1
|
Lee AK, Welander PV. A geranylgeranyl reductase homolog required for cholesterol production in Myxococcota. J Bacteriol 2025; 207:e0049524. [PMID: 40067012 PMCID: PMC12004948 DOI: 10.1128/jb.00495-24] [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: 11/20/2024] [Accepted: 02/14/2025] [Indexed: 04/18/2025] Open
Abstract
Myxococcota is a phylum of sterol-producing bacteria. They exhibit a clade depth for sterol biosynthesis unparalleled in the bacterial domain and produce sterols of a biosynthetic complexity that rivals eukaryotes. Additionally, the sterol biosynthesis pathways found in this phylum have been proposed as a potential source for sterol biosynthesis in the last eukaryotic common ancestor, lending evolutionary importance to our understanding of this pathway in Myxococcota. However, sterol production has only been characterized in a few species, and outstanding questions about the evolutionary history of this pathway remain. Here, we identify two myxobacteria, Minicystis rosea and Sandaracinus amylolyticus, capable of cholesterol biosynthesis. These two myxobacteria possess a cholesterol biosynthesis pathway that differs in both the ordering and enzymes involved in biosynthesis compared with Enhygromyxa salina, a myxobacterium previously demonstrated to produce cholesterol, as well as the canonical pathways found in eukaryotes. We characterize an alternative bacterial reductase responsible for performing C-24 reduction, further delineating bacterial cholesterol production from eukaryotes. Finally, we examine the distribution and phylogenetic relationships of sterol biosynthesis proteins across both cultured and uncultured Myxococcota species, providing evidence for multiple acquisition events and instances of both horizontal and vertical transfer at the family level. Altogether, this work further demonstrates the capacity of myxobacteria to synthesize eukaryotic sterols but with an underlying diversity in the biochemical reactions that govern sterol synthesis, suggesting a complex evolutionary history and refining our understanding of how myxobacterial cholesterol production relates to their eukaryotic counterparts. IMPORTANCE Sterols are essential and ubiquitous lipids in eukaryotes, but their significance in bacteria is less understood. Sterol production in Myxococcota, a phylum of developmentally complex predatory bacteria, has provided insight into novel sterol biochemistry and prompted discussion regarding the evolution of this pathway within both the eukaryotic and bacterial domains. Here, we characterize cholesterol biosynthesis in two myxobacteria, providing evidence for distinct pathway organization and identifying a unique protein responsible for C-24 reduction. We couple these results with the phylogenomic analysis of sterol biosynthesis within Myxococcota, revealing a complicated evolutionary history marked by vertical and horizontal transfer. This suggests a mosaic acquisition of this pathway in Myxococcota and highlights the complex role myxobacteria may have had in sterol transfer to eukaryotes.
Collapse
Affiliation(s)
- Alysha K. Lee
- Department of Earth Systems Science, Stanford University, Stanford, California, USA
| | - Paula V. Welander
- Department of Earth Systems Science, Stanford University, Stanford, California, USA
| |
Collapse
|
2
|
Chen R, Chen T, Li X, Yu J, Lin M, Wen S, Zhang M, Chen J, Yi B, Zhong H, Li Z. SREBP2 as a central player in cancer progression: potential for targeted therapeutics. Front Pharmacol 2025; 16:1535691. [PMID: 40308757 PMCID: PMC12041066 DOI: 10.3389/fphar.2025.1535691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Recent studies have identified the reprogramming of lipid metabolism as a critical hallmark of malignancy. Enhanced cholesterol uptake and increased cholesterol biosynthesis significantly contribute to the rapid growth of tumors, with cholesterol also playing essential roles in cellular signaling pathways. Targeting cholesterol metabolism has emerged as a promising therapeutic strategy in oncology. The sterol regulatory element-binding protein-2 (SREBP2) serves as a primary transcriptional regulator of genes involved in cholesterol biosynthesis and is crucial for maintaining cholesterol homeostasis. Numerous studies have reported the upregulation of SREBP2 across various cancers, facilitating tumor progression. This review aims to provide a comprehensive overview of the structure, biological functions, and regulatory mechanisms of SREBP2. Furthermore, we summarize that SREBP2 plays a crucial role in various cancers and tumor microenvironment primarily by regulating cholesterol, as well as through several non-cholesterol pathways. We also particularly emphasize therapeutic agents targeting SREBP2 that are currently under investigation. This review seeks to enhance our understanding of SREBP2's involvement in cancer and provide theoretical references for cancer therapies that target SREBP2.
Collapse
Affiliation(s)
- Ruiqi Chen
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Tianyu Chen
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiang Li
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Junfeng Yu
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Min Lin
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Siqi Wen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Man Zhang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jinchi Chen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Bei Yi
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Huage Zhong
- Division of Colorectal and Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, China
| | - Zhao Li
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
3
|
Kato C, Iizuka-Ohashi M, Honda M, Konishi E, Yokota I, Boku S, Mizuta N, Morita M, Sakaguchi K, Taguchi T, Watanabe M, Naoi Y. Additional statin treatment enhances the efficacy of HER2 blockade and improves prognosis in Rac1-high/HER2-positive breast cancer. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167458. [PMID: 39128642 DOI: 10.1016/j.bbadis.2024.167458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 07/26/2024] [Accepted: 08/07/2024] [Indexed: 08/13/2024]
Abstract
The prognosis of HER2-positive breast cancer (BC) has improved with the development of anti-HER2 therapies; however, the problem remains that there are still cases where anti-HER2 therapies do not respond well. We found that the expression of SREBF2, a master transcriptional factor in the mevalonate pathway, was correlated with ERBB2 (HER2) expression and a poor prognosis in HER2-positive BC. The target gene expressions of SREBF2 were associated with higher expression of ERBB2 in HER2-positive BC cells. Statins, anti-hypercholesterolemia drugs that inhibit the mevalonate pathway, enhanced the efficacy of HER2-targeting agents with inducing apoptosis in a geranylgeranylation-dependent manner. Mechanistically, statins specifically inhibited membrane localization of Rac1, a target protein of geranylgeranylation, and suppressed the activation of HER2 downstreams AKT and ERK pathways. Consistently, retrospective analysis showed a longer recurrence-free survival in Rac1-high/HER2-positive BC patients treated with HER2-targeting agents with statins than without statins. Our findings thus suggest that Rac1 expression could be used as a biomarker to stratify HER2-positive BC patients that could benefit from dual blockade, i.e., targeting HER2 with inhibition of geranylgeranylation of Rac1 using statins, thereby opening avenues for precision medicine in a new subset of Rac1-high/HER2-positive BC.
Collapse
Affiliation(s)
- Chikage Kato
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mahiro Iizuka-Ohashi
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mizuki Honda
- Department of Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eiichi Konishi
- Department of Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Isao Yokota
- Department of Biostatistics, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shogen Boku
- Cancer Treatment Center, Kansai Medical University Hospital, Osaka, Japan
| | | | - Midori Morita
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Koichi Sakaguchi
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tetsuya Taguchi
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Motoki Watanabe
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Yasuto Naoi
- Department of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
4
|
Lange Y, Steck TL. How active cholesterol coordinates cell cholesterol homeostasis: Test of a hypothesis. Prog Lipid Res 2024; 96:101304. [PMID: 39491591 DOI: 10.1016/j.plipres.2024.101304] [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: 06/27/2024] [Revised: 10/23/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
How do cells coordinate the diverse elements that regulate their cholesterol homeostasis? Our model postulates that membrane cholesterol forms simple complexes with bilayer phospholipids. The phospholipids in the plasma membrane are of high affinity; consequently, they are fully complexed with the sterol. This sets the resting level of plasma membrane cholesterol. Cholesterol in excess of the stoichiometric equivalence point of these complexes has high chemical activity; we refer to it as active cholesterol. It equilibrates with the low affinity phospholipids in the intracellular membranes where it serves as a negative feedback signal to a manifold of regulatory proteins that rein in ongoing cholesterol accretion. We tested the model with a review of the literature regarding fourteen homeostatic proteins in enterocytes. It provided strong albeit indirect support for the following hypothesis. Active cholesterol inhibits cholesterol uptake and biosynthesis by suppressing both the expression and the activity of the gene products activated by SREBP-2; namely, HMGCR, LDLR and NPC1L1. It also reduces free cell cholesterol by serving as the substrate for its esterification by ACAT and for the synthesis of side-chain oxysterols, 27-hydroxycholesterol in particular. The oxysterols drive cholesterol depletion by promoting the destruction of HMGCR and stimulating sterol esterification as well as the activation of LXR. The latter fosters the expression of multiple homeostatic proteins, including four transporters for which active cholesterol is the likely substrate. By nulling active cholesterol, the manifold maintains the cellular sterol at its physiologic set point.
Collapse
Affiliation(s)
- Yvonne Lange
- Department of Pathology, Rush University Medical Center, Chicago, IL 60612, United States of America.
| | - Theodore L Steck
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, United States of America
| |
Collapse
|
5
|
Benatzy Y, Palmer MA, Lütjohann D, Ohno RI, Kampschulte N, Schebb NH, Fuhrmann DC, Snodgrass RG, Brüne B. ALOX15B controls macrophage cholesterol homeostasis via lipid peroxidation, ERK1/2 and SREBP2. Redox Biol 2024; 72:103149. [PMID: 38581859 PMCID: PMC11002893 DOI: 10.1016/j.redox.2024.103149] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/02/2024] [Indexed: 04/08/2024] Open
Abstract
Macrophage cholesterol homeostasis is crucial for health and disease and has been linked to the lipid-peroxidizing enzyme arachidonate 15-lipoxygenase type B (ALOX15B), albeit molecular mechanisms remain obscure. We performed global transcriptome and immunofluorescence analysis in ALOX15B-silenced primary human macrophages and observed a reduction of nuclear sterol regulatory element-binding protein (SREBP) 2, the master transcription factor of cellular cholesterol biosynthesis. Consequently, SREBP2-target gene expression was reduced as were the sterol biosynthetic intermediates desmosterol and lathosterol as well as 25- and 27-hydroxycholesterol. Mechanistically, suppression of ALOX15B reduced lipid peroxidation in primary human macrophages and thereby attenuated activation of mitogen-activated protein kinase ERK1/2, which lowered SREBP2 abundance and activity. Low nuclear SREBP2 rendered both, ALOX15B-silenced and ERK1/2-inhibited macrophages refractory to SREBP2 activation upon blocking the NPC intracellular cholesterol transporter 1. These studies suggest a regulatory mechanism controlling macrophage cholesterol homeostasis based on ALOX15B-mediated lipid peroxidation and concomitant ERK1/2 activation.
Collapse
Affiliation(s)
- Yvonne Benatzy
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Megan A Palmer
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Dieter Lütjohann
- Institute for Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | - Rei-Ichi Ohno
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Nadja Kampschulte
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Nils Helge Schebb
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Dominik C Fuhrmann
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany.
| | - Ryan G Snodgrass
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany; Western Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Davis, CA, USA.
| | - Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt, Germany.
| |
Collapse
|
6
|
Wei G, Zhu H, Zhou Y, Pan Y, Yi B, Bai Y. Single-cell sequencing revealed metabolic reprogramming and its transcription factor regulatory network in prostate cancer. Transl Oncol 2024; 44:101925. [PMID: 38447277 PMCID: PMC11391037 DOI: 10.1016/j.tranon.2024.101925] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/19/2023] [Accepted: 02/28/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND/AIMS Prostate cancer is the most frequently diagnosed cancer among men in the United States and is the second leading cause of cancer-related deaths in men. The incidence of prostate cancer is gradually rising due to factors such as aging demographics and changes in dietary habits. The objective of this study is to investigate the metabolic reprogramming changes occurring in prostate cancer and identify potential therapeutic targets. METHODS In this study, we utilized single-cell sequencing to comprehensively characterize the alterations in metabolism and the regulatory role of transcription factors in various subtypes of prostate cancer. RESULTS In comparison to benign prostate tissue, prostate cancer displayed substantial metabolic variations, notably exhibiting heightened activity in fatty acid metabolism and cholesterol metabolism. This metabolic reprogramming not only influenced cellular energy utilization but also potentially impacted the activity of the androgen receptor (AR) pathway through the synthesis of endogenous steroid hormones. Through our analysis of transcription factor activity, we identified the crucial role of SREBPs, which are transcription factors associated with lipid metabolism, in prostate cancer. Encouragingly, the inhibitor Betulin effectively suppresses prostate cancer growth, highlighting its potential as a therapeutic agent for prostate cancer treatment.
Collapse
Affiliation(s)
- Guojiang Wei
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, PR China; Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, PR China.
| | - Hongcai Zhu
- Department of Medical Oncology, Hanzhong Central Hospital, Hanzhong, Shaanxi 723000, PR China
| | - Yupeng Zhou
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, PR China; Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, PR China
| | - Yang Pan
- Department of Urology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Bocun Yi
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, PR China; Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, PR China
| | - Yangkai Bai
- Department of Urology, Hanzhong Central Hospital, Hanzhong, Shaanxi 723000, PR China; Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, PR China.
| |
Collapse
|
7
|
Chandrasekaran P, Weiskirchen R. The Role of SCAP/SREBP as Central Regulators of Lipid Metabolism in Hepatic Steatosis. Int J Mol Sci 2024; 25:1109. [PMID: 38256181 PMCID: PMC10815951 DOI: 10.3390/ijms25021109] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/09/2024] [Accepted: 01/14/2024] [Indexed: 01/24/2024] Open
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is rapidly increasing worldwide at an alarming pace, due to an increase in obesity, sedentary and unhealthy lifestyles, and unbalanced dietary habits. MASLD is a unique, multi-factorial condition with several phases of progression including steatosis, steatohepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma. Sterol element binding protein 1c (SREBP1c) is the main transcription factor involved in regulating hepatic de novo lipogenesis. This transcription factor is synthesized as an inactive precursor, and its proteolytic maturation is initiated in the membrane of the endoplasmic reticulum upon stimulation by insulin. SREBP cleavage activating protein (SCAP) is required as a chaperon protein to escort SREBP from the endoplasmic reticulum and to facilitate the proteolytic release of the N-terminal domain of SREBP into the Golgi. SCAP inhibition prevents activation of SREBP and inhibits the expression of genes involved in triglyceride and fatty acid synthesis, resulting in the inhibition of de novo lipogenesis. In line, previous studies have shown that SCAP inhibition can resolve hepatic steatosis in animal models and intensive research is going on to understand the effects of SCAP in the pathogenesis of human disease. This review focuses on the versatile roles of SCAP/SREBP regulation in de novo lipogenesis and the structure and molecular features of SCAP/SREBP in the progression of hepatic steatosis. In addition, recent studies that attempt to target the SCAP/SREBP axis as a therapeutic option to interfere with MASLD are discussed.
Collapse
Affiliation(s)
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), Rheinisch-Westfälische Technische Hochschule (RWTH) University Hospital Aachen, D-52074 Aachen, Germany
| |
Collapse
|
8
|
Chen J, Chan TTH, Zhou J. Lipid metabolism in the immune niche of tumor-prone liver microenvironment. J Leukoc Biol 2024; 115:68-84. [PMID: 37474318 DOI: 10.1093/jleuko/qiad081] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/23/2023] [Accepted: 07/06/2023] [Indexed: 07/22/2023] Open
Abstract
The liver is a common primary site not only for tumorigenesis, but also for cancer metastasis. Advanced cancer patients with liver metastases also show reduced response rates and survival benefits when treated with immune checkpoint inhibitors. Accumulating evidence has highlighted the importance of the liver immune microenvironment in determining tumorigenesis, metastasis-organotropism, and immunotherapy resistance. Various immune cells such as T cells, natural killer and natural killer T cells, macrophages and dendritic cells, and stromal cells including liver sinusoidal endothelial cells, Kupffer cells, hepatic stellate cells, and hepatocytes are implicated in contributing to the immune niche of tumor-prone liver microenvironment. In parallel, as the major organ for lipid metabolism, the increased abundance of lipids and their metabolites is linked to processes crucial for nonalcoholic fatty liver disease and related liver cancer development. Furthermore, the proliferation, differentiation, and functions of hepatic immune and stromal cells are also reported to be regulated by lipid metabolism. Therefore, targeting lipid metabolism may hold great potential to reprogram the immunosuppressive liver microenvironment and synergistically enhance the immunotherapy efficacy in the circumstance of liver metastasis. In this review, we describe how the hepatic microenvironment adapts to the lipid metabolic alterations in pathologic conditions like nonalcoholic fatty liver disease. We also illustrate how these immunometabolic alterations promote the development of liver cancers and immunotherapy resistance. Finally, we discuss the current therapeutic options and hypothetic combination immunotherapies for the treatment of advanced liver cancers.
Collapse
Affiliation(s)
- Jintian Chen
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| | - Thomas T H Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| | - Jingying Zhou
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| |
Collapse
|
9
|
Liu Z, Zheng X, Chen J, Zheng L, Ma Z, Chen L, Deng M, Tang H, Zhou L, Kang T, Wu Y, Liu Z. NFYC-37 promotes tumor growth by activating the mevalonate pathway in bladder cancer. Cell Rep 2023; 42:112963. [PMID: 37561631 DOI: 10.1016/j.celrep.2023.112963] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/12/2023] [Accepted: 07/26/2023] [Indexed: 08/12/2023] Open
Abstract
Dysregulation of transcription is a hallmark of cancer, including bladder cancer (BLCA). CRISPR-Cas9 screening using a lentivirus library with single guide RNAs (sgRNAs) targeting human transcription factors and chromatin modifiers is used to reveal genes critical for the proliferation and survival of BLCA cells. As a result, the nuclear transcription factor Y subunit gamma (NFYC)-37, but not NFYC-50, is observed to promote cell proliferation and tumor growth in BLCA. Mechanistically, NFYC-37 interacts with CBP and SREBP2 to activate mevalonate pathway transcription, promoting cholesterol biosynthesis. However, NFYC-50 recruits more of the arginine methyltransferase CARM1 than NFYC-37 to methylate CBP, which prevents the CBP-SREBP2 interaction and subsequently inhibits the mevalonate pathway. Importantly, statins targeting the mevalonate pathway can suppress NFYC-37-induced cell proliferation and tumor growth, indicating the need for conducting a clinical trial with statins for treating patients with BLCA and high NFYC-37 levels, as most patients with BLCA have high NFYC-37 levels.
Collapse
Affiliation(s)
- Zefu Liu
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China; Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Xianchong Zheng
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China; Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Jiawei Chen
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China; Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Lisi Zheng
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Zikun Ma
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China; Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Lei Chen
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China; Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Minhua Deng
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China; Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Huancheng Tang
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China; Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Liwen Zhou
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Tiebang Kang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China.
| | - Yuanzhong Wu
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China.
| | - Zhuowei Liu
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China; Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China; Department of Urology, Sun Yat-sen University Cancer Center Gansu Hospital, Lanzhou 730000, Gansu, China.
| |
Collapse
|
10
|
Kennewick KT, Bensinger SJ. Decoding the crosstalk between mevalonate metabolism and T cell function. Immunol Rev 2023; 317:71-94. [PMID: 36999733 DOI: 10.1111/imr.13200] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/12/2023] [Accepted: 03/16/2023] [Indexed: 04/01/2023]
Abstract
The mevalonate pathway is an essential metabolic pathway in T cells regulating development, proliferation, survival, differentiation, and effector functions. The mevalonate pathway is a complex, branched pathway composed of many enzymes that ultimately generate cholesterol and nonsterol isoprenoids. T cells must tightly control metabolic flux through the branches of the mevalonate pathway to ensure sufficient isoprenoids and cholesterol are available to meet cellular demands. Unbalanced metabolite flux through the sterol or the nonsterol isoprenoid branch is metabolically inefficient and can have deleterious consequences for T cell fate and function. Accordingly, there is tight regulatory control over metabolic flux through the branches of this essential lipid synthetic pathway. In this review we provide an overview of how the branches of the mevalonate pathway are regulated in T cells and discuss our current understanding of the relationship between mevalonate metabolism, cholesterol homeostasis and T cell function.
Collapse
Affiliation(s)
- Kelly T Kennewick
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, USA
| | - Steven J Bensinger
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, USA
| |
Collapse
|
11
|
Maier CR, Hartmann O, Prieto-Garcia C, Al-Shami KM, Schlicker L, Vogel FCE, Haid S, Klann K, Buck V, Münch C, Schmitz W, Einig E, Krenz B, Calzado MA, Eilers M, Popov N, Rosenfeldt MT, Diefenbacher ME, Schulze A. USP28 controls SREBP2 and the mevalonate pathway to drive tumour growth in squamous cancer. Cell Death Differ 2023:10.1038/s41418-023-01173-6. [PMID: 37202505 DOI: 10.1038/s41418-023-01173-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/16/2023] [Accepted: 04/25/2023] [Indexed: 05/20/2023] Open
Abstract
SREBP2 is a master regulator of the mevalonate pathway (MVP), a biosynthetic process that drives the synthesis of dolichol, heme A, ubiquinone and cholesterol and also provides substrates for protein prenylation. Here, we identify SREBP2 as a novel substrate for USP28, a deubiquitinating enzyme that is frequently upregulated in squamous cancers. Our results show that silencing of USP28 reduces expression of MVP enzymes and lowers metabolic flux into this pathway. We also show that USP28 binds to mature SREBP2, leading to its deubiquitination and stabilisation. USP28 depletion rendered cancer cells highly sensitive to MVP inhibition by statins, which was rescued by the addition of geranyl-geranyl pyrophosphate. Analysis of human tissue microarrays revealed elevated expression of USP28, SREBP2 and MVP enzymes in lung squamous cell carcinoma (LSCC) compared to lung adenocarcinoma (LADC). Moreover, CRISPR/Cas-mediated deletion of SREBP2 selectively attenuated tumour growth in a KRas/p53/LKB1 mutant mouse model of lung cancer. Finally, we demonstrate that statins synergise with a dual USP28/25 inhibitor to reduce viability of SCC cells. Our findings suggest that combinatorial targeting of MVP and USP28 could be a therapeutic strategy for the treatment of squamous cell carcinomas.
Collapse
Affiliation(s)
- Carina R Maier
- German Cancer Research Center, Division of Tumor Metabolism and Microenvironment, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
- Protein Stability and Cancer Group, Department of Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074, Würzburg, Germany
| | - Oliver Hartmann
- Protein Stability and Cancer Group, Department of Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074, Würzburg, Germany
| | - Cristian Prieto-Garcia
- Protein Stability and Cancer Group, Department of Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074, Würzburg, Germany
- Institute of Biochemistry II, Goethe University Frankfurt, Theodor-Stern-Kai 7, Haus 75, 60590, Frankfurt am Main, Germany
| | - Kamal M Al-Shami
- German Cancer Research Center, Division of Tumor Metabolism and Microenvironment, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Lisa Schlicker
- German Cancer Research Center, Division of Tumor Metabolism and Microenvironment, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Felix C E Vogel
- German Cancer Research Center, Division of Tumor Metabolism and Microenvironment, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Silke Haid
- German Cancer Research Center, Division of Tumor Metabolism and Microenvironment, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Kevin Klann
- Institute of Biochemistry II, Goethe University Frankfurt, Theodor-Stern-Kai 7, Haus 75, 60590, Frankfurt am Main, Germany
| | - Viktoria Buck
- Institute of Pathology, Julius Maximilians University and Comprehensive Cancer Center (CCC) Mainfranken, Josef-Schneider-Strasse 2, 97080, Würzburg, Germany
| | - Christian Münch
- Institute of Biochemistry II, Goethe University Frankfurt, Theodor-Stern-Kai 7, Haus 75, 60590, Frankfurt am Main, Germany
| | - Werner Schmitz
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| | - Elias Einig
- Internal Medicine VIII-Clinical Tumor Biology, University of Tübingen, Otfried-Müller-Straße 14, 72076, Tübingen, Germany
| | - Bastian Krenz
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| | - Marco A Calzado
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
- Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Martin Eilers
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| | - Nikita Popov
- Internal Medicine VIII-Clinical Tumor Biology, University of Tübingen, Otfried-Müller-Straße 14, 72076, Tübingen, Germany
| | - Mathias T Rosenfeldt
- Institute of Pathology, Julius Maximilians University and Comprehensive Cancer Center (CCC) Mainfranken, Josef-Schneider-Strasse 2, 97080, Würzburg, Germany
| | - Markus E Diefenbacher
- Protein Stability and Cancer Group, Department of Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074, Würzburg, Germany.
| | - Almut Schulze
- German Cancer Research Center, Division of Tumor Metabolism and Microenvironment, Im Neuenheimer Feld 581, 69120, Heidelberg, Germany.
| |
Collapse
|
12
|
Li Y, Wu S, Zhao X, Hao S, Li F, Wang Y, Liu B, Zhang D, Wang Y, Zhou H. Key events in cancer: Dysregulation of SREBPs. Front Pharmacol 2023; 14:1130747. [PMID: 36969840 PMCID: PMC10030587 DOI: 10.3389/fphar.2023.1130747] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
Lipid metabolism reprogramming is an important hallmark of tumor progression. Cancer cells require high levels of lipid synthesis and uptake not only to support their continued replication, invasion, metastasis, and survival but also to participate in the formation of biological membranes and signaling molecules. Sterol regulatory element binding proteins (SREBPs) are core transcription factors that control lipid metabolism and the expression of important genes for lipid synthesis and uptake. A growing number of studies have shown that SREBPs are significantly upregulated in human cancers and serve as intermediaries providing a mechanistic link between lipid metabolism reprogramming and malignancy. Different subcellular localizations, including endoplasmic reticulum, Golgi, and nucleus, play an indispensable role in regulating the cleavage maturation and activity of SREBPs. In this review, we focus on the relationship between aberrant regulation of SREBPs activity in three organelles and tumor progression. Because blocking the regulation of lipid synthesis by SREBPs has gradually become an important part of tumor therapy, this review also summarizes and analyzes several current mainstream strategies.
Collapse
Affiliation(s)
- Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Shouwang Wu
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Xiaodong Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Shiming Hao
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Yuxiong Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Difei Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
- *Correspondence: Yishu Wang, Honglan Zhou,
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Yishu Wang, Honglan Zhou,
| |
Collapse
|
13
|
Du Y, Xi M, Li Y, Zheng R, Ding X, Li X, Zhang X, Wang L, Xing J, Hong B. Tilianin improves lipid profile and alleviates atherosclerosis in ApoE -/- mice through up-regulation of SREBP2-mediated LDLR expression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154577. [PMID: 36610166 DOI: 10.1016/j.phymed.2022.154577] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 09/15/2022] [Accepted: 11/22/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND The huge global burden of atherosclerotic cardiovascular diseases (CVDs) represents an urgent unmet need for the development of novel therapeutics. Dracocephalum moldavica L. has been used as a traditional Uygur medicine to treat various CVDs for centuries. Tilianin is a major flavonoid component of D. moldavica L. and has potential for preventing atherosclerosis. However, the molecular mechanisms that tilianin attenuate atherosclerosis are far from fully understood. PURPOSES The purpose of this study is to investigate the efficiency and underlying mechanisms of tilianin in controlling lipid profile and preventing atherogenesis. METHODS The lipid-lowering effect of tilianin was evaluated in C57BL/6 and ApoE-/- mice by systematically determining serum biochemical parameters. The effects of tilianin on the atherosclerotic lesion were observed in aortic roots and whole aortas of ApoE-/- mice with oil red O staining. Caecal content from ApoE-/- mice were collected for 16S rRNA gene sequence analysis to assess the structure of the gut microbiota. The inhibition of hepatosteatosis was verified by histological examination, and a liver transcriptome analysis was performed to elucidate the tilianin-induced hepatic transcriptional alterations. Effects of tilianin on the expression and function of LDLR were examined in HepG2 cells and ApoE-/- mice. Further mechanisms underlying the efficacy of tilianin were investigated in HepG2 cells. RESULTS Tilianin treatment improved lipid profiles in C57BL/6 and dyslipidemic ApoE-/- mice, especially reducing the serum LDL-cholesterol (LDL-C) level. Significant reductions of atherosclerotic lesion area and hepatosteatosis were observed in tilianin-treated ApoE-/- mice. The altered gut microbial composition in tilianin groups was associated with lipid metabolism and atherosclerosis. The liver transcriptome revealed that tilianin regulated the transcription of lipid metabolism-related genes. Then both in vitro and in vivo analyses revealed the potent effect of tilianin to enhance hepatic LDLR expression and its mediated LDL-C uptake. Further studies confirmed a critical role of SREBP2 in hepatic LDLR up-regulation by tilianin via increasing precursor and thus mature nuclear SREBP2 level. CONCLUSION This study demonstrated the lipid-lowering effect of tilianin through SREBP2-mediated transcriptional activation of LDLR. Our findings reveal a novel anti-atherosclerotic mechanism of tilianin and underlie its potential clinical use in modulating CVDs with good availability and affordability.
Collapse
Affiliation(s)
- Yu Du
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Mei Xi
- Xinjiang Key Laboratory of Uighur Medicines, Xinjiang Institute of Materia Medica, Urumchi 830004, China
| | - Yihua Li
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ruifang Zheng
- Xinjiang Key Laboratory of Uighur Medicines, Xinjiang Institute of Materia Medica, Urumchi 830004, China
| | - Xiaotian Ding
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xingxing Li
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiumin Zhang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Li Wang
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jianguo Xing
- Xinjiang Key Laboratory of Uighur Medicines, Xinjiang Institute of Materia Medica, Urumchi 830004, China.
| | - Bin Hong
- NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
14
|
Chemical chaperones ameliorate neurodegenerative disorders in Derlin-1-deficient mice via improvement of cholesterol biosynthesis. Sci Rep 2022; 12:21840. [PMID: 36528738 PMCID: PMC9759528 DOI: 10.1038/s41598-022-26370-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
There are no available therapies targeting the underlying molecular mechanisms of neurodegenerative diseases. Although chaperone therapies that alleviate endoplasmic reticulum (ER) stress recently showed promise in the treatment of neurodegenerative diseases, the detailed mechanisms remain unclear. We previously reported that mice with central nervous system-specific deletion of Derlin-1, which encodes an essential component for ER quality control, are useful as models of neurodegenerative diseases such as spinocerebellar degeneration. Cholesterol biosynthesis is essential for brain development, and its disruption inhibits neurite outgrowth, causing brain atrophy. In this study, we report a novel mechanism by which chemical chaperones ameliorate brain atrophy and motor dysfunction. ER stress was induced in the cerebella of Derlin-1 deficiency mice, whereas the administration of a chemical chaperone did not alleviate ER stress. However, chemical chaperone treatment ameliorated cholesterol biosynthesis impairment through SREBP-2 activation and simultaneously relieved brain atrophy and motor dysfunction. Altogether, these findings demonstrate that ER stress may not be the target of action of chaperone therapies and that chemical chaperone-mediated improvement of brain cholesterol biosynthesis is a promising novel therapeutic strategy for neurodegenerative diseases.
Collapse
|
15
|
Jiang SY, Yang X, Yang Z, Li JW, Xu MQ, Qu YX, Tang JJ, Li YF, Wang L, Shao YW, Meng XY, Hu H, Song BL, Rao Y, Qi W. Discovery of an insulin-induced gene binding compound that ameliorates nonalcoholic steatohepatitis by inhibiting sterol regulatory element-binding protein-mediated lipogenesis. Hepatology 2022; 76:1466-1481. [PMID: 35102596 DOI: 10.1002/hep.32381] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/14/2022] [Accepted: 01/27/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND AIMS NASH is associated with high levels of cholesterol and triglyceride (TG) in the liver; however, there is still no approved pharmacological therapy. Synthesis of cholesterol and TG is controlled by sterol regulatory element-binding protein (SREBP), which is found to be abnormally activated in NASH patients. We aim to discover small molecules for treating NASH by inhibiting the SREBP pathway. APPROACH AND RESULTS Here, we identify a potent SREBP inhibitor, 25-hydroxylanosterol (25-HL). 25-HL binds to insulin-induced gene (INSIG) proteins, stimulates the interaction between INSIG and SCAP, and retains them in the endoplasmic reticulum, thereby suppressing SREBP activation and inhibiting lipogenesis. In NASH mouse models, 25-HL lowers levels of cholesterol and TG in serum and the liver, enhances energy expenditure to prevent obesity, and improves insulin sensitivity. 25-HL dramatically ameliorates hepatic steatosis, inflammation, ballooning, and fibrosis through down-regulating the expression of lipogenic genes. Furthermore, 25-HL exhibits both prophylactic and therapeutic efficacies of alleviating NASH and atherosclerosis in amylin liver NASH model diet-treated Ldlr-/- mice, and reduces the formation of cholesterol crystals and associated crown-like structures of Kupffer cells. Notably, 25-HL lowers lipid contents in serum and the liver to a greater extent than lovastatin or obeticholic acid. 25-HL shows a good safety and pharmacokinetics profile. CONCLUSIONS This study provides the proof of concept that inhibiting SREBP activation by targeting INSIG to lower lipids could be a promising strategy for treating NASH. It suggests the translational potential of 25-HL in human NASH and demonstrates the critical role of SREBP-controlled lipogenesis in the progression of NASH by pharmacological inhibition.
Collapse
Affiliation(s)
- Shi-You Jiang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China.,Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xinglin Yang
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, China
| | - Zimo Yang
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, China
| | - Jue-Wan Li
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Meng-Qiang Xu
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yu-Xiu Qu
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jing-Jie Tang
- The State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yun-Feng Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Liguo Wang
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, China
| | - Yi-Wen Shao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, Shandong University Cheeloo Medical College, School of Basic Medical Sciences, Jinan, China.,The Research Center of Stem Cell and Regenerative Medicine, Shandong University Cheeloo Medical College, School of Basic Medical Sciences, Jinan, China
| | - Xin-Yuan Meng
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, Shandong University Cheeloo Medical College, School of Basic Medical Sciences, Jinan, China.,The Research Center of Stem Cell and Regenerative Medicine, Shandong University Cheeloo Medical College, School of Basic Medical Sciences, Jinan, China
| | - Huili Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Molecular Medicine and Genetics, Shandong University Cheeloo Medical College, School of Basic Medical Sciences, Jinan, China.,The Research Center of Stem Cell and Regenerative Medicine, Shandong University Cheeloo Medical College, School of Basic Medical Sciences, Jinan, China
| | - Bao-Liang Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 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, China
| | - Wei Qi
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
16
|
Zhu C, Dong X, Wang X, Zheng Y, Qiu J, Peng Y, Xu J, Chai Z, Liu C. Multiple Roles of SIRT2 in Regulating Physiological and Pathological Signal Transduction. Genet Res (Camb) 2022; 2022:9282484. [PMID: 36101744 PMCID: PMC9444453 DOI: 10.1155/2022/9282484] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 07/01/2022] [Accepted: 07/08/2022] [Indexed: 11/18/2022] Open
Abstract
Sirtuin 2 (SIRT2), as a member of the sirtuin family, has representative features of evolutionarily highly conserved nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase activity. In addition, SIRT2, as the only sirtuin protein colocalized with tubulin in the cytoplasm, has its own functions and characteristics. In recent years, studies have increasingly shown that SIRT2 can participate in the regulation of gene expression and regulate signal transduction in the metabolic pathway mainly through its post-translational modification of target genes; thus, SIRT2 has become a key centre in the metabolic pathway and participates in the pathological process of metabolic disorder-related diseases. In this paper, it is discussed that SIRT2 can regulate all aspects of gene expression, including epigenetic modification, replication, transcription and translation, and post-translational modification, which enables SIRT2 to participate in energy metabolism in life activities, and it is clarified that SIRT2 is involved in metabolic process-specific signal transduction mechanisms. Therefore, SIRT2 can be involved in metabolic disorder-related inflammation and oxidative stress, thereby triggering the occurrence of metabolic disorder-related diseases, such as neurodegenerative diseases, tumours, diabetes, and cardiovascular diseases. Currently, although the role of SIRT2 in some diseases is still controversial, given the multiple roles of SIRT2 in regulating physiological and pathological signal transduction, SIRT2 has become a key target for disease treatment. It is believed that with increasing research, the clinical application of SIRT2 will be promoted.
Collapse
Affiliation(s)
- Changhui Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, Shandong, China
- Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Xue Dong
- Department of Education, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Xiwei Wang
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Yingying Zheng
- Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Juanjuan Qiu
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250014, China
| | - Yanling Peng
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250014, China
| | - Jiajun Xu
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250014, China
| | - Zhengbin Chai
- Department of Clinical Laboratory Medicine, Shandong Public Health Clinical Center, Shandong University, Jinan 250102, China
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Chunyan Liu
- Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| |
Collapse
|
17
|
Miyata S, Kodaka M, Kikuchi A, Matsunaga Y, Shoji K, Kuan YC, Iwase M, Takeda K, Katsuta R, Ishigami K, Matsumoto Y, Suzuki T, Yamamoto Y, Sato R, Inoue J. Sulforaphane suppresses the activity of sterol regulatory element-binding proteins (SREBPs) by promoting SREBP precursor degradation. Sci Rep 2022; 12:8715. [PMID: 35610278 PMCID: PMC9130306 DOI: 10.1038/s41598-022-12347-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 05/09/2022] [Indexed: 12/26/2022] Open
Abstract
Sterol regulatory element-binding proteins (SREBPs) are transcription factors that regulate various genes involved in cholesterol and fatty acid synthesis. In this study, we describe that naturally occurring isothiocyanate sulforaphane (SFaN) impairs fatty acid synthase promoter activity and reduces SREBP target gene (e.g., fatty acid synthase and acetyl-CoA carboxylase 1) expression in human hepatoma Huh-7 cells. SFaN reduced SREBP proteins by promoting the degradation of the SREBP precursor. Amino acids 595–784 of SREBP-1a were essential for SFaN-mediated SREBP-1a degradation. We also found that such SREBP-1 degradation occurs independently of the SREBP cleavage-activating protein and the Keap1-Nrf2 pathway. This study identifies SFaN as an SREBP inhibitor and provides evidence that SFaN could have major potential as a pharmaceutical preparation against hepatic steatosis and obesity.
Collapse
Affiliation(s)
- Shingo Miyata
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Manami Kodaka
- Department of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Akito Kikuchi
- Department of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Yuki Matsunaga
- Department of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Kenta Shoji
- Department of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Yen-Chou Kuan
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan.,Department of Horticulture and Landscape Architecture, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Masamori Iwase
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Keita Takeda
- Department of Chemistry for Life Sciences and Agriculture, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Ryo Katsuta
- Department of Chemistry for Life Sciences and Agriculture, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Ken Ishigami
- Department of Chemistry for Life Sciences and Agriculture, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Yu Matsumoto
- Department of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Tsukasa Suzuki
- Department of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Yuji Yamamoto
- Department of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Ryuichiro Sato
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan.
| | - Jun Inoue
- Department of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, 156-8502, Japan.
| |
Collapse
|
18
|
Ahmed N, Ahmed N, Pezacki JP. miR-383 Regulates Hepatic Lipid Homeostasis and Response to Dengue Virus Infection. ACS Infect Dis 2022; 8:928-941. [PMID: 35254825 DOI: 10.1021/acsinfecdis.1c00470] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Recently, microRNAs (miRNAs), as endogenous noncoding RNAs that inhibit mRNA translation, have been identified to broadly possess functional roles in regulating cellular signaling and metabolic processes due to their chemical and biological properties. In addition, they have emerged to be of critical importance in modulating host-virus interactions, especially for RNA viruses. Herein, we discovered that miR-383-5p targets certain lipid and cholesterol biosynthetic pathways and restricts Dengue virus (DENV) infection in hepatic cells. Global transcriptomics analysis of Huh7 human hepatoma cells overexpressing miR-383-5p revealed enrichment of lipid and cholesterol metabolic processes. Bioinformatics analysis of genes repressed in miR-383-5p overexpressing cells divulged the repression of a key target PLA2G4A, a pro-viral host factor essential for the production of infectious DENV particles. Our study demonstrated the effectiveness of miRNA mimics as tools to study cellular signaling pathways that contribute to viral pathogenesis. Overall, our study identifies miR-383-5p as an interesting host factor during DENV propagation and highlights a potential therapeutic role in the regulation of hepatic lipid metabolism and an antiviral response to DENV.
Collapse
Affiliation(s)
- Nadine Ahmed
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Noreen Ahmed
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - John Paul Pezacki
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| |
Collapse
|
19
|
Shi Q, Chen J, Zou X, Tang X. Intracellular Cholesterol Synthesis and Transport. Front Cell Dev Biol 2022; 10:819281. [PMID: 35386193 PMCID: PMC8978673 DOI: 10.3389/fcell.2022.819281] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 02/01/2022] [Indexed: 12/18/2022] Open
Abstract
Cholesterol homeostasis is related to multiple diseases in humans, including cardiovascular disease, cancer, and neurodegenerative and hepatic diseases. The cholesterol levels in cells are balanced dynamically by uptake, biosynthesis, transport, distribution, esterification, and export. In this review, we focus on de novo cholesterol synthesis, cholesterol synthesis regulation, and intracellular cholesterol trafficking. In addition, the progression of lipid transfer proteins (LTPs) at multiple contact sites between organelles is considered.
Collapse
Affiliation(s)
- Qingyang Shi
- Center of Reproductive Medicine and Center of Prenatal Diagnosis, The First Hospital, Jilin University, Changchun, China
| | - Jiahuan Chen
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, Changchun, China
| | - Xiaodong Zou
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, Changchun, China
| | - Xiaochun Tang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, Changchun, China
- Chongqing Research Institute of Jilin University, Chongqing, China
| |
Collapse
|
20
|
Wall CTJ, Lefebvre G, Metairon S, Descombes P, Wiederkehr A, Santo-Domingo J. Mitochondrial respiratory chain dysfunction alters ER sterol sensing and mevalonate pathway activity. J Biol Chem 2022; 298:101652. [PMID: 35101444 PMCID: PMC8892029 DOI: 10.1016/j.jbc.2022.101652] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/13/2022] [Accepted: 01/25/2022] [Indexed: 12/13/2022] Open
Abstract
Mitochondrial dysfunction induces a strong adaptive retrograde signaling response; however, many of the downstream effectors of this response remain to be discovered. Here, we studied the shared transcriptional responses to three different mitochondrial respiratory chain inhibitors in human primary skin fibroblasts using QuantSeq 3′-RNA-sequencing. We found that genes involved in the mevalonate pathway were concurrently downregulated, irrespective of the respiratory chain complex affected. Targeted metabolomics demonstrated that impaired mitochondrial respiration at any of the three affected complexes also had functional consequences on the mevalonate pathway, reducing levels of cholesterol precursor metabolites. A deeper study of complex I inhibition showed a reduced activity of endoplasmic reticulum–bound sterol-sensing enzymes through impaired processing of the transcription factor Sterol Regulatory Element-Binding Protein 2 and accelerated degradation of the endoplasmic reticulum cholesterol-sensors squalene epoxidase and HMG-CoA reductase. These adaptations of mevalonate pathway activity affected neither total intracellular cholesterol levels nor the cellular free (nonesterified) cholesterol pool. Finally, measurement of intracellular cholesterol using the fluorescent cholesterol binding dye filipin revealed that complex I inhibition elevated cholesterol on intracellular compartments. Taken together, our study shows that mitochondrial respiratory chain dysfunction elevates intracellular free cholesterol levels and therefore attenuates the expression of mevalonate pathway enzymes, which lowers endogenous cholesterol biosynthesis, disrupting the metabolic output of the mevalonate pathway. We conclude that intracellular disturbances in cholesterol homeostasis may alter systemic cholesterol management in diseases associated with declining mitochondrial function.
Collapse
Affiliation(s)
- Christopher Tadhg James Wall
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland; Institute of Bioengineering, Life Science Faculty, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Gregory Lefebvre
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Sylviane Metairon
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Patrick Descombes
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Andreas Wiederkehr
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland; Institute of Bioengineering, Life Science Faculty, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | - Jaime Santo-Domingo
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland; Department of Biochemistry and Molecular Biology, Faculty of Medicine, Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), University of Valladolid and CSIC, Valladolid, Spain.
| |
Collapse
|
21
|
Trindade BC, Ceglia S, Berthelette A, Raso F, Howley K, Muppidi JR, Reboldi A. The cholesterol metabolite 25-hydroxycholesterol restrains the transcriptional regulator SREBP2 and limits intestinal IgA plasma cell differentiation. Immunity 2021; 54:2273-2287.e6. [PMID: 34644558 DOI: 10.1016/j.immuni.2021.09.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/10/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022]
Abstract
Diets high in cholesterol alter intestinal immunity. Here, we examined how the cholesterol metabolite 25-hydroxycholesterol (25-HC) impacts the intestinal B cell response. Mice lacking cholesterol 25-hydroxylase (CH25H), the enzyme generating 25-HC, had higher frequencies of immunoglobulin A (IgA)-secreting antigen-specific B cells upon immunization or infection. 25-HC did not affect class-switch recombination but rather restrained plasma cell (PC) differentiation. 25-HC was produced by follicular dendritic cells and increased in response to dietary cholesterol. Mechanistically, 25-HC restricted activation of the sterol-sensing transcription factor SREBP2, thereby regulating B cell cholesterol biosynthesis. Ectopic expression of SREBP2 in germinal center B cells induced rapid PC differentiation, whereas SREBP2 deficiency reduced PC output in vitro and in vivo. High-cholesterol diet impaired, whereas Ch25h deficiency enhanced, the IgA response against Salmonella and the resulting protection from systemic bacterial dissemination. Thus, a 25-HC-SREBP2 axis shapes the humoral response at the intestinal barrier, providing insight into the effect of high dietary cholesterol in intestinal immunity.
Collapse
Affiliation(s)
- Bruno C Trindade
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Simona Ceglia
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Alyssa Berthelette
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Fiona Raso
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kelsey Howley
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jagan R Muppidi
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrea Reboldi
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
22
|
Giacomini I, Gianfanti F, Desbats MA, Orso G, Berretta M, Prayer-Galetti T, Ragazzi E, Cocetta V. Cholesterol Metabolic Reprogramming in Cancer and Its Pharmacological Modulation as Therapeutic Strategy. Front Oncol 2021; 11:682911. [PMID: 34109128 PMCID: PMC8181394 DOI: 10.3389/fonc.2021.682911] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/06/2021] [Indexed: 12/14/2022] Open
Abstract
Cholesterol is a ubiquitous sterol with many biological functions, which are crucial for proper cellular signaling and physiology. Indeed, cholesterol is essential in maintaining membrane physical properties, while its metabolism is involved in bile acid production and steroid hormone biosynthesis. Additionally, isoprenoids metabolites of the mevalonate pathway support protein-prenylation and dolichol, ubiquinone and the heme a biosynthesis. Cancer cells rely on cholesterol to satisfy their increased nutrient demands and to support their uncontrolled growth, thus promoting tumor development and progression. Indeed, transformed cells reprogram cholesterol metabolism either by increasing its uptake and de novo biosynthesis, or deregulating the efflux. Alternatively, tumor can efficiently accumulate cholesterol into lipid droplets and deeply modify the activity of key cholesterol homeostasis regulators. In light of these considerations, altered pathways of cholesterol metabolism might represent intriguing pharmacological targets for the development of exploitable strategies in the context of cancer therapy. Thus, this work aims to discuss the emerging evidence of in vitro and in vivo studies, as well as clinical trials, on the role of cholesterol pathways in the treatment of cancer, starting from already available cholesterol-lowering drugs (statins or fibrates), and moving towards novel potential pharmacological inhibitors or selective target modulators.
Collapse
Affiliation(s)
- Isabella Giacomini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Federico Gianfanti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, VIMM, Padova, Italy
| | | | - Genny Orso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Tommaso Prayer-Galetti
- Department of Surgery, Oncology and Gastroenterology - Urology, University of Padova, Padova, Italy
| | - Eugenio Ragazzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Veronica Cocetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
23
|
Kong M, Zhu Y, Shao J, Fan Z, Xu Y. The Chromatin Remodeling Protein BRG1 Regulates SREBP Maturation by Activating SCAP Transcription in Hepatocytes. Front Cell Dev Biol 2021; 9:622866. [PMID: 33718362 PMCID: PMC7947303 DOI: 10.3389/fcell.2021.622866] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
Sterol response element binding protein (SREBP) is a master regulator of cellular lipogenesis. One key step in the regulation of SREBP activity is its sequential cleavage and trans-location by several different proteinases including SREBP cleavage activating protein (SCAP). We have previously reported that Brahma related gene 1 (BRG1) directly interacts with SREBP1c and SREBP2 to activate pro-lipogenic transcription in hepatocytes. We report here that BRG1 deficiency resulted in reduced processing and nuclear accumulation of SREBP in the murine livers in two different models of non-alcoholic steatohepatitis (NASH). Exposure of hepatocytes to lipopolysaccharide (LPS) and palmitate (PA) promoted SREBP accumulation in the nucleus whereas BRG1 knockdown or inhibition blocked SREBP maturation. Further analysis revealed that BRG1 played an essential role in the regulation of SCAP expression. Mechanistically, BRG1 interacted with Sp1 and directly bound to the SCAP promoter to activate SCAP transcription. Forced expression of exogenous SCAP partially rescued the deficiency in the expression of SREBP target genes in BRG1-null hepatocytes. In conclusion, our data uncover a novel mechanism by which BRG1 contributes to SREBP-dependent lipid metabolism.
Collapse
Affiliation(s)
- Ming Kong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yuwen Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Jing Shao
- Wu Medical School, Jiangnan University, Wuxi, China
| | - Zhiwen Fan
- Department of Pathology, Affiliated Nanjing Drum Tower Hospital of Nanjing University School of Medicine, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| |
Collapse
|
24
|
Horie T, Nakao T, Miyasaka Y, Nishino T, Matsumura S, Nakazeki F, Ide Y, Kimura M, Tsuji S, Rodriguez RR, Watanabe T, Yamasaki T, Xu S, Otani C, Miyagawa S, Matsushita K, Sowa N, Omori A, Tanaka J, Nishimura C, Nishiga M, Kuwabara Y, Baba O, Watanabe S, Nishi H, Nakashima Y, Picciotto MR, Inoue H, Watanabe D, Nakamura K, Sasaki T, Kimura T, Ono K. microRNA-33 maintains adaptive thermogenesis via enhanced sympathetic nerve activity. Nat Commun 2021; 12:843. [PMID: 33594062 PMCID: PMC7886914 DOI: 10.1038/s41467-021-21107-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 01/12/2021] [Indexed: 12/30/2022] Open
Abstract
Adaptive thermogenesis is essential for survival, and therefore is tightly regulated by a central neural circuit. Here, we show that microRNA (miR)-33 in the brain is indispensable for adaptive thermogenesis. Cold stress increases miR-33 levels in the hypothalamus and miR-33−/− mice are unable to maintain body temperature in cold environments due to reduced sympathetic nerve activity and impaired brown adipose tissue (BAT) thermogenesis. Analysis of miR-33f/f dopamine-β-hydroxylase (DBH)-Cre mice indicates the importance of miR-33 in Dbh-positive cells. Mechanistically, miR-33 deficiency upregulates gamma-aminobutyric acid (GABA)A receptor subunit genes such as Gabrb2 and Gabra4. Knock-down of these genes in Dbh-positive neurons rescues the impaired cold-induced thermogenesis in miR-33f/fDBH-Cre mice. Conversely, increased gene dosage of miR-33 in mice enhances thermogenesis. Thus, miR-33 in the brain contributes to maintenance of BAT thermogenesis and whole-body metabolism via enhanced sympathetic nerve tone through suppressing GABAergic inhibitory neurotransmission. This miR-33-mediated neural mechanism may serve as a physiological adaptive defense mechanism for several stresses including cold stress. Adaptive thermogenesis is regulated by central neuronal circuits. Here, the authors show that microRNA-33 in the brain contributes to the maintenance of brown adipose tissue thermogenesis and whole-body energy balance via enhanced sympathetic nerve tone, and regulating the expression of GABAa receptor subunits.
Collapse
Affiliation(s)
- Takahiro Horie
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Tetsushi Nakao
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yui Miyasaka
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Nishino
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigenobu Matsumura
- Laboratory of Physiological Functions of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Fumiko Nakazeki
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuya Ide
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shuhei Tsuji
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Randolph Ruiz Rodriguez
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshimitsu Watanabe
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Yamasaki
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sijia Xu
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Chiharu Otani
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sawa Miyagawa
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuki Matsushita
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naoya Sowa
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Aoi Omori
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Jin Tanaka
- Laboratory of Physiological Functions of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Chika Nishimura
- Department of Biological Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masataka Nishiga
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuhide Kuwabara
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Osamu Baba
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shin Watanabe
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hitoo Nishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuhiro Nakashima
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Marina R Picciotto
- Department of Psychiatry and Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan.,Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Dai Watanabe
- Department of Biological Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuhiro Nakamura
- Department of Integrative Physiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tsutomu Sasaki
- Laboratory of Nutrition Chemistry, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koh Ono
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
25
|
Song S, Niu M, Liang Q, Wang L, Min H, Peng Y, Wang H, Gao Q. Statin Treatment Induced a Lipogenic Expression Hierarchical Network Centered by SREBF2 in the Liver. Front Endocrinol (Lausanne) 2021; 12:573824. [PMID: 34349727 PMCID: PMC8326809 DOI: 10.3389/fendo.2021.573824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
Statin treatment is a major prevention treatment for hypercholesterolemia and the management of patients with increased risk of cardiovascular disease (CVD) due to its protective effects. However, its long-term safety was questioned regarding its potential role in new-onset type 2 diabetes mellitus, and its effect on gene regulation in the liver is not yet fully understood. By reanalyzing the transcriptome of the livers of patients with obesity and hypercholesterolemia, this study shows a multiple module organization that is related to various clinical metabolic parameters and identified an expression hierarchical network involving cholesterol and fatty acid syntheses in the liver of statin-treated patients. The key genes of the network were validated by QPCR in the hepatocytes upon statin treatment. The upregulation of the key enzymes involving the synthesis of Acetyl-CoA and the induction of gentle global acetylation of pan-protein and histone H4 in hepatocytes were observed. The study provides an overall view of the statin effect on transcriptional and post-transcriptional regulation of genes in the liver.
Collapse
Affiliation(s)
- Shiyu Song
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Mengyuan Niu
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Qiao Liang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Lei Wang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Haiyan Min
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yuming Peng
- Department of General Practice of Central Hospital of Karamay, Karamay, China
| | - Hongwei Wang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, China
- *Correspondence: Qian Gao, ; Hongwei Wang,
| | - Qian Gao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- *Correspondence: Qian Gao, ; Hongwei Wang,
| |
Collapse
|
26
|
Li Y, Wu S. Curcumin inhibits the proteolytic process of SREBP-2 by first inhibiting the expression of S1P rather than directly inhibiting SREBP-2 expression. Food Sci Nutr 2021; 9:209-216. [PMID: 33473285 PMCID: PMC7802558 DOI: 10.1002/fsn3.1985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022] Open
Abstract
Many studies have demonstrated that curcumin can downregulate mRNA levels of sterol regulatory element-binding proteins (SREBP-2); however, our study did not find similar results. This study was designed to demonstrate that curcumin inhibits the proteolytic process of SREBP-2 by first inhibiting the expression of membrane-bound transcription factor site-1 protease (S1P) rather than directly inhibiting SREBP-2 expression. After curcumin treatment, Caco-2 cells were collected to observe the dose- and time-dependent dynamics of precursor and mature SREBP-2, transcription factor-specific protein 1 (SP-1), and SREBP cleavage-activating protein (SCAP). After curcumin treatment, SREBP-2 distribution was detected in the cells and S1P protein expression was examined. Curcumin could downregulate mRNA levels of SREBP2, SP-1 and SCAP, but it did not simultaneously downregulate the expression of precursor SREBP-2 (pSREBP-2) and SCAP. Curcumin can inhibit the proteolytic process of SREBP-2, reduce the production of mature SREBP-2 (mSREBP-2), and change the cellular distribution of SREBP-2. The inhibitory effect of curcumin on SP-1 protein expression is short-acting. Curcumin could downregulate the mRNA and protein expression of S1P, but has no obvious inhibitory effect on the mRNA and protein expression of S2P (site-2 protease). Curcumin can inhibit the SREBP-2 proteolytic process to reduce mSREBP-2 which functions as a transcription factor, affecting the regulation of cholesterol metabolism-related genes. Curcumin does not directly inhibit the expression of mSREBP-2 protein, and it has no such inhibitory effect for at least a short period of time, although curcumin does reduce the amount of mSREBP-2 protein. S1P is a key protease in the hydrolysis of pSREBP-2 into mSREBP-2. Therefore, curcumin may decrease the amount of mSREBP-2 by directly inhibiting the expression of S1P mRNA and protein.
Collapse
Affiliation(s)
- Yongnan Li
- The Sixth General SurgeryBiliary & Vascular surgeryShengjing Hospital of China Medical UniversityShenyang CityChina
| | - Shuodong Wu
- The Sixth General SurgeryBiliary & Vascular surgeryShengjing Hospital of China Medical UniversityShenyang CityChina
| |
Collapse
|
27
|
Chae HS, Kim HJ, Ko HJ, Lee CH, Choi YH, Chin YW. Transcriptome Analysis Illuminates a Hub Role of SREBP2 in Cholesterol Metabolism by α-Mangostin. ACS OMEGA 2020; 5:31126-31136. [PMID: 33324821 PMCID: PMC7726933 DOI: 10.1021/acsomega.0c04282] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/09/2020] [Indexed: 05/08/2023]
Abstract
Whole-transcriptome analysis of α-mangostin-treated HepG2 cells revealed that genes relevant to lipid and cholesterol metabolic processes responded to α-mangostin treatment. α-Mangostin downregulated a series of cholesterol biosynthetic genes, including SQLE, HMGCR, and LSS, and controlled specific cholesterol trafficking-associated genes such as ABCA1, SOAT1, and PCSK9. In particular, the downregulation of SREBP2 expression highlighted SREBP2 as a key transcriptional factor controlling lipid or cholesterol metabolic processes. Gene network analysis of SREBP2 and responses of its target proteins demonstrated that the effect of α-mangostin on HepG2 cells was mediated by the downregulation of SREBP2 expression, which was further supported by the reduction of the amount of SREBP2-SCAP complex. In the presence of exogenous cholesterols, α-mangostin downregulated SREBP2 expression and suppressed PCSK9 synthesis, which might contribute to the increased cholesterol uptake in cells, in part explaining the cholesterol-lowering effect of α-mangostin.
Collapse
Affiliation(s)
- Hee-Sung Chae
- College
of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University Seoul, 32 Dongguk-lo, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Republic of Korea
| | - Hyun Ji Kim
- College
of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University Seoul, 32 Dongguk-lo, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Republic of Korea
| | - Hyun-Jeong Ko
- Laboratory
of Microbiology and Immunology, College of Pharmacy, Kangwon National University, 1 Gangwondaehakgil, Chuncheon-si, Gangwon-do 24341, Republic of Korea
| | - Chang Hoon Lee
- College
of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University Seoul, 32 Dongguk-lo, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Republic of Korea
| | - Young Hee Choi
- College
of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University Seoul, 32 Dongguk-lo, Ilsandong-gu, Goyang-si, Gyeonggi-do 10326, Republic of Korea
| | - Young-Won Chin
- College
of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-lo, Gwanak-gu, Seoul 08826, Republic
of Korea
- E-mail: . Phone: +82 2 880 7859
| |
Collapse
|
28
|
Jiang T, Zhang G, Lou Z. Role of the Sterol Regulatory Element Binding Protein Pathway in Tumorigenesis. Front Oncol 2020; 10:1788. [PMID: 33014877 PMCID: PMC7506081 DOI: 10.3389/fonc.2020.01788] [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: 08/24/2019] [Accepted: 08/11/2020] [Indexed: 12/15/2022] Open
Abstract
Metabolic changes are a major feature of tumors, including various metabolic forms, such as energy, lipid, and amino acid metabolism. Sterol regulatory element binding proteins (SREBPs) are important modules in regulating lipid metabolism and play an essential role in metabolic diseases. In the previous decades, the regulatory range of SREBPs has been markedly expanded. It was found that SREBPs also played a critical role in tumor development. SREBPs are involved in energy supply, lipid supply, immune environment and inflammatory environment shaping in tumor cells, and as a protective umbrella to support the malignant proliferation of tumor cells. Natural medicine and traditional Chinese medicine, as an important part of drug therapy, demonstrates the multifaceted effects of SREBPs regulation. This review summarizes the core processes in the involvement of SREBPs in tumors and provides a comprehensive understanding of the pathways through which natural drugs target the SREBP pathway and regulate tumor progression.
Collapse
Affiliation(s)
- Tao Jiang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Guangji Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhaohuan Lou
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
29
|
Oni TE, Biffi G, Baker LA, Hao Y, Tonelli C, Somerville TD, Deschênes A, Belleau P, Hwang CI, Sánchez-Rivera FJ, Cox H, Brosnan E, Doshi A, Lumia RP, Khaledi K, Park Y, Trotman LC, Lowe SW, Krasnitz A, Vakoc CR, Tuveson DA. SOAT1 promotes mevalonate pathway dependency in pancreatic cancer. J Exp Med 2020; 217:151922. [PMID: 32633781 PMCID: PMC7478739 DOI: 10.1084/jem.20192389] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 03/28/2020] [Accepted: 05/12/2020] [Indexed: 12/31/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis, and new therapies are needed. Altered metabolism is a cancer vulnerability, and several metabolic pathways have been shown to promote PDAC. However, the changes in cholesterol metabolism and their role during PDAC progression remain largely unknown. Here we used organoid and mouse models to determine the drivers of altered cholesterol metabolism in PDAC and the consequences of its disruption on tumor progression. We identified sterol O-acyltransferase 1 (SOAT1) as a key player in sustaining the mevalonate pathway by converting cholesterol to inert cholesterol esters, thereby preventing the negative feedback elicited by unesterified cholesterol. Genetic targeting of Soat1 impairs cell proliferation in vitro and tumor progression in vivo and reveals a mevalonate pathway dependency in p53 mutant PDAC cells that have undergone p53 loss of heterozygosity (LOH). In contrast, pancreatic organoids lacking p53 mutation and p53 LOH are insensitive to SOAT1 loss, indicating a potential therapeutic window for inhibiting SOAT1 in PDAC.
Collapse
Affiliation(s)
- Tobiloba E. Oni
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY,Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY
| | - Giulia Biffi
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY,Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Lindsey A. Baker
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY
| | - Yuan Hao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| | - Claudia Tonelli
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY
| | | | - Astrid Deschênes
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY
| | | | - Chang-il Hwang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY,Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA
| | | | - Hilary Cox
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| | - Erin Brosnan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY
| | - Abhishek Doshi
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY
| | - Rebecca P. Lumia
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY
| | - Kimia Khaledi
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY
| | - Youngkyu Park
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY
| | | | - Scott W. Lowe
- Department of Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, NY,Howard Hughes Medical Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | | | | | - David A. Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY,Correspondence to David A. Tuveson:
| |
Collapse
|
30
|
Mechanisms and regulation of cholesterol homeostasis. Nat Rev Mol Cell Biol 2019; 21:225-245. [DOI: 10.1038/s41580-019-0190-7] [Citation(s) in RCA: 1094] [Impact Index Per Article: 182.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2019] [Indexed: 12/14/2022]
|
31
|
Oteng A, Loregger A, van Weeghel M, Zelcer N, Kersten S. Industrial Trans Fatty Acids Stimulate SREBP2-Mediated Cholesterogenesis and Promote Non-Alcoholic Fatty Liver Disease. Mol Nutr Food Res 2019; 63:e1900385. [PMID: 31327168 PMCID: PMC6790681 DOI: 10.1002/mnfr.201900385] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/01/2019] [Indexed: 12/24/2022]
Abstract
SCOPE The mechanisms underlying the deleterious effects of trans fatty acids on plasma cholesterol and non-alcoholic fatty liver disease (NAFLD) are unclear. Here, the aim is to investigate the molecular mechanisms of action of industrial trans fatty acids. METHODS AND RESULTS Hepa1-6 hepatoma cells were incubated with elaidate, oleate, or palmitate. C57Bl/6 mice were fed diets rich in trans-unsaturated, cis-unsaturated, or saturated fatty acids. Transcriptomics analysis of Hepa1-6 cells shows that elaidate but not oleate or palmitate induces expression of genes involved in cholesterol biosynthesis. Induction of cholesterogenesis by elaidate is mediated by increased sterol regulatory element-binding protein 2 (SREBP2) activity and is dependent on SREBP cleavage-activating protein (SCAP), yet independent of liver-X receptor and ubiquitin regulatory X domain-containing protein 8. Elaidate decreases intracellular free cholesterol levels and represses the anticholesterogenic effect of exogenous cholesterol. In mice, the trans-unsaturated diet increases the ratio of liver to gonadal fat mass, steatosis, hepatic cholesterol levels, alanine aminotransferase activity, and fibrosis markers, suggesting enhanced NAFLD, compared to the cis-unsaturated and saturated diets. CONCLUSION Elaidate induces cholesterogenesis in vitro by activating the SCAP-SREBP2 axis, likely by lowering intracellular free cholesterol and attenuating cholesterol-dependent repression of SCAP. This pathway potentially underlies the increase in liver cholesterol and NAFLD by industrial trans fatty acids.
Collapse
Affiliation(s)
- Antwi‐Boasiako Oteng
- Nutrition, Metabolism and Genomics GroupDivision of Human Nutrition and HealthWageningen University6708 WEWageningenThe Netherlands
| | - Anke Loregger
- Department of Medical BiochemistryAcademic Medical CenterUniversity of Amsterdam1105 AZAmsterdamThe Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic DiseasesAmsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences1105 AZAmsterdamThe Netherlands
| | - Noam Zelcer
- Department of Medical BiochemistryAcademic Medical CenterUniversity of Amsterdam1105 AZAmsterdamThe Netherlands
| | - Sander Kersten
- Nutrition, Metabolism and Genomics GroupDivision of Human Nutrition and HealthWageningen University6708 WEWageningenThe Netherlands
| |
Collapse
|
32
|
Yamasaki S, Tomihara T, Kimura G, Ueno Y, Ketema RM, Sato S, Mukai Y, Sikder T, Kurasaki M, Hosokawa T, Saito T. Long-term effects of maternal resveratrol intake during lactation on cholesterol metabolism in male rat offspring. Int J Food Sci Nutr 2019; 71:226-234. [PMID: 31290360 DOI: 10.1080/09637486.2019.1639638] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Resveratrol (RSV) can protect against non-communicable diseases by improving cholesterol metabolism. However, it is unclear that effects of maternal RSV intake on health of adult offspring. In this study, we examined effects of maternal RSV intake during lactation on cholesterol metabolism in adult male rat offspring. Female Wistar rats were fed a control diet (CON) supplemented with or without RSV (20 mg/kg body weight/day) during their lactation period. Male offspring were weaned onto a standard diet and maintained on this diet for 36 weeks. As a result, plasma cholesterol level significantly decreased in RSV offspring compared to CON offspring. Furthermore, a decrease in hepatic 3-hydroxy-3-methylglutaryl-CoA reductase level and an increase in hepatic LDL-receptor level were observed in the RSV offspring. These results indicate that maternal RSV intake causes long-term decrease in plasma cholesterol level in the offspring through suppression of hepatic cholesterol biosynthesis and promotion of hepatic cholesterol uptake.
Collapse
Affiliation(s)
- Shojiro Yamasaki
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Tomomi Tomihara
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Goh Kimura
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Yukako Ueno
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | | | - Shin Sato
- Department of Nutrition, Aomori University of Health and Welfare, Aomori, Japan
| | - Yuuka Mukai
- Faculty of Health and Social Work, Kanagawa University of Human Services, Yokosuka, Japan
| | - Tajuddin Sikder
- Department of Public Health and Informatics, Jahangirnagar University, Dhaka, Bangladesh
| | - Masaaki Kurasaki
- Faculty of Environmental Earth Science, Hokkaido University, Sapporo, Japan
| | - Toshiyuki Hosokawa
- Institute for the Advancement of Higher Education, Hokkaido University, Sapporo, Japan
| | - Takeshi Saito
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan.,Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| |
Collapse
|
33
|
Iwase M, Watanabe K, Shimizu M, Suzuki T, Yamamoto Y, Inoue J, Sato R. Chrysin reduces the activity and protein level of mature forms of sterol regulatory element-binding proteins. Biosci Biotechnol Biochem 2019; 83:1740-1746. [PMID: 31021712 DOI: 10.1080/09168451.2019.1608806] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Sterol regulatory element-binding proteins (SREBPs) are transcription factors that regulate the expression of genes involved in fatty acid and cholesterol biosynthetic pathways. The present study showed that the flavonoid chrysin impairs the fatty acid synthase promoter. Chrysin reduces the expression of SREBP target genes, such as fatty acid synthase, in human hepatoma Huh-7 cells and impairs de novo synthesis of fatty acids and cholesterol. Moreover, it reduces the endogenous mature, transcriptionally active forms of SREBPs, which are generated by the proteolytic processing of precursor forms. In addition, chrysin reduces the enforced expressing mature forms of SREBPs and their transcriptional activity. The ubiquitin-proteasome system is not involved in the chrysin-mediated reduction of SREBPs mature forms. These results suggest that chrysin suppresses SREBP activity, at least partially, via the degradation of SREBPs mature forms. Abbreviations: ACC1: acetyl-CoA carboxylase 1; DMEM: Dulbecco's modified Eagle's medium; FAS: fatty acid synthase; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; 25-HC: 25-hydroxycholesterol; HMGCS: HMG-CoA synthase; LDH: lactate dehydrogenase; LPDS: lipoprotein-deficient serum; PI3K: phosphatidylinositol 3-kinase; SCD1: stearoyl-CoA desaturase; SREBPs: sterol regulatory element-binding proteins.
Collapse
Affiliation(s)
- Masamori Iwase
- a Food Biochemistry laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo , Tokyo , Japan
| | - Kyoko Watanabe
- b Department of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture , Tokyo , Japan
| | - Makoto Shimizu
- a Food Biochemistry laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo , Tokyo , Japan
| | - Tsukasa Suzuki
- b Department of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture , Tokyo , Japan
| | - Yuji Yamamoto
- b Department of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture , Tokyo , Japan
| | - Jun Inoue
- a Food Biochemistry laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo , Tokyo , Japan.,b Department of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture , Tokyo , Japan
| | - Ryuichiro Sato
- a Food Biochemistry laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo , Tokyo , Japan.,c Nutri-Life Science laboratory, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo , Tokyo , Japan
| |
Collapse
|
34
|
Shu Z, Gao Y, Zhang G, Zhou Y, Cao J, Wan D, Zhu X, Xiong W. A functional interaction between Hippo-YAP signalling and SREBPs mediates hepatic steatosis in diabetic mice. J Cell Mol Med 2019; 23:3616-3628. [PMID: 30821074 PMCID: PMC6484311 DOI: 10.1111/jcmm.14262] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/21/2019] [Accepted: 02/11/2019] [Indexed: 12/14/2022] Open
Abstract
The Hippo pathway is an evolutionarily conserved regulator of organ size and tumorigenesis that negatively regulates cell growth and survival. Whether the Hippo pathway regulates cell metabolism is unknown. Here, we report that in the nucleus of hepatocytes, Yes‐associated protein(YAP)—the terminal effector of the Hippo pathway—directly interacts with sterol regulatory element binding proteins (SREBP‐1c and SREBP‐2) on the promoters of the fatty acid synthase (FAS) and 30‐hydroxylmethyl glutaryl coenzyme A reductase (HMGCR), thereby stimulating their transcription and promoting hepatocyte lipogenesis and cholesterol synthesis. In diet‐induced diabetic mice, either Lats1 overexpression or YAP knockdown protects against hepatic steatosis and hyperlipidaemia through suppression of the interaction between YAP and SREBP‐1c/SREBP‐2. These results suggest that YAP is a nuclear co‐factor of SREBPs and that the Hippo pathway negatively affects hepatocyte lipogenesis by inhibiting the function of YAP‐SREBP complexes.
Collapse
Affiliation(s)
- Zhiping Shu
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Gao
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guopeng Zhang
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Zhou
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Cao
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongyi Wan
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohua Zhu
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenqian Xiong
- Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Tsai JW, Kostyleva R, Chen PL, Rivas-Serna IM, Clandinin MT, Meinertzhagen IA, Clandinin TR. Transcriptional Feedback Links Lipid Synthesis to Synaptic Vesicle Pools in Drosophila Photoreceptors. Neuron 2019; 101:721-737.e4. [PMID: 30737130 PMCID: PMC8053036 DOI: 10.1016/j.neuron.2019.01.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 12/03/2018] [Accepted: 01/09/2019] [Indexed: 02/06/2023]
Abstract
Neurons can maintain stable synaptic connections across adult life. However, the signals that regulate expression of synaptic proteins in the mature brain are incompletely understood. Here, we describe a transcriptional feedback loop between the biosynthesis and repertoire of specific phospholipids and the synaptic vesicle pool in adult Drosophila photoreceptors. Mutations that disrupt biosynthesis of a subset of phospholipids cause degeneration of the axon terminal and loss of synaptic vesicles. Although degeneration of the axon terminal is dependent on neural activity, activation of sterol regulatory element binding protein (SREBP) is both necessary and sufficient to cause synaptic vesicle loss. Our studies demonstrate that SREBP regulates synaptic vesicle levels by interacting with tetraspanins, critical organizers of membranous organelles. SREBP is an evolutionarily conserved regulator of lipid biosynthesis in non-neuronal cells; our studies reveal a surprising role for this feedback loop in maintaining synaptic vesicle pools in the adult brain.
Collapse
Affiliation(s)
- Jessica W Tsai
- Department of Neurobiology, Stanford University, Fairchild D200, 299 W. Campus Drive, Stanford, CA 94305, USA
| | - Ripsik Kostyleva
- Department of Psychology and Neuroscience, Life Sciences Centre, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Pei-Ling Chen
- Department of Neurobiology, Stanford University, Fairchild D200, 299 W. Campus Drive, Stanford, CA 94305, USA
| | - Irma Magaly Rivas-Serna
- Department of Agriculture, Food, and Nutritional Science, Alberta Institute of Human Nutrition, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - M Thomas Clandinin
- Department of Agriculture, Food, and Nutritional Science, Alberta Institute of Human Nutrition, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Ian A Meinertzhagen
- Department of Psychology and Neuroscience, Life Sciences Centre, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Thomas R Clandinin
- Department of Neurobiology, Stanford University, Fairchild D200, 299 W. Campus Drive, Stanford, CA 94305, USA.
| |
Collapse
|
36
|
Shao F, Wang X, Yu J, Shen K, Qi C, Gu Z. Expression of miR-33 from an SREBP2 intron inhibits the expression of the fatty acid oxidation-regulatory genes CROT and HADHB in chicken liver. Br Poult Sci 2019; 60:115-124. [PMID: 30698464 DOI: 10.1080/00071668.2018.1564242] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
1. Limiting the growth of adipose tissue in chickens is a major issue in the poultry industry. In chickens, de novo synthesis of lipids occurs primarily in the liver. Thus, it is necessary to understand how fatty acid accumulation in the liver is controlled. The miR-33 is an intronic microRNA (miRNA) of the chicken sterol regulatory element binding transcription factor 2 (SREBF2), which is a master switch in activating many genes involved in the uptake and synthesis of cholesterol, triglycerides, fatty acids and phospholipids. 2. In the current study, the genes CROT and HADHB known to encode enzymes critical for fatty acid oxidation were predicted to be potential targets of miR-33 in chickens via the miRNA target prediction programs 'miRanda' and 'TargetScan'. Co-transfection and dual-luciferase reporter assays showed that the expression of luciferase reporter gene linked to the 3'-untranslated region (3'UTR) of the chicken CROT and HADHB mRNA was down-regulated by overexpression of the chicken miR-33 (P < 0.05). This down-regulation was completely abolished when the predicted miR-33 target sites in the CROT and HADHB 3'UTR were mutated. 3. Transfecting miR-33 mimics into the LMH cells led to a decrease in the mRNA expression of CROT and HADHB (P < 0.01), and this transfection had a similar effect on the proteins (P < 0.05). In contrast, the expression of CROT in primary chicken hepatocytes was up-regulated after transfection with the miR-33 inhibitor LNA-anti-miR-33 (P < 0.05). 4. Using quantitative RT-PCR, it was shown that the expression of miR-33 was increased in the chicken liver from day 0 to day 49 of age, whereas the CROT and HADHB mRNA levels decreased during the same period. 5. These findings support the conclusion that miR-33 might play an important role in lipid metabolism in the chicken liver by negatively regulating the expression of the CROT and HADHB genes, which encode enzymes critical for lipid oxidation.
Collapse
Affiliation(s)
- F Shao
- a Department of Life Science and Technology , Changshu Institute of Technology , Changshu, Jiangsu , China.,b Medical Research Centre , The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University , Changzhou, Jiangsu , China
| | - X Wang
- c Jiangsu Institute of Poultry Science , Yangzhou , Jiangsu , China
| | - J Yu
- a Department of Life Science and Technology , Changshu Institute of Technology , Changshu, Jiangsu , China
| | - K Shen
- b Medical Research Centre , The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University , Changzhou, Jiangsu , China
| | - C Qi
- b Medical Research Centre , The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University , Changzhou, Jiangsu , China
| | - Z Gu
- a Department of Life Science and Technology , Changshu Institute of Technology , Changshu, Jiangsu , China
| |
Collapse
|
37
|
Guo C, Chi Z, Jiang D, Xu T, Yu W, Wang Z, Chen S, Zhang L, Liu Q, Guo X, Zhang X, Li W, Lu L, Wu Y, Song BL, Wang D. Cholesterol Homeostatic Regulator SCAP-SREBP2 Integrates NLRP3 Inflammasome Activation and Cholesterol Biosynthetic Signaling in Macrophages. Immunity 2018; 49:842-856.e7. [PMID: 30366764 DOI: 10.1016/j.immuni.2018.08.021] [Citation(s) in RCA: 212] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 07/25/2018] [Accepted: 08/27/2018] [Indexed: 11/28/2022]
Abstract
Cholesterol metabolism has been linked to immune functions, but the mechanisms by which cholesterol biosynthetic signaling orchestrates inflammasome activation remain unclear. Here, we have shown that NLRP3 inflammasome activation is integrated with the maturation of cholesterol master transcription factor SREBP2. Importantly, SCAP-SREBP2 complex endoplasmic reticulum-to-Golgi translocation was required for optimal activation of the NLRP3 inflammasome both in vitro and in vivo. Enforced cholesterol biosynthetic signaling by sterol depletion or statins promoted NLPR3 inflammasome activation. However, this regulation did not predominantly depend on changes in cholesterol homeostasis controlled by the transcriptional activity of SREBP2, but relied on the escort activity of SCAP. Mechanistically, NLRP3 associated with SCAP-SREBP2 to form a ternary complex which translocated to the Golgi apparatus adjacent to a mitochondrial cluster for optimal inflammasome assembly. Our study reveals that, in addition to controlling cholesterol biosynthesis, SCAP-SREBP2 also serves as a signaling hub integrating cholesterol metabolism with inflammation in macrophages.
Collapse
Affiliation(s)
- Chuansheng Guo
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China; Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China
| | - Zhexu Chi
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China; Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China
| | - Danlu Jiang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China; Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China
| | - Ting Xu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China; Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China
| | - Weiwei Yu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, P.R. China
| | - Zhen Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China; Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China
| | - Sheng Chen
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China; Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China
| | - Li Zhang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China; Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China
| | - Qianyun Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, P.R. China
| | - Xingchen Guo
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, P.R. China
| | - Xue Zhang
- Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China; Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China
| | - Wenxin Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, P.R. China
| | - Linrong Lu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China; Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China
| | - Yingliang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, P.R. China
| | - Bao-Liang Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, P.R. China
| | - Di Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China; Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou 310058, P.R. China.
| |
Collapse
|
38
|
Weber K, Casali C, Gaveglio V, Pasquaré S, Morel Gómez E, Parra L, Erjavec L, Perazzo C, Fernández Tome MC. TAG synthesis and storage under osmotic stress. A requirement for preserving membrane homeostasis in renal cells. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:1108-1120. [DOI: 10.1016/j.bbalip.2018.06.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/11/2018] [Accepted: 06/12/2018] [Indexed: 12/14/2022]
|
39
|
de la Roche M, Hamilton C, Mortensen R, Jeyaprakash AA, Ghosh S, Anand PK. Trafficking of cholesterol to the ER is required for NLRP3 inflammasome activation. J Cell Biol 2018; 217:3560-3576. [PMID: 30054450 PMCID: PMC6168277 DOI: 10.1083/jcb.201709057] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 12/19/2017] [Accepted: 07/13/2018] [Indexed: 01/01/2023] Open
Abstract
Cellular lipid metabolism is being increasingly recognized to influence inflammatory responses. de la Roche et al. reveal that cellular sterol trafficking to the endoplasmic reticulum is required for the assembly and the activation of the NLRP3 inflammasome, thereby coupling lipid homeostasis to innate immune signaling. Cellular lipids determine membrane integrity and fluidity and are being increasingly recognized to influence immune responses. Cellular cholesterol requirements are fulfilled through biosynthesis and uptake programs. In an intricate pathway involving the lysosomal cholesterol transporter NPC1, the sterol gets unequally distributed across intracellular compartments. By using pharmacological and genetic approaches targeting NPC1, we reveal that blockade of cholesterol trafficking through the late endosome–lysosome pathway blunts NLRP3 inflammasome activation. Altered cholesterol localization at the plasma membrane (PM) in Npc1−/− cells abrogated AKT–mTOR signaling by TLR4. However, the inability to activate the NLRP3 inflammasome was traced to perturbed cholesterol trafficking to the ER but not the PM. Accordingly, acute cholesterol depletion in the ER membranes by statins abrogated casp-1 activation and IL-1β secretion and ablated NLRP3 inflammasome assembly. By contrast, assembly and activation of the AIM2 inflammasome progressed unrestricted. Together, this study reveals ER sterol levels as a metabolic rheostat for the activation of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Marianne de la Roche
- Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London, UK
| | - Claire Hamilton
- Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London, UK
| | - Rebecca Mortensen
- Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London, UK
| | - A Arockia Jeyaprakash
- Wellcome Trust Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Sanjay Ghosh
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Paras K Anand
- Infectious Diseases and Immunity, Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
40
|
Inoue J, Miyata S, Shimizu M, Sato R. Isoxanthohumol stimulates ubiquitin-proteasome-dependent degradation of precursor forms of sterol regulatory element-binding proteins. Biosci Biotechnol Biochem 2018; 82:1591-1598. [PMID: 29804513 DOI: 10.1080/09168451.2018.1478715] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Sterol regulatory element-binding proteins (SREBPs) are transcription factors that regulate a wide variety of genes involved in fatty acid and cholesterol synthesis. In the present study, we identified that isoxanthohumol (IXN) suppressed SREBP activity. Low concentrations of IXN (10 and 30 μM) reduced the amount of mature forms of SREBPs, while high concentration of IXN (100 μM) reduced both precursor and mature forms of SREBPs in Huh-7 cells. The IXN-mediated decrease in the precursor forms of SREBPs in Huh-7 cells was completely abolished by culturing cells under sterol-supplemented conditions and was partly abolished by treatment with a proteasome inhibitor, MG132, but not a lysosome inhibitor, NH4Cl. Moreover, IXN accelerated the ubiquitination of the precursor forms of SREBP-1a. These results suggest that IXN suppresses SREBP activity, at least in part, via ubiquitin-proteasome-dependent degradation of the precursor forms of SREBPs. ABBREVIATIONS ACC1: acetyl-CoA carboxylase 1; DMEM: Dulbecco's modified Eagle's medium; ER: endoplasmic reticulum; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; 25-HC: 25-hydroxycholesterol; HMGCR: HMG-CoA reductase; HMGCS: HMG-CoA synthase; Insig: insulin-induced gene; IXN: isoxanthohumol; LPDS: lipoprotein-deficient serum; SCAP: SREBP cleavage-activating protein; SCD1: stearoyl-CoA desaturase; SREBPs: sterol regulatory element-binding proteins; XN: xanthohumol.
Collapse
Affiliation(s)
- Jun Inoue
- a Department of Agricultural Chemistry, Faculty of Applied Biosciences , Tokyo University of Agriculture , Tokyo , Japan.,b Food Biochemistry laboratory, Department of Applied Biological Chemistry , Graduate School of Agricultural and Life Sciences, The University of Tokyo , Tokyo , Japan
| | - Shingo Miyata
- b Food Biochemistry laboratory, Department of Applied Biological Chemistry , Graduate School of Agricultural and Life Sciences, The University of Tokyo , Tokyo , Japan
| | - Makoto Shimizu
- b Food Biochemistry laboratory, Department of Applied Biological Chemistry , Graduate School of Agricultural and Life Sciences, The University of Tokyo , Tokyo , Japan
| | - Ryuichiro Sato
- b Food Biochemistry laboratory, Department of Applied Biological Chemistry , Graduate School of Agricultural and Life Sciences, The University of Tokyo , Tokyo , Japan.,c Nutri-Life Science laboratory, Department of Applied Biological Chemistry , Graduate School of Agricultural and Life Sciences, The University of Tokyo , Tokyo , Japan
| |
Collapse
|
41
|
Heilos D, Röhrl C, Pirker C, Englinger B, Baier D, Mohr T, Schwaiger M, Iqbal SM, van Schoonhoven S, Klavins K, Eberhart T, Windberger U, Taibon J, Sturm S, Stuppner H, Koellensperger G, Dornetshuber-Fleiss R, Jäger W, Lemmens-Gruber R, Berger W. Altered membrane rigidity via enhanced endogenous cholesterol synthesis drives cancer cell resistance to destruxins. Oncotarget 2018; 9:25661-25680. [PMID: 29876015 PMCID: PMC5986646 DOI: 10.18632/oncotarget.25432] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/25/2018] [Indexed: 12/31/2022] Open
Abstract
Destruxins, secondary metabolites of entomopathogenic fungi, exert a wide variety of interesting characteristics ranging from antiviral to anticancer effects. Although their mode of action was evaluated previously, the molecular mechanisms of resistance development are unknown. Hence, we have established destruxin-resistant sublines of HCT116 colon carcinoma cells by selection with the most prevalent derivatives, destruxin (dtx)A, dtxB and dtxE. Various cell biological and molecular techniques were applied to elucidate the regulatory mechanisms underlying these acquired and highly stable destruxin resistance phenotypes. Interestingly, well-known chemoresistance-mediating ABC efflux transporters were not the major players. Instead, in dtxA- and dtxB-resistant cells a hyper-activated mevalonate pathway was uncovered resulting in increased de-novo cholesterol synthesis rates and elevated levels of lanosterol, cholesterol as well as several oxysterol metabolites. Accordingly, inhibition of the mevalonate pathway at two different steps, using either statins or zoledronic acid, significantly reduced acquired but also intrinsic destruxin resistance. Vice versa, cholesterol supplementation protected destruxin-sensitive cells against their cytotoxic activity. Additionally, an increased cell membrane adhesiveness of dtxA-resistant as compared to parental cells was detected by atomic force microscopy. This was paralleled by a dramatically reduced ionophoric capacity of dtxA in resistant cells when cultured in absence but not in presence of statins. Summarizing, our results suggest a reduced ionophoric activity of destruxins due to cholesterol-mediated plasma membrane re-organization as molecular mechanism underlying acquired destruxin resistance in human colon cancer cells. Whether this mechanism might be valid also in other cell types and organisms exposed to destruxins e.g. as bio-insecticides needs to be evaluated.
Collapse
Affiliation(s)
- Daniela Heilos
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Comprehensive Cancer Center of the Medical University of Vienna, Vienna, Austria
- Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Clemens Röhrl
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Christine Pirker
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Comprehensive Cancer Center of the Medical University of Vienna, Vienna, Austria
| | - Bernhard Englinger
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Comprehensive Cancer Center of the Medical University of Vienna, Vienna, Austria
| | - Dina Baier
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Comprehensive Cancer Center of the Medical University of Vienna, Vienna, Austria
- Decentralized Biomedical Facilities of the Medical University of Vienna, Vienna, Austria
| | - Thomas Mohr
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Comprehensive Cancer Center of the Medical University of Vienna, Vienna, Austria
| | - Michaela Schwaiger
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | | | - Sushilla van Schoonhoven
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Comprehensive Cancer Center of the Medical University of Vienna, Vienna, Austria
| | | | - Tanja Eberhart
- Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Ursula Windberger
- Decentralized Biomedical Facilities of the Medical University of Vienna, Vienna, Austria
| | - Judith Taibon
- Institute of Pharmacy, Pharmacognosy and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Sonja Sturm
- Institute of Pharmacy, Pharmacognosy and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Hermann Stuppner
- Institute of Pharmacy, Pharmacognosy and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Gunda Koellensperger
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Vienna Metabolomics Center, University of Vienna, Vienna, Austria
| | - Rita Dornetshuber-Fleiss
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Comprehensive Cancer Center of the Medical University of Vienna, Vienna, Austria
- Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Walter Jäger
- Department of Pharmaceutical Chemistry, Division of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Rosa Lemmens-Gruber
- Department of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Comprehensive Cancer Center of the Medical University of Vienna, Vienna, Austria
| |
Collapse
|
42
|
Endosomal-Lysosomal Cholesterol Sequestration by U18666A Differentially Regulates Amyloid Precursor Protein (APP) Metabolism in Normal and APP-Overexpressing Cells. Mol Cell Biol 2018. [PMID: 29530923 DOI: 10.1128/mcb.00529-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Amyloid β (Aβ) peptide, derived from amyloid precursor protein (APP), plays a critical role in the development of Alzheimer's disease. Current evidence indicates that altered levels or subcellular distribution of cholesterol can regulate Aβ production and clearance, but it remains unclear how cholesterol sequestration within the endosomal-lysosomal (EL) system can influence APP metabolism. Thus, we evaluated the effects of U18666A, which triggers cholesterol redistribution within the EL system, on mouse N2a cells expressing different levels of APP in the presence or absence of extracellular cholesterol and lipids provided by fetal bovine serum (FBS). Our results reveal that U18666A and FBS differentially increase the levels of APP and its cleaved products, the α-, β-, and η-C-terminal fragments, in N2a cells expressing normal levels of mouse APP (N2awt), higher levels of human wild-type APP (APPwt), or "Swedish" mutant APP (APPsw). The cellular levels of Aβ1-40/Aβ1-42 were markedly increased in U18666A-treated APPwt and APPsw cells. Our studies further demonstrate that APP and its cleaved products are partly accumulated in the lysosomes, possibly due to decreased clearance. Finally, we show that autophagy inhibition plays a role in mediating U18666A effects. Collectively, these results suggest that altered levels and distribution of cholesterol and lipids can differentially regulate APP metabolism depending on the nature of APP expression.
Collapse
|
43
|
DeBose-Boyd RA, Ye J. SREBPs in Lipid Metabolism, Insulin Signaling, and Beyond. Trends Biochem Sci 2018; 43:358-368. [PMID: 29500098 DOI: 10.1016/j.tibs.2018.01.005] [Citation(s) in RCA: 232] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 12/22/2022]
Abstract
Sterol regulatory element-binding proteins (SREBPs) are a family of membrane-bound transcription factors that activate genes encoding enzymes required for synthesis of cholesterol and unsaturated fatty acids. SREBPs are controlled by multiple mechanisms at the level of mRNA synthesis, proteolytic activation, and transcriptional activity. In this review, we summarize the recent findings that contribute to the current understanding of the regulation of SREBPs and their physiologic roles in maintenance of lipid homeostasis, insulin signaling, innate immunity, and cancer development.
Collapse
Affiliation(s)
- Russell A DeBose-Boyd
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9046, USA.
| | - Jin Ye
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9046, USA.
| |
Collapse
|
44
|
Aikawa T, Holm ML, Kanekiyo T. ABCA7 and Pathogenic Pathways of Alzheimer's Disease. Brain Sci 2018; 8:E27. [PMID: 29401741 PMCID: PMC5836046 DOI: 10.3390/brainsci8020027] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/01/2018] [Accepted: 02/03/2018] [Indexed: 12/22/2022] Open
Abstract
The ATP-binding cassette (ABC) reporter family functions to regulate the homeostasis of phospholipids and cholesterol in the central nervous system, as well as peripheral tissues. ABCA7 belongs to the A subfamily of ABC transporters, which shares 54% sequence identity with ABCA1. While ABCA7 is expressed in a variety of tissues/organs, including the brain, recent genome-wide association studies (GWAS) have identified ABCA7 gene variants as susceptibility loci for late-onset Alzheimer's disease (AD). More important, subsequent genome sequencing analyses have revealed that premature termination codon mutations in ABCA7 are associated with the increased risk for AD. Alzheimer's disease is a progressive neurodegenerative disease and the most common cause of dementia, where the accumulation and deposition of amyloid-β (Aβ) peptides cleaved from amyloid precursor protein (APP) in the brain trigger the pathogenic cascade of the disease. In consistence with human genetic studies, increasing evidence has demonstrated that ABCA7 deficiency exacerbates Aβ pathology using in vitro and in vivo models. While ABCA7 has been shown to mediate phagocytic activity in macrophages, ABCA7 is also involved in the microglial Aβ clearance pathway. Furthermore, ABCA7 deficiency results in accelerated Aβ production, likely by facilitating endocytosis and/or processing of APP. Taken together, current evidence suggests that ABCA7 loss-of-function contributes to AD-related phenotypes through multiple pathways. A better understanding of the function of ABCA7 beyond lipid metabolism in both physiological and pathological conditions becomes increasingly important to explore AD pathogenesis.
Collapse
Affiliation(s)
- Tomonori Aikawa
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| | - Marie-Louise Holm
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| |
Collapse
|
45
|
Kelsey I, Zbinden M, Byles V, Torrence M, Manning BD. mTORC1 suppresses PIM3 expression via miR-33 encoded by the SREBP loci. Sci Rep 2017; 7:16112. [PMID: 29170467 PMCID: PMC5701013 DOI: 10.1038/s41598-017-16398-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/13/2017] [Indexed: 12/31/2022] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) is a central regulator of cell growth that is often aberrantly activated in cancer. However, mTORC1 inhibitors, such as rapamycin, have limited effectiveness as single agent cancer therapies, with feedback mechanisms inherent to the signaling network thought to diminish the anti-tumor effects of mTORC1 inhibition. Here, we identify the protein kinase and proto-oncogene PIM3 as being repressed downstream of mTORC1 signaling. PIM3 expression is suppressed in cells with loss of the tuberous sclerosis complex (TSC) tumor suppressors, which exhibit growth factor-independent activation of mTORC1, and in the mouse liver upon feeding-induced activation of mTORC1. Inhibition of mTORC1 with rapamycin induces PIM3 transcript and protein levels in a variety of settings. Suppression of PIM3 involves the sterol regulatory element-binding (SREBP) transcription factors SREBP1 and 2, whose activation and mRNA expression are stimulated by mTORC1 signaling. We find that PIM3 repression is mediated by miR-33, an intronic microRNA encoded within the SREBP loci, the expression of which is decreased with rapamycin. These results demonstrate that PIM3 is induced upon mTORC1 inhibition, with potential implications for the effects of mTORC1 inhibitors in TSC, cancers, and the many other disease settings influenced by aberrant mTORC1 signaling.
Collapse
Affiliation(s)
- Ilana Kelsey
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Marie Zbinden
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Vanessa Byles
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Margaret Torrence
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Brendan D Manning
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
46
|
Mohamed A, Viveiros A, Williams K, Posse de Chaves E. Aβ inhibits SREBP-2 activation through Akt inhibition. J Lipid Res 2017; 59:1-13. [PMID: 29122977 PMCID: PMC5748492 DOI: 10.1194/jlr.m076703] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 10/21/2017] [Indexed: 12/20/2022] Open
Abstract
We previously demonstrated that oligomeric amyloid β42 (oAβ42) inhibits the mevalonate pathway impairing cholesterol synthesis and protein prenylation. Enzymes of the mevalonate pathway are regulated by the transcription factor SREBP-2. Here, we show that in several neuronal types challenged with oAβ42, SREBP-2 activation is reduced. Moreover, SREBP-2 activation is also decreased in the brain cortex of the Alzheimer's disease (AD) mouse model, TgCRND8, suggesting that SREBP-2 may be affected in vivo early in the disease. We demonstrate that oAβ42 does not affect enzymatic cleavage of SREBP-2 per se, but may impair SREBP-2 transport from the endoplasmic reticulum (ER) to the Golgi. Trafficking of SREBP-2 from the ER to the Golgi requires protein kinase B (Akt) activation. oAβ42 significantly reduces Akt phosphorylation and this decrease is responsible for the decline in SREBP-2 activation. Overexpression of constitutively active Akt prevents the effect of oAβ42 on SREBP-2 and the downstream inhibition of cholesterol synthesis and protein prenylation. Our work provides a novel mechanistic link between Aβ and the mevalonate pathway, which will impact the views on issues related to cholesterol, isoprenoids, and statins in AD. We also identify SREBP-2 as an indirect target of Akt in neurons, which may play a role in the cross-talk between AD and diabetes.
Collapse
Affiliation(s)
- Amany Mohamed
- Department of Pharmacology and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Anissa Viveiros
- Department of Pharmacology and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Kathleen Williams
- Department of Pharmacology and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Elena Posse de Chaves
- Department of Pharmacology and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
47
|
Branched-chain amino acids prevent hepatic fibrosis and development of hepatocellular carcinoma in a non-alcoholic steatohepatitis mouse model. Oncotarget 2017; 8:18191-18205. [PMID: 28212548 PMCID: PMC5392319 DOI: 10.18632/oncotarget.15304] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 12/27/2016] [Indexed: 01/06/2023] Open
Abstract
Oral supplementation with branched-chain amino acids (BCAA; leucine, isoleucine, and valine) in patients with liver cirrhosis potentially suppresses the incidence of hepatocellular carcinoma (HCC) and improves event-free survival. However, the detailed mechanisms of BCAA action have not been fully elucidated. BCAA were administered to atherogenic and high-fat (Ath+HF) diet-induced nonalcoholic steatohepatitis (NASH) model mice. Liver histology, tumor incidence, and gene expression profiles were evaluated. Ath+HF diet mice developed hepatic tumors at a high frequency at 68 weeks. BCAA supplementation significantly improved hepatic steatosis, inflammation, fibrosis, and tumors in Ath+HF mice at 68 weeks. GeneChip analysis demonstrated the significant resolution of pro-fibrotic gene expression by BCAA supplementation. The anti-fibrotic effect of BCAA was confirmed further using platelet-derived growth factor C transgenic mice, which develop hepatic fibrosis and tumors. In vitro, BCAA restored the transforming growth factor (TGF)-β1-stimulated expression of pro-fibrotic genes in hepatic stellate cells (HSC). In hepatocytes, BCAA restored TGF-β1-induced apoptosis, lipogenesis, and Wnt/β-Catenin signaling, and inhibited the transformation of WB-F344 rat liver epithelial stem-like cells. BCAA repressed the promoter activity of TGFβ1R1 by inhibiting the expression of the transcription factor NFY and histone acetyltransferase p300. Interestingly, the inhibitory effect of BCAA on TGF-β1 signaling was mTORC1 activity-dependent, suggesting the presence of negative feedback regulation from mTORC1 to TGF-β1 signaling. Thus, BCAA induce an anti-fibrotic effect in HSC, prevent apoptosis in hepatocytes, and decrease the incidence of HCC; therefore, BCAA supplementation would be beneficial for patients with advanced liver fibrosis with a high risk of HCC.
Collapse
|
48
|
Korošec T, Tomažin U, Horvat S, Keber R, Salobir J. The diverse effects of α- and γ-tocopherol on chicken liver transcriptome. Poult Sci 2017; 96:667-680. [PMID: 27587731 DOI: 10.3382/ps/pew296] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 07/11/2016] [Indexed: 01/11/2023] Open
Abstract
α-Tocopherol is the form of vitamin E with the highest biological value and is almost exclusively considered as vitamin E in feed and feed supplements. Because γ-tocopherol, the predominant form of vitamin E naturally present in chicken feed, is not considered as a source of vitamin E, its re-evaluation with newer methods might be important.Despite γ-tocopherol's lower estimated biological value, it has been shown to be effective in reducing reactive nitrogen species, regulating immune and inflammatory processes, and diminishing the risk of metabolic perturbations and associated diseases. A 30-day nutritional trial in broiler chickens (Ross 308) was conducted to investigate how specific forms of vitamin E (α- and γ-tocopherol) and their combination impact liver gene expression when oxidative susceptibility of the organism is induced by high n-3 polyunsaturated fatty acids (PUFA) intake (linseed oil). Thirty-six one-day-old male broilers were fed a diet enriched with 5% linseed oil. A control group (Cont; N = 10) was used as a reference group, Tα (N = 10) was supplemented with 67 mg/kg RRR-α-tocopherol, Tγ (N = 8) with 67 mg/kg RRR-γ-tocopherol, and Tαγ (N = 8) with a combination of 33.5 mg/kg of each tocopherol. Beside oxidative stress indicators, whole chicken genome microarray analysis was performed on liver RNA and selected differentially expressed genes were confirmed by real time quantitative PCR. α-Tocopherol alone and in combination with γ-tocopherol was able to prevent lipid oxidation, which was also supported by transcriptome analysis. The effect of γ-tocopherol was evident in the expression of genes involved in inflammatory processes and immune response, while α-tocopherol affected genes involved in lipid and cholesterol metabolism. Both isomers of vitamin E influenced the transcription of genes, which are related to improved fat oxidation and enhanced glucose sparing.
Collapse
Affiliation(s)
- Tamara Korošec
- University of Ljubljana, Biotechnical Faculty, Department of Animal Science, Groblje 3, 1230 Domžale, Slovenia
| | - Urška Tomažin
- Agricultural Institute of Slovenia, Hacquetova ulica 17, 1000 Ljubljana, Slovenia
| | - Simon Horvat
- University of Ljubljana, Biotechnical Faculty, Department of Animal Science, Groblje 3, 1230 Domžale, Slovenia.,National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia
| | - Rok Keber
- Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Janez Salobir
- University of Ljubljana, Biotechnical Faculty, Department of Animal Science, Groblje 3, 1230 Domžale, Slovenia
| |
Collapse
|
49
|
Li Y, Song Y, Zhao M, Guo Y, Yu C, Chen W, Shao S, Xu C, Zhou X, Zhao L, Zhang Z, Bo T, Xia Y, Proud CG, Wang X, Wang L, Zhao J, Gao L. A novel role for CRTC2 in hepatic cholesterol synthesis through SREBP-2. Hepatology 2017; 66:481-497. [PMID: 28395113 PMCID: PMC5575482 DOI: 10.1002/hep.29206] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/29/2017] [Accepted: 04/06/2017] [Indexed: 02/06/2023]
Abstract
Cholesterol synthesis is regulated by the transcription factor sterol regulatory element binding protein 2 (SREBP-2) and its target gene 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR), which is the rate-limiting enzyme in cholesterol synthesis. Cyclic adenosine monophosphate-responsive element (CRE) binding protein-regulated transcription coactivator (CRTC) 2 is the master regulator of glucose metabolism. However, the effect of CRTC2 on cholesterol and its potential molecular mechanism remain unclear. Here, we demonstrated that CRTC2 expression and liver cholesterol content were increased in patients with high serum cholesterol levels who underwent resection of liver hemangiomas, as well as in mice fed a 4% cholesterol diet. Mice with adenovirus-mediated CRTC2 overexpression also showed elevated lipid levels in both serum and liver tissues. Intriguingly, hepatic de novo cholesterol synthesis was markedly increased under these conditions. In contrast, CRTC2 ablation in mice fed a 4% cholesterol diet (18 weeks) showed decreased lipid levels in serum and liver tissues compared with those in littermate wild-type mice. The expression of lipogenic genes (SREBP-2 and HMGCR) was consistent with hepatic CRTC2 levels. In vivo imaging showed enhanced adenovirus-mediated HMGCR-luciferase activity in adenovirus-mediated CRTC2 mouse livers; however, the activity was attenuated after mutation of CRE or sterol regulatory element sequences in the HMGCR reporter construct. The effect of CRTC2 on HMGCR in mouse livers was alleviated upon SREBP-2 knockdown. CRTC2 modulated SREBP-2 transcription by CRE binding protein, which recognizes the half-site CRE sequence in the SREBP-2 promoter. CRTC2 reduced the nuclear protein expression of forkhead box O1 and subsequently increased SREBP-2 transcription by binding insulin response element 1, rather than insulin response element 2, in the SREBP-2 promoter. CONCLUSION CRTC2 regulates the transcription of SREBP-2 by interfering with the recognition of insulin response element 1 in the SREBP-2 promoter by forkhead box O1, thus inducing SREBP-2/HMGCR signaling and subsequently facilitating hepatic cholesterol synthesis. (Hepatology 2017;66:481-497).
Collapse
|
50
|
Wang WA, Liu WX, Durnaoglu S, Lee SK, Lian J, Lehner R, Ahnn J, Agellon LB, Michalak M. Loss of Calreticulin Uncovers a Critical Role for Calcium in Regulating Cellular Lipid Homeostasis. Sci Rep 2017; 7:5941. [PMID: 28725049 PMCID: PMC5517566 DOI: 10.1038/s41598-017-05734-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/02/2017] [Indexed: 01/28/2023] Open
Abstract
A direct link between Ca2+ and lipid homeostasis has not been definitively demonstrated. In this study, we show that manipulation of ER Ca2+ causes the re-distribution of a portion of the intracellular unesterified cholesterol to a pool that is not available to the SCAP-SREBP complex. The SREBP processing pathway in ER Ca2+ depleted cells remained fully functional and responsive to changes in cellular cholesterol status but differed unexpectedly in basal activity. These findings establish the role of Ca2+ in determining the reference set-point for controlling cellular lipid homeostasis. We propose that ER Ca2+ status is an important determinant of the basal sensitivity of the sterol sensing mechanism inherent to the SREBP processing pathway.
Collapse
Affiliation(s)
- Wen-An Wang
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Wen-Xin Liu
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Serpen Durnaoglu
- Department of Life Sciences, Research Institute for Natural Sciences, BK21 Plus Life Science for BDR Team, Research Institute of Natural Science, Hanyang University, Seoul, 133-791, South Korea
| | - Sun-Kyung Lee
- Department of Life Sciences, Research Institute for Natural Sciences, BK21 Plus Life Science for BDR Team, Research Institute of Natural Science, Hanyang University, Seoul, 133-791, South Korea
| | - Jihong Lian
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Richard Lehner
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Joohong Ahnn
- Department of Life Sciences, Research Institute for Natural Sciences, BK21 Plus Life Science for BDR Team, Research Institute of Natural Science, Hanyang University, Seoul, 133-791, South Korea
| | - Luis B Agellon
- School of Human Nutrition, McGill University, Ste. Anne de Bellevue, Quebec, H9X 3V9, Canada.
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada.
| |
Collapse
|