Yue Wu
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, P.R. China
| | - Lei Song
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences PHOENIX Center, Beijing Institute of LifeOmics, Beijing 102206, P.R. China
| | - Haiyan Yang
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Ming He
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, P.R. China
| | - Hongying Gao
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, P.R. China
| | - Bao-Liang Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, P.R. China
| | - Jie Luo
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, P.R. China
| | - Yu Rao
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, P.R. China
| |
Collapse
|
27
|
Ikpa PT, Meijsen KF, Nieuwenhuijze ND, Dulla K, de Jonge HR, Bijvelds MJ. Transcriptome analysis of the distal small intestine of Cftr null mice. Genomics 2020; 112:1139-1150. [DOI: 10.1016/j.ygeno.2019.06.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/18/2019] [Accepted: 06/24/2019] [Indexed: 12/22/2022]
|
28
|
Capell-Hattam IM, Sharpe LJ, Qian L, Hart-Smith G, Prabhu AV, Brown AJ. Twin enzymes, divergent control: The cholesterogenic enzymes DHCR14 and LBR are differentially regulated transcriptionally and post-translationally. J Biol Chem 2020; 295:2850-2865. [PMID: 31911440 PMCID: PMC7049974 DOI: 10.1074/jbc.ra119.011323] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/13/2019] [Indexed: 01/07/2023] Open
Abstract
Cholesterol synthesis is a tightly regulated process, both transcriptionally and post-translationally. Transcriptional control of cholesterol synthesis is relatively well-understood. However, of the ∼20 enzymes in cholesterol biosynthesis, post-translational regulation has only been examined for a small number. Three of the four sterol reductases in cholesterol production, 7-dehydrocholesterol reductase (DHCR7), 14-dehydrocholesterol reductase (DHCR14), and lamin-B receptor (LBR), share evolutionary ties with a high level of sequence homology and predicted structural homology. DHCR14 and LBR uniquely share the same Δ-14 reductase activity in cholesterol biosynthesis, yet little is known about their post-translational regulation. We have previously identified specific modes of post-translational control of DHCR7, but it is unknown whether these regulatory mechanisms are shared by DHCR14 and LBR. Using CHO-7 cells stably expressing epitope-tagged DHCR14 or LBR, we investigated the post-translational regulation of these enzymes. We found that DHCR14 and LBR undergo differential post-translational regulation, with DHCR14 being rapidly turned over, triggered by cholesterol and other sterol intermediates, whereas LBR remained stable. DHCR14 is degraded via the ubiquitin-proteasome system, and we identified several DHCR14 and DHCR7 putative interaction partners, including a number of E3 ligases that modulate DHCR14 levels. Interestingly, we found that gene expression across an array of human tissues showed a negative relationship between the C14-sterol reductases; one enzyme or the other tends to be predominantly expressed in each tissue. Overall, our findings indicate that whereas LBR tends to be the constitutively active C14-sterol reductase, DHCR14 levels are tunable, responding to the local cellular demands for cholesterol.
Collapse
Affiliation(s)
- Isabelle M Capell-Hattam
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - Laura J Sharpe
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - Lydia Qian
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - Gene Hart-Smith
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales 2052, Australia; Department of Molecular Sciences, Macquarie University, Macquarie Park, New South Wales 2109, Australia
| | - Anika V Prabhu
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales 2052, Australia
| | - Andrew J Brown
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, New South Wales 2052, Australia.
| |
Collapse
|
29
|
Petroglou D, Kanellos I, Savopoulos C, Kaiafa G, Chrysochoou A, Skantzis P, Daios S, Hatzitolios AI, Giannoglou G. The LDL-Receptor and its Molecular Properties: From Theory to Novel Biochemical and Pharmacological Approaches in Reducing LDL-cholesterol. Curr Med Chem 2020; 27:317-333. [PMID: 29865996 DOI: 10.2174/0929867325666180604114819] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 02/25/2018] [Accepted: 05/31/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND The Low-Density Lipoprotein (LDL) Receptor (LDL-R) is a transmembrane protein playing a crucial role in effective lipid homeostasis. Various therapeutic agents have been used in the management of dyslipidemias, however, the outcome of therapeutic target is debated. OBJECTIVE The aim of this review is to summarize and fully understand the current concept regarding LDL-R and its molecular properties, metabolic pathway, factors affecting LDL-R activity and all available pharmacological interventions. Additionally, non-lipid related properties of LDL-R are also referred. METHODS Literature from the PubMed database was extracted to identify papers between 1984 to 2017 regarding LDL-R and therapeutic agents on dyslipidemia management. RESULTS We analyzed basic data regarding agents associated with LDL-R (Sterol Regulating Element-Binding Proteins - SREBPs, Protein ARH, IDOL, Thyroid Hormones, Haematologic Disorders, Protein convertase subtilisin kexintype 9 - PCSK-9, ApoC-III) as well as non-lipid related properties of LDL-R, while all relevant (common and novel) pharmacological interventions (statins, fibrates, cholesterol absorption inhibitors, bile acid sequestrants and PCSK- 9) are also referred. CONCLUSION LDL-R and its molecular properties are involved in lipid homeostasis, so potentially sets the therapeutic goals in cardiovascular patients, which is usually debated. Further research is needed in order to fully understand its properties, as well as to find the potential pharmacological interventions that could be beneficial in cholesterol homeostasis and various morbidities in order to reach the most appropriate therapeutic goal.
Collapse
Affiliation(s)
- Dimitrios Petroglou
- 1st Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ilias Kanellos
- 1st Propedeutic Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christos Savopoulos
- 1st Propedeutic Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgia Kaiafa
- 1st Propedeutic Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anastasios Chrysochoou
- 1st Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Panagiotis Skantzis
- 1st Propedeutic Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Stylianos Daios
- 1st Propedeutic Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Apostolos I Hatzitolios
- 1st Propedeutic Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios Giannoglou
- 1st Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
30
|
Zhou ZS, Li MX, Liu J, Jiao H, Xia JM, Shi XJ, Zhao H, Chu L, Liu J, Qi W, Luo J, Song BL. Competitive oxidation and ubiquitylation on the evolutionarily conserved cysteine confer tissue-specific stabilization of Insig-2. Nat Commun 2020; 11:379. [PMID: 31953408 PMCID: PMC6969111 DOI: 10.1038/s41467-019-14231-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 12/19/2019] [Indexed: 11/09/2022] Open
Abstract
Insig-2 is an ER membrane protein negatively controlling lipid biosynthesis. Here, we find that Insig-2 is increased in the tissues, including liver, but unaltered in the muscle of gp78-deficient mice. In hepatocytes and undifferentiated C2C12 myoblasts, Insig-2 is ubiquitylated on Cys215 by gp78 and degraded. However, the C215 residue is oxidized by elevated reactive oxygen species (ROS) during C2C12 myoblasts differentiating into myotubes, preventing Insig-2 from ubiquitylation and degradation. The stabilized Insig-2 downregulates lipogenesis through inhibiting the SREBP pathway, helping to channel the carbon flux to ATP generation and protecting myotubes from lipid over-accumulation. Evolutionary analysis shows that the YECK (in which C represents Cys215 in human Insig-2) tetrapeptide sequence in Insig-2 is highly conserved in amniotes but not in aquatic amphibians and fishes, suggesting it may have been shaped by differential selection. Together, this study suggests that competitive oxidation-ubiquitylation on Cys215 of Insig-2 senses ROS and prevents muscle cells from lipid accumulation.
Collapse
Affiliation(s)
- Zhang-Sen Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Mei-Xin Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Jie Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Hengwu Jiao
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Jing-Ming Xia
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Xiong-Jie Shi
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Huabin Zhao
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Liping Chu
- School of Life Science and Technology, ShanghaiTech Universiy, Shanghai, 201203, China
| | - Jingrong Liu
- School of Life Science and Technology, ShanghaiTech Universiy, Shanghai, 201203, China
| | - Wei Qi
- School of Life Science and Technology, ShanghaiTech Universiy, Shanghai, 201203, China
| | - Jie Luo
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Bao-Liang Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
31
|
Coates HW, Chua NK, Brown AJ. Consulting prostate cancer cohort data uncovers transcriptional control: Regulation of the MARCH6 gene. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1656-1668. [PMID: 31422115 DOI: 10.1016/j.bbalip.2019.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 08/01/2019] [Accepted: 08/14/2019] [Indexed: 12/28/2022]
Abstract
Cholesterol accumulation is a hallmark of prostate cancer (PCa) enabled by the upregulation of its synthesis, which presents a potential therapeutic target. This pathway is suppressed by the E3 ubiquitin ligase membrane-associated RING-CH-type finger 6 (MARCH6); however, little is known of MARCH6 regulation, particularly at the transcriptional level. Here, we consulted large transcriptomic PCa datasets to investigate transcription factors and DNA sequence elements that regulate the MARCH6 gene. Amongst 498 primary PCa tissues of The Cancer Genome Atlas, we identified a striking positive correlation between MARCH6 and androgen receptor (AR) gene expression (r = 0.81, p < 1 × 10-117) that held in other primary tumour datasets. Two putative androgen response elements were identified in the MARCH6 gene using motif prediction and mining of publicly accessible chromatin immunoprecipitation-sequencing data. However, MARCH6 expression was not androgen-responsive in luciferase reporter and qRT-PCR assays. Instead, we established that the MARCH6-AR correlation in primary PCa is due to common regulation by the transcription factor Sp1. We located a region 100 bp downstream of the MARCH6 transcriptional start site that contains three Sp1 binding sites and strongly upregulates promoter activity. The functionality of this region, and Sp1-mediated upregulation of MARCH6, was confirmed using pharmacological and genetic inhibition of Sp1. Moreover, modulation of Sp1 activity affected the stability of squalene monooxygenase, a cholesterol biosynthesis enzyme and MARCH6 substrate. We thus establish Sp1 as the first known regulator of the MARCH6 gene and demonstrate that interrogation of transcriptomic datasets can assist in the de novo inference of transcriptional regulation.
Collapse
Affiliation(s)
- Hudson W Coates
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, Australia
| | - Ngee Kiat Chua
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, Australia
| | - Andrew J Brown
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, Australia.
| |
Collapse
|
32
|
McClellan AJ, Laugesen SH, Ellgaard L. Cellular functions and molecular mechanisms of non-lysine ubiquitination. Open Biol 2019; 9:190147. [PMID: 31530095 PMCID: PMC6769291 DOI: 10.1098/rsob.190147] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Protein ubiquitination is of great cellular importance through its central role in processes such as degradation, DNA repair, endocytosis and inflammation. Canonical ubiquitination takes place on lysine residues, but in the past 15 years non-lysine ubiquitination on serine, threonine and cysteine has been firmly established. With the emerging importance of non-lysine ubiquitination, it is crucial to identify the responsible molecular machinery and understand the mechanistic basis for non-lysine ubiquitination. Here, we first provide an overview of the literature that has documented non-lysine ubiquitination. Informed by these examples, we then discuss the molecular mechanisms and cellular implications of non-lysine ubiquitination, and conclude by outlining open questions and future perspectives in the field.
Collapse
Affiliation(s)
- Amie J McClellan
- Division of Science and Mathematics, Bennington College, 1 College Drive, Bennington, VT 05201, USA
| | - Sophie Heiden Laugesen
- Department of Biology, Linderstrøm-Lang Centre for Protein Science, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Lars Ellgaard
- Department of Biology, Linderstrøm-Lang Centre for Protein Science, University of Copenhagen, 2200 Copenhagen N, Denmark
| |
Collapse
|
33
|
Cholesterol transport through the peroxisome-ER membrane contacts tethered by PI(4,5)P 2 and extended synaptotagmins. SCIENCE CHINA-LIFE SCIENCES 2019; 62:1117-1135. [PMID: 31144242 DOI: 10.1007/s11427-019-9569-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 10/26/2022]
Abstract
Most mammalian cells take up cholesterol from low-density lipoproteins (LDLs) via receptor-mediated endocytosis. After reaching lysosomes, LDL-derived cholesterol continues to transport to downstream organelles including the ER for specific structural and functional needs. Peroxisomes are recently found to receive cholesterol from lysosomes through lysosome-peroxisome membrane contacts. However, whether and how cholesterol is conveyed from peroxisomes to the ER remain unknown. Here, by combining high-resolution microscopic analyses and in vitro reconstitution of highly purified organelles or artificial liposomes, we demonstrate that peroxisomes form membrane contacts with the ER through the interaction between peroxisomal PI(4,5)P2 and ER-resident extended synaptotagmin-1, 2 and 3 (E-Syts). Depletion of peroxisomal PI(4,5)P2 or E-Syts markedly decreases peroxisome-ER membrane contacts and induces cholesterol accumulation in lysosomes. Furthermore, we show that cholesterol is delivered from 3H-labeled peroxisomes or PI(4,5)P2-containing liposomes to the ER in vitro, and that the presence of peroxisomes augments cholesterol transfer from lysosomes to the ER. Together, our study reveals a new cholesterol transport pathway along the lysosome-peroxisome-ER membrane contacts in the cell.
Collapse
|
34
|
Malhotra P, Soni V, Yamanashi Y, Takada T, Suzuki H, Gill RK, Saksena S, Dudeja PK, Alrefai WA. Mechanisms of Niemann-Pick type C1 Like 1 protein degradation in intestinal epithelial cells. Am J Physiol Cell Physiol 2019; 316:C559-C566. [PMID: 30789754 PMCID: PMC6482670 DOI: 10.1152/ajpcell.00465.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/24/2018] [Accepted: 02/12/2019] [Indexed: 01/06/2023]
Abstract
Intestinal Niemann-Pick C1 Like 1 (NPC1L1) protein plays a key role in cholesterol absorption. A decrease in NPC1L1 expression has been implicated in lowering plasma cholesterol and mitigating the risk for coronary heart disease. Little is known about the mechanisms responsible for NPC1L1 protein degradation that upon activation may lead to a reduction in NPC1L1 protein levels in intestinal epithelial cells (IECs). In current studies, the human intestinal Caco-2 and HuTu-80 cell lines expressing NPC1L1-hemagglutinin fusion protein were used to investigate the mechanisms of NPC1L1 protein degradation. Incubation with the proteasome inhibitors MG-132 and lactacystin (10 μM, 24 h) significantly increased NPC1L1 protein levels in IECs. Also, the inhibition of the lysosomal pathway with bafilomycin A1 (80 nM, 24 h) resulted in a significant increase in NPC1L1 protein levels. Immunoprecipitation studies showed that NPC1L1 protein is both a poly- and monoubiquinated polypeptide and that the inhibition of the proteasomal pathway remarkably increased the level of the polyubiquinated NPC1L1. The surface expression of NPC1L1 was increased by the inhibition of both proteasomal and lysosomal pathways. Furthermore, the pharmacological inhibition of mitogen-activated protein kinase pathway (PD-98059, 15 μM, 24 h) and siRNA silencing of ERK1/2 resulted in a significant decrease in NPC1L1 protein levels in IECs. In conclusion, our results showed that basal level of intestinal cholesterol transporter NPC1L1 protein is modulated by both ubiquitin proteasome- and lysosome-dependent degradation as well as by ERK1/2-dependent pathway. The modulation of these pathways may provide novel clues for therapeutic intervention to inhibit cholesterol absorption and lower plasma cholesterol.
Collapse
Affiliation(s)
- Pooja Malhotra
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Vinay Soni
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Yoshihide Yamanashi
- Department of Pharmacy, The University of Tokyo Hospital , Tokyo , Japan
- Faculty of Medicine, The University of Tokyo , Tokyo , Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital , Tokyo , Japan
- Faculty of Medicine, The University of Tokyo , Tokyo , Japan
| | - Hiroshi Suzuki
- Department of Pharmacy, The University of Tokyo Hospital , Tokyo , Japan
- Faculty of Medicine, The University of Tokyo , Tokyo , Japan
| | - Ravinder K Gill
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Seema Saksena
- Jesse Brown Veterans Affairs Medical Center , Chicago, Illinois
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Pradeep K Dudeja
- Jesse Brown Veterans Affairs Medical Center , Chicago, Illinois
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Waddah A Alrefai
- Jesse Brown Veterans Affairs Medical Center , Chicago, Illinois
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| |
Collapse
|
35
|
van der Kant R, Langness VF, Herrera CM, Williams DA, Fong LK, Leestemaker Y, Steenvoorden E, Rynearson KD, Brouwers JF, Helms JB, Ovaa H, Giera M, Wagner SL, Bang AG, Goldstein LSB. Cholesterol Metabolism Is a Druggable Axis that Independently Regulates Tau and Amyloid-β in iPSC-Derived Alzheimer's Disease Neurons. Cell Stem Cell 2019; 24:363-375.e9. [PMID: 30686764 PMCID: PMC6414424 DOI: 10.1016/j.stem.2018.12.013] [Citation(s) in RCA: 227] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 09/26/2018] [Accepted: 12/17/2018] [Indexed: 02/07/2023]
Abstract
Genetic, epidemiologic, and biochemical evidence suggests that predisposition to Alzheimer's disease (AD) may arise from altered cholesterol metabolism, although the molecular pathways that may link cholesterol to AD phenotypes are only partially understood. Here, we perform a phenotypic screen for pTau accumulation in AD-patient iPSC-derived neurons and identify cholesteryl esters (CE), the storage product of excess cholesterol, as upstream regulators of Tau early during AD development. Using isogenic induced pluripotent stem cell (iPSC) lines carrying mutations in the cholesterol-binding domain of APP or APP null alleles, we found that while CE also regulate Aβ secretion, the effects of CE on Tau and Aβ are mediated by independent pathways. Efficacy and toxicity screening in iPSC-derived astrocytes and neurons showed that allosteric activation of CYP46A1 lowers CE specifically in neurons and is well tolerated by astrocytes. These data reveal that CE independently regulate Tau and Aβ and identify a druggable CYP46A1-CE-Tau axis in AD.
Collapse
Affiliation(s)
- Rik van der Kant
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam de Boelelaan 1087, 1081 HV Amsterdam, the Netherlands
| | - Vanessa F Langness
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Cheryl M Herrera
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel A Williams
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lauren K Fong
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yves Leestemaker
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, the Netherlands
| | - Evelyne Steenvoorden
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Kevin D Rynearson
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jos F Brouwers
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University Yalelaan 2, 3584 CM Utrecht, the Netherlands
| | - J Bernd Helms
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University Yalelaan 2, 3584 CM Utrecht, the Netherlands
| | - Huib Ovaa
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, the Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Steven L Wagner
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA; Research Biologist, VA San Diego Healthcare System, La Jolla, CA 92161, USA
| | - Anne G Bang
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Lawrence S B Goldstein
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA.
| |
Collapse
|
36
|
Králová J, Král V. Fluorescent Probes for Monitoring Cholesterol Trafficking in Cells. Folia Biol (Praha) 2019; 65:1-10. [PMID: 31171077 DOI: 10.14712/fb2019065010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Cellular cholesterol plays fundamental and diverse roles in many biological processes and affects the pathology of various diseases. Comprehensive and detailed understanding of the cellular functions and characteristics of cholesterol requires visualization of its subcellular distribution, which can be achieved by fluorescence microscopy. Many attempts have been made to develop fluorescent cholesterol reporters, but so far, none of them seems to be ideal for studying all aspects of cholesterol management. To meet the requirements for the right probe remains a great challenge, and progress in this field continues. The main objective of this review is to not only present the current state of the art, but also critically evaluate the applicability of individual probes and for what purpose they can be used to obtain relevant data. Hence, the data obtained with different probes might provide complementary information to build an integrated picture about the cellular cholesterol.
Collapse
Affiliation(s)
- J Králová
- CZ-OPENSCREEN, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - V Král
- University of Chemistry and Technology, Prague, Czech Republic
| |
Collapse
|
37
|
Tan JME, Cook ECL, van den Berg M, Scheij S, Zelcer N, Loregger A. Differential use of E2 ubiquitin conjugating enzymes for regulated degradation of the rate-limiting enzymes HMGCR and SQLE in cholesterol biosynthesis. Atherosclerosis 2018; 281:137-142. [PMID: 30658189 DOI: 10.1016/j.atherosclerosis.2018.12.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/03/2018] [Accepted: 12/13/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND AIMS Cholesterol is an essential lipid for cellular function and membrane integrity, and hence its cellular levels and distribution must be tightly regulated. Biosynthesis of cholesterol is ramped when its cellular levels are low. Herein, the ER-resident and rate-limiting enzymes 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) and squalene monooxygenase (SQLE) play a prominent role. We have recently reported that MARCH6, an E3 ubiquitin ligase, specifically promotes cholesterol-stimulated ubiquitylation and subsequent proteasomal degradation of SQLE, but not of HMGCR. To further delineate how post-translational regulation of SQLE and HMGCR is differentially achieved, we hypothesized that their sterol-dependent degradation machinery makes use of distinct E2 ubiquitin conjugating enzymes. METHODS To study this possibility, we therefore used a CRISPR/Cas9-based approach to screen for ER-associated degradation (ERAD)-associated E2 enzymes that are essential for MARCH6-dependent degradation of SQLE. RESULTS We report here the identification of UBE2J2 as the primary E2 ubiquitin conjugating enzyme essential for this process in mammalian cells, in contrast to UBE2G2, which is essential for sterol-stimulated degradation of HMGCR. We demonstrate that ablating UBE2J2 disturbs cholesterol-accelerated SQLE degradation in multiple human cell types, including cells of hepatic origin, and that the ability of UBE2J2 to support SQLE degradation critically depends on its enzymatic activity. CONCLUSIONS Our findings establish UBE2J2 as an important partner of MARCH6 in cholesterol-stimulated degradation of SQLE, thereby contributing to the complex regulation of cellular cholesterol homeostasis.
Collapse
Affiliation(s)
- Josephine M E Tan
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, the Netherlands
| | - Emma C L Cook
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, the Netherlands
| | - Marlene van den Berg
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, the Netherlands
| | - Saskia Scheij
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, the Netherlands
| | - Noam Zelcer
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, the Netherlands.
| | - Anke Loregger
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, the Netherlands.
| |
Collapse
|
38
|
Li L, Xu L, Chen W, Li X, Xia Q, Zheng L, Duan Q, Zhang H, Zhao Y. Reduced Annexin A1 Secretion by ABCA1 Causes Retinal Inflammation and Ganglion Cell Apoptosis in a Murine Glaucoma Model. Front Cell Neurosci 2018; 12:347. [PMID: 30364320 PMCID: PMC6193130 DOI: 10.3389/fncel.2018.00347] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/18/2018] [Indexed: 12/15/2022] Open
Abstract
Variants near the ATP-binding cassette transporter A1 (ABCA1) gene are associated with elevated intraocular pressure and newly discovered risk factors for glaucoma. Previous studies have shown an association between ABCA1 deficiency and retinal inflammation. Using a mouse model of ischemia-reperfusion (IR) induced by acute intraocular pressure elevation, we found that the retinal expression of ABCA1 protein was decreased. An induction of ABCA1 expression by liver X receptor agonist TO901317 reduced retinal ganglion cell (RGC) apoptosis after IR and promoted membrane translocation and secretion of the anti-inflammatory factor annexin A1 (ANXA1). Moreover, ABCA1 and ANXA1 co-localized in cell membranes, and the interaction domain is amino acid 196 to 274 of ANXA1 fragment. TO901317 also reduced microglia migration and activation and decreased the expression of pro-inflammatory cytokines interleukin (IL)-17A and IL-1β, which could be reversed by the ANXA1 receptor blocker Boc2. Overexpression of TANK-binding kinase 1 (TBK1) increased ABCA1 degradation, which was reversed by the proteasome inhibitor carbobenzoxy-L-leucyl-L-leucyl-L-leucinal (MG132). Silencing Tbk1 with siRNA increased ABCA1 expression and promoted ANXA1 membrane translocation. These results indicate a novel IR mechanism, that leads via TBK1 activation to ABCA1 ubiquitination. This degradation decreases ANXA1 secretion, thus facilitating retinal inflammation and RGC apoptosis. Our findings suggest a potential treatment strategy to prevent RGC apoptosis in retinal ischemia and glaucoma.
Collapse
Affiliation(s)
- Lu Li
- Department of Ophthalmology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Department of Ophthalmology, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
| | - Lingjuan Xu
- Department of Ophthalmology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Chen
- Department of Ophthalmology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Li
- Key Laboratory of Neurological Diseases, Department of Neurobiology, Ministry of Education, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Xia
- Key Laboratory of Neurological Diseases, Department of Neurobiology, Ministry of Education, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Zheng
- Key Laboratory of Neurological Diseases, Department of Neurobiology, Ministry of Education, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Qiming Duan
- Gladstone Institutes, San Francisco, CA, United States
| | - Hong Zhang
- Department of Ophthalmology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yin Zhao
- Department of Ophthalmology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
39
|
Tegge AN, Rodrigues RR, Larkin AL, Vu L, Murali TM, Rajagopalan P. Transcriptomic Analysis of Hepatic Cells in Multicellular Organotypic Liver Models. Sci Rep 2018; 8:11306. [PMID: 30054499 PMCID: PMC6063915 DOI: 10.1038/s41598-018-29455-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 07/11/2018] [Indexed: 02/08/2023] Open
Abstract
Liver homeostasis requires the presence of both parenchymal and non-parenchymal cells (NPCs). However, systems biology studies of the liver have primarily focused on hepatocytes. Using an organotypic three-dimensional (3D) hepatic culture, we report the first transcriptomic study of liver sinusoidal endothelial cells (LSECs) and Kupffer cells (KCs) cultured with hepatocytes. Through computational pathway and interaction network analyses, we demonstrate that hepatocytes, LSECs and KCs have distinct expression profiles and functional characteristics. Our results show that LSECs in the presence of KCs exhibit decreased expression of focal adhesion kinase (FAK) signaling, a pathway linked to LSEC dedifferentiation. We report the novel result that peroxisome proliferator-activated receptor alpha (PPARα) is transcribed in LSECs. The expression of downstream processes corroborates active PPARα signaling in LSECs. We uncover transcriptional evidence in LSECs for a feedback mechanism between PPARα and farnesoid X-activated receptor (FXR) that maintains bile acid homeostasis; previously, this feedback was known occur only in HepG2 cells. We demonstrate that KCs in 3D liver models display expression patterns consistent with an anti-inflammatory phenotype when compared to monocultures. These results highlight the distinct roles of LSECs and KCs in maintaining liver function and emphasize the need for additional mechanistic studies of NPCs in addition to hepatocytes in liver-mimetic microenvironments.
Collapse
Affiliation(s)
- Allison N Tegge
- Department of Computer Science, Virginia Tech, Blacksburg, USA
- Department of Statistics, Virginia Tech, Blacksburg, USA
| | - Richard R Rodrigues
- Genetics, Bioinformatics, and Computational Biology Ph.D. Program, Virginia Tech, Blacksburg, USA
| | - Adam L Larkin
- Department of Chemical Engineering, Virginia Tech, Blacksburg, USA
| | - Lucas Vu
- Department of Chemical Engineering, Virginia Tech, Blacksburg, USA
| | - T M Murali
- Department of Computer Science, Virginia Tech, Blacksburg, USA.
- ICTAS Center for Systems Biology of Engineered Tissues, Virginia Tech, Blacksburg, USA.
| | - Padmavathy Rajagopalan
- Department of Chemical Engineering, Virginia Tech, Blacksburg, USA.
- ICTAS Center for Systems Biology of Engineered Tissues, Virginia Tech, Blacksburg, USA.
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, USA.
| |
Collapse
|
40
|
Wangeline MA, Vashistha N, Hampton RY. Proteostatic Tactics in the Strategy of Sterol Regulation. Annu Rev Cell Dev Biol 2018; 33:467-489. [PMID: 28992438 DOI: 10.1146/annurev-cellbio-111315-125036] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In eukaryotes, the synthesis and uptake of sterols undergo stringent multivalent regulation. Both individual enzymes and transcriptional networks are controlled to meet changing needs of the many sterol pathway products. Regulation is tailored by evolution to match regulatory constraints, which can be very different in distinct species. Nevertheless, a broadly conserved feature of many aspects of sterol regulation is employment of proteostasis mechanisms to bring about control of individual proteins. Proteostasis is the set of processes that maintain homeostasis of a dynamic proteome. Proteostasis includes protein quality control pathways for the detection, and then the correction or destruction, of the many misfolded proteins that arise as an unavoidable feature of protein-based life. Protein quality control displays not only the remarkable breadth needed to manage the wide variety of client molecules, but also extreme specificity toward the misfolded variants of a given protein. These features are amenable to evolutionary usurpation as a means to regulate proteins, and this approach has been used in sterol regulation. We describe both well-trod and less familiar versions of the interface between proteostasis and sterol regulation and suggest some underlying ideas with broad biological and clinical applicability.
Collapse
Affiliation(s)
- Margaret A Wangeline
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093;
| | - Nidhi Vashistha
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093;
| | - Randolph Y Hampton
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093;
| |
Collapse
|
41
|
Ma X, Bi E, Huang C, Lu Y, Xue G, Guo X, Wang A, Yang M, Qian J, Dong C, Yi Q. Cholesterol negatively regulates IL-9-producing CD8 + T cell differentiation and antitumor activity. J Exp Med 2018; 215:1555-1569. [PMID: 29743292 PMCID: PMC5987919 DOI: 10.1084/jem.20171576] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 03/19/2018] [Accepted: 04/23/2018] [Indexed: 12/17/2022] Open
Abstract
CD8+ T cells can be polarized into IL-9-secreting (Tc9) cells. We previously showed that adoptive therapy using tumor-specific Tc9 cells generated stronger antitumor responses in mouse melanoma than classical Tc1 cells. To understand why Tc9 cells exert stronger antitumor responses, we used gene profiling to compare Tc9 and Tc1 cells. Tc9 cells expressed different levels of cholesterol synthesis and efflux genes and possessed significantly lower cholesterol content than Tc1 cells. Unique to Tc9, but not other CD8+ or CD4+ T cell subsets, manipulating cholesterol content in polarizing Tc9 cells significantly affected IL-9 expression and Tc9 differentiation and antitumor response in vivo. Mechanistic studies showed that IL-9 was indispensable for Tc9 cell persistence and antitumor effects, and cholesterol or its derivatives inhibited IL-9 expression by activating liver X receptors (LXRs), leading to LXR Sumoylation and reduced p65 binding to Il9 promoter. Our study identifies cholesterol as a critical regulator of Tc9 cell differentiation and function.
Collapse
Affiliation(s)
- Xingzhe Ma
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Enguang Bi
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Chunjian Huang
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Yong Lu
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Gang Xue
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Xing Guo
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Aibo Wang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Maojie Yang
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Jianfei Qian
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Qing Yi
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
42
|
Abstract
The objective of this study was to assess the effects of proteasome inhibition on the development of burn-induced hypermetabolism. Rats underwent 30-40% total BSA scald burn or sham injury. The proteasome inhibitor bortezomib (0.1 mg/kg) or vehicle (n = 10) was administered i.p. 3× weekly starting at 2 hours (early bortezomib, n = 20) or 48 hours (late-bortezomib, n = 13) postburn. Body weights were determined weekly. Resting energy expenditures (REE) were measured at days 0 (baseline), 7, 14, 21, and 42 postburn. At day 42, blood and pectoral muscle were harvested. Routine blood chemistry parameters were analyzed. Proteasome content, proteasome peptidase activities, and ubiquitin-protein conjugates were measured in muscle extracts. As compared with sham-vehicle-treated animals, specific proteasome activities were increased after burn and vehicle treatment. Bortezomib treatment inhibited proteasome activities and increased ubiquitin-protein conjugates after sham and burn injury. Bortezomib treatment did not affect REE after sham procedure. REE significantly increased by 47% within 7 days and remained elevated until day 42 after burn and vehicle treatment. After early-bortezomib treatment, burn-induced increases in REE were delayed and significantly reduced by 42% at day 42, as compared with vehicle treatment. With late-bortezomib treatment, burn-induced increases in REE were also delayed but not attenuated at day 42. Mortality was 20% with vehicle, 65% (median survival time: 1.875 days) with early-bortezomib and 25% with late-bortezomib treatment after burns (P < .05 early-bortezomib vs vehicle and late-bortezomib). Proteasome inhibition delays development of burn-induced hypermetabolism. Although proteasome inhibition early after burn injury reduces the hypermetabolic response, it significantly increases early burn-associated mortality.
Collapse
|
43
|
Gil-Ramírez A, Morales D, Soler-Rivas C. Molecular actions of hypocholesterolaemic compounds from edible mushrooms. Food Funct 2018; 9:53-69. [DOI: 10.1039/c7fo00835j] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Edible mushrooms contain bioactive compounds able to modulate the expression of genes related to absorption, biosynthesis and transport of cholesterol and regulation of its homeostasis.
Collapse
Affiliation(s)
- Alicia Gil-Ramírez
- Department of Production and Characterization of Novel Foods
- CIAL – Research Institute in Food Science (UAM+CSIC)
- C/Nicolas Cabrera 9
- Campus de Cantoblanco
- Universidad Autonoma de Madrid
| | - Diego Morales
- Department of Production and Characterization of Novel Foods
- CIAL – Research Institute in Food Science (UAM+CSIC)
- C/Nicolas Cabrera 9
- Campus de Cantoblanco
- Universidad Autonoma de Madrid
| | - Cristina Soler-Rivas
- Department of Production and Characterization of Novel Foods
- CIAL – Research Institute in Food Science (UAM+CSIC)
- C/Nicolas Cabrera 9
- Campus de Cantoblanco
- Universidad Autonoma de Madrid
| |
Collapse
|
44
|
Edvinsson M, Tallkvist J, Nyström-Rosander C, Ilbäck NG. Cholesterol uptake in the mouse aorta increases during Chlamydia pneumoniae infection. Pathog Dis 2017; 75:2966466. [PMID: 28158541 DOI: 10.1093/femspd/ftx004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 02/01/2017] [Indexed: 01/08/2023] Open
Abstract
Chlamydia pneumoniae has been suggested as a stimulator of the atherosclerotic process. Mice fed a normal diet were infected intranasally with C. pneumoniae and given one intraperitoneal injection of 14C-cholesterol tracer per day for 12 days. Bacteria were demonstrated in the aorta in the early phase of infection and in lungs and liver throughout the study period of 20 days. 14C-cholesterol was not affected in the heart but increased in the blood, liver and aorta on day 4 when the infection was clinically most severe. Furthermore, on day 20 14C-cholesterol tended to be increased in the aorta. Accordingly, copper- and zinc levels and expressions of the infection biomarkers Cxcl2 and Ifng increased in the liver on day 4 with a tendency of increased of copper, zinc and Ifng on day 20. In mice where bacteria could be cultivated from the lungs, expressions of cholesterol transporters Abca1 and Idol were both increased in the liver on day 4. The increased levels of 14C-cholesterol in blood and aorta together with increased Abca1 and Idol in the liver during C. pneumoniae infection in mice fed a normal diet suggest that this pathogen may have a role in the initiation of the atherosclerotic process.
Collapse
Affiliation(s)
- Marie Edvinsson
- Department of Medical Sciences, Section of Infectious Diseases, Uppsala University, 75185 Uppsala, Sweden
| | - Jonas Tallkvist
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, 75007 Uppsala, Sweden
| | - Christina Nyström-Rosander
- Department of Medical Sciences, Section of Infectious Diseases, Uppsala University, 75185 Uppsala, Sweden
| | - Nils-Gunnar Ilbäck
- Department of Medical Sciences, Section of Infectious Diseases, Uppsala University, 75185 Uppsala, Sweden.,Risk Benefit Assessment Department, National Food Agency, 75126 Uppsala, Sweden
| |
Collapse
|
45
|
Loregger A, Raaben M, Tan J, Scheij S, Moeton M, van den Berg M, Gelberg-Etel H, Stickel E, Roitelman J, Brummelkamp T, Zelcer N. Haploid Mammalian Genetic Screen Identifies UBXD8 as a Key Determinant of HMGCR Degradation and Cholesterol Biosynthesis. Arterioscler Thromb Vasc Biol 2017; 37:2064-2074. [PMID: 28882874 PMCID: PMC5671778 DOI: 10.1161/atvbaha.117.310002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 08/29/2017] [Indexed: 01/23/2023]
Abstract
Supplemental Digital Content is available in the text. Objective— The cellular demand for cholesterol requires control of its biosynthesis by the mevalonate pathway. Regulation of HMGCR (3-hydroxy-3-methylglutaryl coenzyme A reductase), a rate-limiting enzyme in this pathway and the target of statins, is a key control point herein. Accordingly, HMGCR is subject to negative and positive regulation. In particular, the ability of oxysterols and intermediates of the mevalonate pathway to stimulate its proteasomal degradation is an exquisite example of metabolically controlled feedback regulation. To define the genetic determinants that govern this process, we conducted an unbiased haploid mammalian genetic screen. Approach and Results— We generated human haploid cells with mNeon fused to endogenous HMGCR using CRISPR/Cas9 and used these cells to interrogate regulation of HMGCR abundance in live cells. This resulted in identification of known and new regulators of HMGCR, and among the latter, UBXD8 (ubiquitin regulatory X domain-containing protein 8), a gene that has not been previously implicated in this process. We demonstrate that UBXD8 is an essential determinant of metabolically stimulated degradation of HMGCR and of cholesterol biosynthesis in multiple cell types. Accordingly, UBXD8 ablation leads to aberrant cholesterol synthesis due to loss of feedback control. Mechanistically, we show that UBXD8 is necessary for sterol-stimulated dislocation of ubiquitylated HMGCR from the endoplasmic reticulum membrane en route to proteasomal degradation, a function dependent on its UBX domain. Conclusions— We establish UBXD8 as a previously unrecognized determinant that couples flux across the mevalonate pathway to control of cholesterol synthesis and demonstrate the feasibility of applying mammalian haploid genetics to study metabolic traits.
Collapse
Affiliation(s)
- Anke Loregger
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Matthijs Raaben
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Josephine Tan
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Saskia Scheij
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Martina Moeton
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Marlene van den Berg
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Hila Gelberg-Etel
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Elmer Stickel
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Joseph Roitelman
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Thijn Brummelkamp
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Noam Zelcer
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.).
| |
Collapse
|
46
|
Jovandaric MZ, Nikolic TV, Milenkovic SJ, Otaševic BS, Bankovic VV, Ivanovski PI, Jesic MM. Lipids on the Second Day in Ischemic and Normoxemic Term Neonates. Fetal Pediatr Pathol 2017; 36:276-281. [PMID: 28453380 DOI: 10.1080/15513815.2017.1313914] [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: 10/19/2022]
Abstract
INTRODUCTION In hypoxic newborns requiring oxygen, lipid peroxidation affects the peripheral blood lipids. OBJECTIVES Determine the influence of perinatal oxygen therapy for hypoxia on serum lipid concentrations on the second day of life. MATERIALS AND METHODS Our study included 50 newborns with perinatal hypoxia requiring oxygen and 50 healthy newborns without oxygen therapy. Arterialized capillary blood was taken for categorization of hypoxia (pO2) after birth in both groups. Lipid concentrations: total cholesterol (TC), high density lipoproteins (HDL), low density lipoproteins (LDL), and triglycerides (TG) were measured on day 2 in both groups. RESULTS TC, LDL, HDL, TG, HC03 levels were statistically lower in the study group compared to the control one, while pCO2 and BE levels were statistically higher in newborns with perinatal hypoxia. CONCLUSION Lower lipid levels in hypoxic newborns may suggest that circulating lipids are oxidized, peroxidized, and removed from the peripheral circulation.
Collapse
Affiliation(s)
- Miljana Z Jovandaric
- a Department Neonatology, Clinic for Ginecology and Obstetrics , Clinical Centre of Serbia , Belgrade , Serbia
| | - Tatjana V Nikolic
- a Department Neonatology, Clinic for Ginecology and Obstetrics , Clinical Centre of Serbia , Belgrade , Serbia
| | - Svetlana J Milenkovic
- a Department Neonatology, Clinic for Ginecology and Obstetrics , Clinical Centre of Serbia , Belgrade , Serbia
| | - Biljana S Otaševic
- a Department Neonatology, Clinic for Ginecology and Obstetrics , Clinical Centre of Serbia , Belgrade , Serbia
| | - Violeta V Bankovic
- a Department Neonatology, Clinic for Ginecology and Obstetrics , Clinical Centre of Serbia , Belgrade , Serbia
| | - Petar I Ivanovski
- b Department of Hematology , University Children's Hospital, School of Medicine, University of Belgrade , Belgrade , Serbia
| | - Milos M Jesic
- c Department of Neonatology , University Children's Hospital, School of Medicine, University of Belgrade , Belgrade , Serbia
| |
Collapse
|
47
|
Wang YJ, Bian Y, Luo J, Lu M, Xiong Y, Guo SY, Yin HY, Lin X, Li Q, Chang CCY, Chang TY, Li BL, Song BL. Cholesterol and fatty acids regulate cysteine ubiquitylation of ACAT2 through competitive oxidation. Nat Cell Biol 2017; 19:808-819. [PMID: 28604676 PMCID: PMC5518634 DOI: 10.1038/ncb3551] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 05/10/2017] [Indexed: 12/26/2022]
Abstract
Ubiquitin linkage to cysteine is an unconventional modification targeting protein for degradation. However, the physiological regulation of cysteine ubiquitylation is still mysterious. Here we found that ACAT2, a cellular enzyme converting cholesterol and fatty acid to cholesteryl esters, was ubiquitylated on Cys277 for degradation when the lipid level was low. gp78-Insigs catalysed Lys48-linked polyubiquitylation on this Cys277. A high concentration of cholesterol and fatty acid, however, induced cellular reactive oxygen species (ROS) that oxidized Cys277, resulting in ACAT2 stabilization and subsequently elevated cholesteryl esters. Furthermore, ACAT2 knockout mice were more susceptible to high-fat diet-associated insulin resistance. By contrast, expression of a constitutively stable form of ACAT2 (C277A) resulted in higher insulin sensitivity. Together, these data indicate that lipid-induced stabilization of ACAT2 ameliorates lipotoxicity from excessive cholesterol and fatty acid. This unconventional cysteine ubiquitylation of ACAT2 constitutes an important mechanism for sensing lipid-overload-induced ROS and fine-tuning lipid homeostasis.
Collapse
Affiliation(s)
- Yong-Jian Wang
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu, China
| | - Yan Bian
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Jie Luo
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| | - Ming Lu
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Ying Xiong
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Shu-Yuan Guo
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hui-Yong Yin
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xu Lin
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qin Li
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Catherine CY Chang
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Ta-Yuan Chang
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Bo-Liang Li
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Bao-Liang Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| |
Collapse
|
48
|
Identification of the ER-resident E3 ubiquitin ligase RNF145 as a novel LXR-regulated gene. PLoS One 2017; 12:e0172721. [PMID: 28231341 PMCID: PMC5322959 DOI: 10.1371/journal.pone.0172721] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/08/2017] [Indexed: 12/12/2022] Open
Abstract
Cellular cholesterol metabolism is subject to tight regulation to maintain adequate levels of this central lipid molecule. Herein, the sterol-responsive Liver X Receptors (LXRs) play an important role owing to their ability to reduce cellular cholesterol load. In this context, identifying the full set of LXR-regulated genes will contribute to our understanding of their role in cholesterol metabolism. Using global transcriptional analysis we report here the identification of RNF145 as an LXR-regulated target gene. We demonstrate that RNF145 is regulated by LXRs in both human and mouse primary cells and cell lines, and in vivo in mice. Regulation of RNF145 by LXR depends on a functional LXR-element in its proximal promotor. Consistent with LXR-dependent regulation of Rnf145 we show that regulation is lost in macrophages and fibroblasts from Lxrαβ(-/-) mice, and also in vivo in livers of Lxrα(-/-) mice treated with the LXR synthetic ligand T0901317. RNF145 is closely related to RNF139/TRC8, an E3 ligase implicated in control of SREBP processing. However, silencing of RNF145 in HepG2 or HeLa cells does not impair SREBP1/2 processing and sterol-responsive gene expression in these cells. Similar to TRC8, we demonstrate that RNF145 is localized to the ER and that it possesses intrinsic E3 ubiquitin ligase activity. In summary, we report the identification of RNF145 as an ER-resident E3 ubiquitin ligase that is transcriptionally controlled by LXR.
Collapse
|
49
|
Lawson JS. Multiple Infectious Agents and the Origins of Atherosclerotic Coronary Artery Disease. Front Cardiovasc Med 2016; 3:30. [PMID: 27672638 PMCID: PMC5018484 DOI: 10.3389/fcvm.2016.00030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 08/29/2016] [Indexed: 12/24/2022] Open
Abstract
Although deaths due to atherosclerotic coronary artery disease (ACAD) have fallen dramatically during the past 50 years, ACAD remains as the leading cause of death in all continents, except Africa, where deaths due to infections are still dominant. Although food and nutrition have a proven role in atherosclerosis, the underlying causes of ACAD remain unknown. This is despite a century of intensive research dominated by investigations into the saturated fat hypothesis. In this review, it is hypothesized that the rise and fall in ACAD during the past 100 years is primarily due to the parallel rise and fall in the prevalence of coronary atheroma, the underlying disease. It is further hypothesized that infectious pathogens initiate atherosclerosis mainly during infancy and childhood. It is speculated that widespread use of antibiotics and vaccines against bacterial and viral infections may be the reason for the dramatic fall in coronary atheroma and ACAD during the past 50 years. The relevant evidence and a working hypothesis are included in this review.
Collapse
Affiliation(s)
- James S. Lawson
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
50
|
Affiliation(s)
- Andrew J Brown
- From the School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia (A.J.B.); and Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY (J.H.)
| | - Joanne Hsieh
- From the School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia (A.J.B.); and Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY (J.H.).
| |
Collapse
|