1
|
Chen M, Yang Y, Chen S, He Z, Du L. Targeting squalene epoxidase in the treatment of metabolic-related diseases: current research and future directions. PeerJ 2024; 12:e18522. [PMID: 39588004 PMCID: PMC11587872 DOI: 10.7717/peerj.18522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024] Open
Abstract
Metabolic-related diseases are chronic diseases caused by multiple factors, such as genetics and the environment. These diseases are difficult to cure and seriously affect human health. Squalene epoxidase (SQLE), the second rate-limiting enzyme in cholesterol synthesis, plays an important role in cholesterol synthesis and alters the gut microbiota and tumor immunity. Research has shown that SQLE is expressed in many tissues and organs and is involved in the occurrence and development of various metabolic-related diseases, such as cancer, nonalcoholic fatty liver disease, diabetes mellitus, and obesity. SQLE inhibitors, such as terbinafine, NB598, natural compounds, and their derivatives, can effectively ameliorate fungal infections, nonalcoholic fatty liver disease, and cancer. In this review, we provide an overview of recent research progress on the role of SQLE in metabolic-related diseases. Further research on the regulation of SQLE expression is highly important for developing drugs for the treatment of metabolic-related diseases with good pharmacological activity.
Collapse
Affiliation(s)
- Mingzhu Chen
- School of Basic Medical Sciences, Chengdu University of Chinese Medicine, Chengdu, Sichuan Province, China
| | - Yongqi Yang
- Harbin Medical University, Department of Pharmacology, College of Pharmacy, Harbin, Heilongjiang Province, China
| | - Shiting Chen
- School of Basic Medical Sciences, Chengdu University of Chinese Medicine, Chengdu, Sichuan Province, China
| | - Zhigang He
- School of Basic Medical Sciences, Chengdu University of Chinese Medicine, Chengdu, Sichuan Province, China
| | - Lian Du
- School of Basic Medical Sciences, Chengdu University of Chinese Medicine, Chengdu, Sichuan Province, China
| |
Collapse
|
2
|
Montesinos J, Kabra K, Uceda M, Larrea D, Agrawal R, Tamucci K, Pera M, Ferre A, Gomez-Lopez N, Yun T, Velasco K, Schon E, Area-Gomez E. The contribution of mitochondria-associated ER membranes to cholesterol homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.622945. [PMID: 39605513 PMCID: PMC11601226 DOI: 10.1101/2024.11.11.622945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Cellular demands for cholesterol are met by a balance between its biosynthesis in the endoplasmic reticulum (ER) and its uptake from lipoproteins. Cholesterol levels in intracellular membranes form a gradient maintained by a complex network of mechanisms including the control of the expression, compartmentalization and allosteric modulation of the enzymes that balance endogenous and exogenous sources of cholesterol. Low-density lipoproteins (LDLs) are internalized and delivered to lysosomal compartments to release their cholesterol content, which is then distributed within cellular membranes. High-density lipoproteins (HDLs), on the other hand, can transfer their cholesterol content directly into cellular membranes through the action of receptors such as the scavenger receptor B type 1 (SR-B1; gene SCARB1). We show here that SR-B1-mediated exogenous cholesterol internalization from HDL stimulates the formation of lipid-raft subdomains in the ER known as mitochondria-associated ER membranes (MAM), that, in turn, suppress de novo cholesterol biosynthesis machinery. We propose that MAM is a regulatory hub for cholesterol homeostasis that offers a novel dimension for understanding the intracellular regulation of this important lipid.
Collapse
Affiliation(s)
- J. Montesinos
- Centro de Investigaciones Biológicas Margarita Salas. CSIC. Madrid, Spain. CIBERNED
| | - K. Kabra
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, USA
| | - M. Uceda
- Centro de Investigaciones Biológicas Margarita Salas. CSIC. Madrid, Spain. CIBERNED
| | - D. Larrea
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - R.R. Agrawal
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, USA
| | - K.A. Tamucci
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, USA
| | - M. Pera
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - A.C. Ferre
- Centro de Investigaciones Biológicas Margarita Salas. CSIC. Madrid, Spain. CIBERNED
| | - N. Gomez-Lopez
- Centro de Investigaciones Biológicas Margarita Salas. CSIC. Madrid, Spain. CIBERNED
| | - T.D. Yun
- Centro de Investigaciones Biológicas Margarita Salas. CSIC. Madrid, Spain. CIBERNED
| | - K.R. Velasco
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - E.A. Schon
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - E. Area-Gomez
- Centro de Investigaciones Biológicas Margarita Salas. CSIC. Madrid, Spain. CIBERNED
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
3
|
Tao Z, Li C, Zhang A, Zhang Z, Huang J, Harnud S. Study of transport, tissue distribution, depletion, and hepatotoxicity of Cyadox, a quinoxaline-1,4-dioxide derivative. Front Pharmacol 2024; 15:1401275. [PMID: 39376613 PMCID: PMC11456426 DOI: 10.3389/fphar.2024.1401275] [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: 03/15/2024] [Accepted: 09/06/2024] [Indexed: 10/09/2024] Open
Abstract
Background Cyadox (CYA) is a derivative of quinoxaline 1,4-dioxide and a safe and effective synthetic antibacterial agent. Objective This study aimed to explore the drug transport in blood, distribution, depletion and hepatotoxicity of drugs in animals. Methods The transport of CYA in blood was studied using fluorescence, circular dichroism (CD) and molecular docking methods. Tissue distribution and depletion of CYA in rats were evaluated following oral administration of [3H]-CYA at different doses. Hepatotoxicity of drugs evaluated by transcriptomics. Results During transport in the bloodstream, the drug binds to bovine serum albumin (BSA) by hydrogen bonding and has only one binding site. Hydrogen bonds were formed between O (2) of CYA and ARG208, O (3) of CYA and LEU480, VAL481. The secondary protein conformation of BSA changed after binding with an increase in α-helix and a decrease in β-strand. After a single oral administration of [3H]-CYA, it was excreted rapidly within 7 days, with 34.81% from the urine and 60.25% from the feces. Higher and sustained levels of radioactivity were detected in the liver during the post-dose period, suggesting that the drug may concentrate in the liver. The transcriptomic data indicates that CYA exhibits low hepatotoxicity. However, there are indications that it may have an impact on steroid biosynthesis. Conclusion This study could serve as a basis for conducting further studies on the use of CYA in food animals and improving the pharmacologic, pharmacokinetic, and toxicologic effects of CYA on food animals.
Collapse
Affiliation(s)
- Zhu Tao
- Research Center for Ecotoxicology and Food Safety, Hubei Engineering University, Xiaogan, China
- College of Life Science and Technology, Hubei Engineering University, Xiaogan, China
- Hubei Province Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hubei Engineering University, Xiaogan, China
- Hubei Key Laboratory of Quality Control of Characteristic Fruits and Vegetables, Hubei Engineering University, Xiaogan, China
| | - Changchun Li
- College of Life Science and Technology, Hubei Engineering University, Xiaogan, China
- Hubei Province Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hubei Engineering University, Xiaogan, China
- Hubei Key Laboratory of Quality Control of Characteristic Fruits and Vegetables, Hubei Engineering University, Xiaogan, China
| | - Aiqun Zhang
- Research Center for Ecotoxicology and Food Safety, Hubei Engineering University, Xiaogan, China
- College of Life Science and Technology, Hubei Engineering University, Xiaogan, China
- Hubei Key Laboratory of Quality Control of Characteristic Fruits and Vegetables, Hubei Engineering University, Xiaogan, China
| | - Zhilin Zhang
- College of Life Science and Technology, Hubei Engineering University, Xiaogan, China
- Hubei Province Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hubei Engineering University, Xiaogan, China
- Hubei Key Laboratory of Quality Control of Characteristic Fruits and Vegetables, Hubei Engineering University, Xiaogan, China
| | - Jing Huang
- College of Life Science and Technology, Hubei Engineering University, Xiaogan, China
- Hubei Province Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hubei Engineering University, Xiaogan, China
- Hubei Key Laboratory of Quality Control of Characteristic Fruits and Vegetables, Hubei Engineering University, Xiaogan, China
| | - Sechenchogt Harnud
- Research Center for Ecotoxicology and Food Safety, Hubei Engineering University, Xiaogan, China
- College of Life Science and Technology, Hubei Engineering University, Xiaogan, China
- Hubei Province Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hubei Engineering University, Xiaogan, China
- Hubei Key Laboratory of Quality Control of Characteristic Fruits and Vegetables, Hubei Engineering University, Xiaogan, China
| |
Collapse
|
4
|
Yu T, Hou Z, Wang H, Chang S, Song X, Zheng W, Zheng L, Wei J, Lu Z, Chen J, Zhou Y, Chen M, Sun S, Jiang Q, Jin L, Ma Y, Xu Z. Soybean steroids improve crop abiotic stress tolerance and increase yield. PLANT BIOTECHNOLOGY JOURNAL 2024; 22:2333-2347. [PMID: 38600703 PMCID: PMC11258977 DOI: 10.1111/pbi.14349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 02/20/2024] [Accepted: 03/20/2024] [Indexed: 04/12/2024]
Abstract
Sterols have long been associated with diverse fields, such as cancer treatment, drug development, and plant growth; however, their underlying mechanisms and functions remain enigmatic. Here, we unveil a critical role played by a GmNF-YC9-mediated CCAAT-box transcription complex in modulating the steroid metabolism pathway within soybeans. Specifically, this complex directly activates squalene monooxygenase (GmSQE1), which is a rate-limiting enzyme in steroid synthesis. Our findings demonstrate that overexpression of either GmNF-YC9 or GmSQE1 significantly enhances soybean stress tolerance, while the inhibition of SQE weakens this tolerance. Field experiments conducted over two seasons further reveal increased yields per plant in both GmNF-YC9 and GmSQE1 overexpressing plants under drought stress conditions. This enhanced stress tolerance is attributed to the reduction of abiotic stress-induced cell oxidative damage. Transcriptome and metabolome analyses shed light on the upregulation of multiple sterol compounds, including fucosterol and soyasaponin II, in GmNF-YC9 and GmSQE1 overexpressing soybean plants under stress conditions. Intriguingly, the application of soybean steroids, including fucosterol and soyasaponin II, significantly improves drought tolerance in soybean, wheat, foxtail millet, and maize. These findings underscore the pivotal role of soybean steroids in countering oxidative stress in plants and offer a new research strategy for enhancing crop stress tolerance and quality from gene regulation to chemical intervention.
Collapse
Affiliation(s)
- Tai‐Fei Yu
- State Key Laboratory of Crop Gene Resources and BreedingInstitute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS)BeijingChina
| | - Ze‐Hao Hou
- State Key Laboratory of Crop Gene Resources and BreedingInstitute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS)BeijingChina
| | - Hai‐Long Wang
- Beijing Key Laboratory of Agricultural Genetic Resources and BiotechnologyInstitute of Biotechnology, Beijing Academy of Agriculture and Forestry SciencesBeijingChina
| | - Shi‐Yang Chang
- Department of Histology and EmbryologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Xin‐Yuan Song
- Agro‐biotechnology Research InstituteJilin Academy of Agriculture SciencesChangchunChina
| | - Wei‐Jun Zheng
- State Key Laboratory of Crop Stress Biology for Arid Areas/Northwest Agricultural and Forestry UniversityYanglingChina
| | - Lei Zheng
- State Key Laboratory of Crop Gene Resources and BreedingInstitute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS)BeijingChina
| | - Ji‐Tong Wei
- State Key Laboratory of Crop Gene Resources and BreedingInstitute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS)BeijingChina
| | - Zhi‐Wei Lu
- State Key Laboratory of Crop Gene Resources and BreedingInstitute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS)BeijingChina
| | - Jun Chen
- State Key Laboratory of Crop Gene Resources and BreedingInstitute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS)BeijingChina
| | - Yong‐Bin Zhou
- State Key Laboratory of Crop Gene Resources and BreedingInstitute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS)BeijingChina
| | - Ming Chen
- State Key Laboratory of Crop Gene Resources and BreedingInstitute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS)BeijingChina
| | - Su‐Li Sun
- State Key Laboratory of Crop Gene Resources and BreedingInstitute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS)BeijingChina
| | - Qi‐Yan Jiang
- State Key Laboratory of Crop Gene Resources and BreedingInstitute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS)BeijingChina
- College of Agronomy/College of Life SciencesJilin Agricultural UniversityChangchunChina
| | - Long‐Guo Jin
- State Key Laboratory of Crop Gene Resources and BreedingInstitute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS)BeijingChina
| | - You‐Zhi Ma
- State Key Laboratory of Crop Gene Resources and BreedingInstitute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS)BeijingChina
- College of Agronomy/College of Life SciencesJilin Agricultural UniversityChangchunChina
| | - Zhao‐Shi Xu
- State Key Laboratory of Crop Gene Resources and BreedingInstitute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS)BeijingChina
- College of Agronomy/College of Life SciencesJilin Agricultural UniversityChangchunChina
- National Nanfan Research Institute (Sanya)Chinese Academy of Agricultural Sciences / Seed Industry LaboratorySanyaChina
| |
Collapse
|
5
|
Zheng Y, Lu Q, Cao J, Liu Y, Liu H, Jin J, Zhang Z, Yang Y, Zhu X, Han D, Xie S. Supplementation of Mangiferin to a High-Starch Diet Alleviates Hepatic Injury and Lipid Accumulation Potentially through Modulating Cholesterol Metabolism in Channel Catfish ( Ictalurus punctatus). Antioxidants (Basel) 2024; 13:722. [PMID: 38929161 PMCID: PMC11200457 DOI: 10.3390/antiox13060722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Starch is a common source of carbohydrates in aqua feed. High-starch diet can cause hepatic injury and lipid accumulation in fish. Mangiferin (MGF) can regulate lipid metabolism and protect the liver, but there is limited research on its effects in fish. In the present study, we investigated whether MGF could ameliorate high-starch-induced hepatic damage and lipid accumulation in channel catfish. The channel catfish (Ictalurus punctatus) were fed one of four experimental diets for eight weeks: a control diet (NCD), a high-starch diet (HCD), an HCD supplemented with 100 mg/kg MGF (100 MGF), and an HCD supplemented with 500 mg/kg MGF (500 MGF). The results demonstrated that the weight gain rate (WGR) (p = 0.031), specific growth rate (SGR) (p = 0.039), and feed conversion efficiency (FCE) (p = 0.040) of the 500 MGF group were significantly higher than those of the NCD group. MGF supplementation alleviated liver damage and improved antioxidant capacity (T-AOC) compared to those of the HCD group (p = 0.000). In addition, dietary MGF significantly reduced plasma glucose (GLU) (p = 0.000), triglyceride (TG) (p= 0.001), and low-density lipoprotein cholesterol (LDL) (p = 0.000) levels. It is noteworthy that MGF significantly reduced the plasma total cholesterol (TC) levels (p = 0.000) and liver TC levels (p = 0.005) of channel catfish. Dietary MGF improves cholesterol homeostasis by decreasing the expression of genes that are involved in cholesterol synthesis and transport (hmgcr, sqle, srebf2, sp1, and ldlr) and increasing the expression of genes that are involved in cholesterol catabolism (cyp7a1). Among them, the largest fold decrease in squalene epoxidase (sqle) expression levels was observed in the 100 MGF or 500 MGF groups compared with the HCD group, with a significant decrease of 3.64-fold or 2.20-fold (p = 0.008). And the 100 MGF or 500 MGF group had significantly decreased (by 1.67-fold or 1.94-fold) Sqle protein levels compared to those of the HCD group (p = 0.000). In primary channel catfish hepatocytes, MGF significantly down-regulated the expression of sqle (p = 0.030) and reduced cholesterol levels (p = 0.000). In NCTC 1469 cells, MGF significantly down-regulated the expression of sqle (p = 0.000) and reduced cholesterol levels (p = 0.024). In conclusion, MGF effectively inhibits sqle expression and reduces cholesterol accumulation. The current study shows how MGF supplementation regulates the metabolism and accumulation of cholesterol in channel catfish, providing a theoretical basis for the use of MGF as a dietary supplement in aquaculture.
Collapse
Affiliation(s)
- Yutong Zheng
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China;
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Q.L.); (J.C.); (Y.L.); (H.L.); (J.J.); (Z.Z.); (Y.Y.); (X.Z.); (S.X.)
| | - Qisheng Lu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Q.L.); (J.C.); (Y.L.); (H.L.); (J.J.); (Z.Z.); (Y.Y.); (X.Z.); (S.X.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingyue Cao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Q.L.); (J.C.); (Y.L.); (H.L.); (J.J.); (Z.Z.); (Y.Y.); (X.Z.); (S.X.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yulong Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Q.L.); (J.C.); (Y.L.); (H.L.); (J.J.); (Z.Z.); (Y.Y.); (X.Z.); (S.X.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haokun Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Q.L.); (J.C.); (Y.L.); (H.L.); (J.J.); (Z.Z.); (Y.Y.); (X.Z.); (S.X.)
| | - Junyan Jin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Q.L.); (J.C.); (Y.L.); (H.L.); (J.J.); (Z.Z.); (Y.Y.); (X.Z.); (S.X.)
| | - Zhimin Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Q.L.); (J.C.); (Y.L.); (H.L.); (J.J.); (Z.Z.); (Y.Y.); (X.Z.); (S.X.)
| | - Yunxia Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Q.L.); (J.C.); (Y.L.); (H.L.); (J.J.); (Z.Z.); (Y.Y.); (X.Z.); (S.X.)
| | - Xiaoming Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Q.L.); (J.C.); (Y.L.); (H.L.); (J.J.); (Z.Z.); (Y.Y.); (X.Z.); (S.X.)
| | - Dong Han
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Q.L.); (J.C.); (Y.L.); (H.L.); (J.J.); (Z.Z.); (Y.Y.); (X.Z.); (S.X.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Shouqi Xie
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; (Q.L.); (J.C.); (Y.L.); (H.L.); (J.J.); (Z.Z.); (Y.Y.); (X.Z.); (S.X.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China
| |
Collapse
|
6
|
Zhang L, Cao Z, Hong Y, He H, Chen L, Yu Z, Gao Y. Squalene Epoxidase: Its Regulations and Links with Cancers. Int J Mol Sci 2024; 25:3874. [PMID: 38612682 PMCID: PMC11011400 DOI: 10.3390/ijms25073874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/09/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
Squalene epoxidase (SQLE) is a key enzyme in the mevalonate-cholesterol pathway that plays a critical role in cellular physiological processes. It converts squalene to 2,3-epoxysqualene and catalyzes the first oxygenation step in the pathway. Recently, intensive efforts have been made to extend the current knowledge of SQLE in cancers through functional and mechanistic studies. However, the underlying mechanisms and the role of SQLE in cancers have not been fully elucidated yet. In this review, we retrospected current knowledge of SQLE as a rate-limiting enzyme in the mevalonate-cholesterol pathway, while shedding light on its potential as a diagnostic and prognostic marker, and revealed its therapeutic values in cancers. We showed that SQLE is regulated at different levels and is involved in the crosstalk with iron-dependent cell death. Particularly, we systemically reviewed the research findings on the role of SQLE in different cancers. Finally, we discussed the therapeutic implications of SQLE inhibitors and summarized their potential clinical values. Overall, this review discussed the multifaceted mechanisms that involve SQLE to present a vivid panorama of SQLE in cancers.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Zheng Cao
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yuheng Hong
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Haihua He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Leifeng Chen
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhentao Yu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Yibo Gao
- Central Laboratory & Shenzhen Key Laboratory of Epigenetics and Precision Medicine for Cancers, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
- State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Laboratory of Translational Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
7
|
Wang T, Gong M, Lu Y, Zhao C, Ling L, Chen J, Ju R. Oxysterol 25-hydroxycholesterol activation of ferritinophagy inhibits the development of squamous intraepithelial lesion of cervix in HPV-positive patients. Cell Death Discov 2024; 10:135. [PMID: 38472192 DOI: 10.1038/s41420-024-01899-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Squamous intraepithelial lesion of cervix (SIL) in human papillomavirus (HPV)-positive patient often undergoes a silent and long-course development, and most of them with high-grade transit to cervical squamous cell carcinoma (CSCC). The oxysterol 25-hydroxycholesterol (25-HC) is associated with HPV inhibition, autophagy and cholesterol synthesis, however, its function in this long process of SIL development remain unclear. In this study, we demonstrate that 25-HC generation is inhibited through HSIL-to-CSCC transition. The 25-HC activates ferritinophagy in the early stage of SIL, promoting the vulnerability of HSILs to ferroptosis. Therefore, maintaining 25-HC level is crucial for suppressing HSIL progression and holds promise for developing novel clinical therapies for CSCC.
Collapse
Affiliation(s)
- Tianming Wang
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Min Gong
- Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yingfei Lu
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chengcheng Zhao
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ling Ling
- Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianquan Chen
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Rong Ju
- Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
8
|
Wu J, Hu W, Yang W, Long Y, Chen K, Li F, Ma X, Li X. Knockdown of SQLE promotes CD8+ T cell infiltration in the tumor microenvironment. Cell Signal 2024; 114:110983. [PMID: 37993027 DOI: 10.1016/j.cellsig.2023.110983] [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: 08/30/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/24/2023]
Abstract
Cholesterol biosynthesis and metabolism are critical aspects that shape the process of tumor development and associated microenvironmental conditions owing to the ability of cholesterol to drive tumor growth and invasion. Squalene Epoxidase (SQLE) is the second rate-limiting enzyme involved in the synthesis of cholesterol. The functional role of SQLE within the tumor microenvironment, however, has yet to be established. Here we show that SQLE is distinctively expressed across most types of cancer, and the expression level is highly correlated with tumor mutation burden and microsatellite instability. Accordingly, SQLE was identified as a prognostic risk factor in cancer patients. In addition, we observed a negative correlation between SQLE expression and immune cell infiltration across multiple cancers, and murine xenograft model further confirmed that SQLE knockdown was associated with enhanced intratumoral CD8+ T cell infiltration. Using next-generation sequencing, we identified 410 genes distinctively expressed in tumors exhibiting SQLE inhibition. KEGG and GO analysis further verified that SQLE altered the immune response in the tumor microenvironment. Furthermore, we found that the metabolism and translation of proteins is the main binding factor with SQLE. Our findings ascertain that SQLE is a potential target in multiple cancers and suppressing SQLE establishes an essential mechanism for shaping tumor microenvironment.
Collapse
Affiliation(s)
- Jun Wu
- Key Laboratory of Brain, Cognition and Education Sciences, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, South China Normal University, Guangzhou 510631, China
| | - Weibin Hu
- Key Laboratory of Brain, Cognition and Education Sciences, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, South China Normal University, Guangzhou 510631, China
| | - Wenhui Yang
- Key Laboratory of Brain, Cognition and Education Sciences, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, South China Normal University, Guangzhou 510631, China
| | - Yihao Long
- Key Laboratory of Brain, Cognition and Education Sciences, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, South China Normal University, Guangzhou 510631, China
| | - Kaizhao Chen
- Key Laboratory of Brain, Cognition and Education Sciences, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, South China Normal University, Guangzhou 510631, China
| | - Fugui Li
- Cancer Research Institute of Zhongshan City, Zhongshan City People's Hospital, Zhongshan 528403, China
| | - Xiaodong Ma
- Key Laboratory of Brain, Cognition and Education Sciences, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education, South China Normal University, Guangzhou 510631, China.
| | - Xun Li
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
9
|
Kiss RS, Chicoine J, Khalil Y, Sladek R, Chen H, Pisaturo A, Martin C, Dale JD, Brudenell TA, Kamath A, Kyei-Boahen J, Hafiane A, Daliah G, Alecki C, Hopes TS, Heier M, Aligianis IA, Lebrun JJ, Aspden J, Paci E, Kerksiek A, Lütjohann D, Clayton P, Wills JC, von Kriegsheim A, Nilsson T, Sheridan E, Handley MT. Comparative proximity biotinylation implicates the small GTPase RAB18 in sterol mobilization and biosynthesis. J Biol Chem 2023; 299:105295. [PMID: 37774976 PMCID: PMC10641524 DOI: 10.1016/j.jbc.2023.105295] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 10/01/2023] Open
Abstract
Loss of functional RAB18 causes the autosomal recessive condition Warburg Micro syndrome. To better understand this disease, we used proximity biotinylation to generate an inventory of potential RAB18 effectors. A restricted set of 28 RAB18 interactions were dependent on the binary RAB3GAP1-RAB3GAP2 RAB18-guanine nucleotide exchange factor complex. Twelve of these 28 interactions are supported by prior reports, and we have directly validated novel interactions with SEC22A, TMCO4, and INPP5B. Consistent with a role for RAB18 in regulating membrane contact sites, interactors included groups of microtubule/membrane-remodeling proteins, membrane-tethering and docking proteins, and lipid-modifying/transporting proteins. Two of the putative interactors, EBP and OSBPL2/ORP2, have sterol substrates. EBP is a Δ8-Δ7 sterol isomerase, and ORP2 is a lipid transport protein. This prompted us to investigate a role for RAB18 in cholesterol biosynthesis. We found that the cholesterol precursor and EBP-product lathosterol accumulates in both RAB18-null HeLa cells and RAB3GAP1-null fibroblasts derived from an affected individual. Furthermore, de novo cholesterol biosynthesis is impaired in cells in which RAB18 is absent or dysregulated or in which ORP2 expression is disrupted. Our data demonstrate that guanine nucleotide exchange factor-dependent Rab interactions are highly amenable to interrogation by proximity biotinylation and may suggest that Micro syndrome is a cholesterol biosynthesis disorder.
Collapse
Affiliation(s)
- Robert S Kiss
- Cardiovascular Health Across the Lifespan (CHAL) Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | - Jarred Chicoine
- Metabolic Disorders and Complications (MEDIC) Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Youssef Khalil
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Robert Sladek
- Metabolic Disorders and Complications (MEDIC) Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - He Chen
- Cardiovascular Health Across the Lifespan (CHAL) Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Alessandro Pisaturo
- Cardiovascular Health Across the Lifespan (CHAL) Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Cyril Martin
- Cardiovascular Health Across the Lifespan (CHAL) Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Jessica D Dale
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, United Kingdom
| | - Tegan A Brudenell
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, United Kingdom
| | - Archith Kamath
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom; Division of Medical Sciences, University of Oxford, Oxford, United Kingdom
| | - Jeffrey Kyei-Boahen
- Department of Medicine, McGill University Health Centre, CHAL Research Program, Montreal, Canada
| | - Anouar Hafiane
- Department of Medicine, McGill University Health Centre, CHAL Research Program, Montreal, Canada
| | - Girija Daliah
- Department of Medicine, McGill University Health Centre, Cancer Research Program, Montreal, Canada
| | - Célia Alecki
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Tayah S Hopes
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Martin Heier
- Department of Clinical Neuroscience for Children, Oslo University Hospital, Oslo, Norway
| | - Irene A Aligianis
- Medical and Developmental Genetics, Medical Research Council Human Genetics Unit, Edinburgh, United Kingdom
| | - Jean-Jacques Lebrun
- Department of Medicine, McGill University Health Centre, Cancer Research Program, Montreal, Canada
| | - Julie Aspden
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Emanuele Paci
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Anja Kerksiek
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Peter Clayton
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Jimi C Wills
- Cancer Research United Kingdom Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom; Firefinch Software Ltd, Edinburgh, United Kingdom
| | - Alex von Kriegsheim
- Cancer Research United Kingdom Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Tommy Nilsson
- Cancer Research Program (CRP), Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Eamonn Sheridan
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, United Kingdom
| | - Mark T Handley
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, United Kingdom; Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|
10
|
Iyer DR, Venkatraman J, Tanguy E, Vitale N, Mahapatra NR. Chromogranin A and its derived peptides: potential regulators of cholesterol homeostasis. Cell Mol Life Sci 2023; 80:271. [PMID: 37642733 PMCID: PMC11072126 DOI: 10.1007/s00018-023-04908-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/31/2023]
Abstract
Chromogranin A (CHGA), a member of the granin family of proteins, has been an attractive therapeutic target and candidate biomarker for several cardiovascular, neurological, and inflammatory disorders. The prominence of CHGA stems from the pleiotropic roles of several bioactive peptides (e.g., catestatin, pancreastatin, vasostatins) generated by its proteolytic cleavage and by their wide anatomical distribution. These peptides are emerging as novel modulators of cardiometabolic diseases that are often linked to high blood cholesterol levels. However, their impact on cholesterol homeostasis is poorly understood. The dynamic nature of cholesterol and its multitudinous roles in almost every aspect of normal body function makes it an integral component of metabolic physiology. A tightly regulated coordination of cholesterol homeostasis is imperative for proper functioning of cellular and metabolic processes. The deregulation of cholesterol levels can result in several pathophysiological states. Although studies till date suggest regulatory roles for CHGA and its derived peptides on cholesterol levels, the mechanisms by which this is achieved still remain unclear. This review aims to aggregate and consolidate the available evidence linking CHGA with cholesterol homeostasis in health and disease. In addition, we also look at common molecular regulatory factors (viz., transcription factors and microRNAs) which could govern the expression of CHGA and genes involved in cholesterol homeostasis under basal and pathological conditions. In order to gain further insights into the pathways mediating cholesterol regulation by CHGA/its derived peptides, a few prospective signaling pathways are explored, which could act as primers for future studies.
Collapse
Affiliation(s)
- Dhanya R Iyer
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Janani Venkatraman
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Emeline Tanguy
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212 and Université de Strasbourg, 5 Rue Blaise Pascal, 67000, Strasbourg, France
| | - Nicolas Vitale
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212 and Université de Strasbourg, 5 Rue Blaise Pascal, 67000, Strasbourg, France.
| | - Nitish R Mahapatra
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India.
| |
Collapse
|
11
|
Weng N, Zhang Z, Tan Y, Zhang X, Wei X, Zhu Q. Repurposing antifungal drugs for cancer therapy. J Adv Res 2023; 48:259-273. [PMID: 36067975 PMCID: PMC10248799 DOI: 10.1016/j.jare.2022.08.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 08/29/2022] [Accepted: 08/29/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Repurposing antifungal drugs in cancer therapy has attracted unprecedented attention in both preclinical and clinical research due to specific advantages, such as safety, high-cost effectiveness and time savings compared with cancer drug discovery. The surprising and encouraging efficacy of antifungal drugs in cancer therapy, mechanistically, is attributed to the overlapping targets or molecular pathways between fungal and cancer pathogenesis. Advancements in omics, informatics and analytical technology have led to the discovery of increasing "off-site" targets from antifungal drugs involved in cancerogenesis, such as smoothened (D477G) inhibition from itraconazole in basal cell carcinoma. AIM OF REVIEW This review illustrates several antifungal drugs repurposed for cancer therapy and reveals the underlying mechanism based on their original target and "off-site" target. Furthermore, the challenges and perspectives for the future development and clinical applications of antifungal drugs for cancer therapy are also discussed, providing a refresh understanding of drug repurposing. KEY SCIENTIFIC CONCEPTS OF REVIEW This review may provide a basic understanding of repurposed antifungal drugs for clinical cancer management, thereby helping antifungal drugs broaden new indications and promote clinical translation.
Collapse
Affiliation(s)
- Ningna Weng
- Department of Abdominal Oncology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Medical Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fujian 350011, PR China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu, China; Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yunhan Tan
- West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xiaoyue Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Zhu
- Department of Abdominal Oncology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
12
|
Zhang Z, Wu W, Jiao H, Chen Y, Ji X, Cao J, Yin F, Yin W. Squalene epoxidase promotes hepatocellular carcinoma development by activating STRAP transcription and TGF-β/SMAD signalling. Br J Pharmacol 2022; 180:1562-1581. [PMID: 36581319 DOI: 10.1111/bph.16024] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 08/20/2022] [Accepted: 09/11/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Squalene epoxidase (SQLE) is a key enzyme involved in cholesterol biosynthesis, but growing evidence also reveals that SQLE is abnormally expressed in some types of malignant tumours, even though the underlying mechanism remains poorly understood. EXPERIMENTAL APPROACH Bioinformatics analysis and RNA sequencing were applied to detect differentially expressed genes in clinical hepatocellular carcinoma (HCC). MTT, colony formation, AnnexinV-FITC/PI, EdU, wound healing, transwell, western blot, qRT-PCR, IHC, F-actin, RNA-sequencing, dual-luciferase reporters, and H&E staining were used to investigate the pharmacological effects and possible mechanisms of SQLE. KEY RESULTS SQLE expression was specifically elevated in HCC, correlating with poor clinical outcomes. SQLE significantly promoted HCC growth, epithelial-mesenchymal transition, and metastasis both in vitro and in vivo. RNA sequencing and functional experiments revealed that the protumourigenic effect of SQLE on HCC was closely related to the activation of TGF-β/SMAD signalling, but the stimulatory effect of SQLE on TGF-β/SMAD signalling and HCC development is critically dependent on STRAP. SQLE expression is well correlated with STRAP in HCC, and further, to amplify TGF-β/SMAD signalling, SQLE even transcriptionally increased STRAP gene expression mediated by AP-2α. Finally, as a chemical inhibitor of SQLE, NB-598 markedly inhibited HCC cell growth and tumour development. CONCLUSIONS AND IMPLICATIONS Taken together, SQLE serves as a novel oncogene in HCC development by activating TGF-β/SMAD signalling. Targeting SQLE could be useful in drug development and therapy for HCC.
Collapse
Affiliation(s)
- Zhirui Zhang
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| | - Wei Wu
- Organ Transplantation Center, Southern District, the First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Hospital), Hefei, China
| | - Hao Jiao
- Department of Pharmacy, Fuyang People's Hospital, Fuyang, China
| | - Yuzhong Chen
- Department of Surgical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xiaojun Ji
- Department of Innovation, Nanjing Chia Tai Tianqing Pharmaceutical Co., Ltd, Nanjing, China
| | - Jing Cao
- Department of Pharmacy, Women's Hospital of Nanjing Medical University/Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Fangzhou Yin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wu Yin
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
13
|
Zou Y, Zhang H, Bi F, Tang Q, Xu H. Targeting the key cholesterol biosynthesis enzyme squalene monooxygenasefor cancer therapy. Front Oncol 2022; 12:938502. [PMID: 36091156 PMCID: PMC9449579 DOI: 10.3389/fonc.2022.938502] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/26/2022] [Indexed: 11/22/2022] Open
Abstract
Cholesterol metabolism is often dysregulated in cancer. Squalene monooxygenase (SQLE) is the second rate-limiting enzyme involved in cholesterol synthesis. Since the discovery of SQLE dysregulation in cancer, compelling evidence has indicated that SQLE plays a vital role in cancer initiation and progression and is a promising therapeutic target for cancer treatment. In this review, we provide an overview of the role and regulation of SQLE in cancer and summarize the updates of antitumor therapy targeting SQLE.
Collapse
Affiliation(s)
- Yuheng Zou
- Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Hongying Zhang
- Laboratory of Oncogene, West China Hospital, Sichuan University, Chengdu, China
| | - Feng Bi
- Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiulin Tang
- Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Qiulin Tang, ; Huanji Xu,
| | - Huanji Xu
- Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Qiulin Tang, ; Huanji Xu,
| |
Collapse
|
14
|
Shi L, Qian Y, Shen Q, He Y, Jia Y, Wang F. The developmental toxicity and transcriptome analyses of zebrafish (Danio rerio) embryos exposed to carbon nanoparticles. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 234:113417. [PMID: 35304337 DOI: 10.1016/j.ecoenv.2022.113417] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 06/14/2023]
Abstract
Long-term and short-term exposure to carbon nanoparticles (CNPs) can affect fetal development and subsequent adverse outcomes including preterm delivery, intrauterine growth restriction, low birth weight, increased health risk linked to cardiovascular, respiratory and nervous systems in adulthood. The adverse developmental outcomes of CNPs were well known, but the underlying mechanisms remain unresolved. In this study, zebrafish embryos were treated with CNPs of 50,100,200 μg/mL and the toxic effects were observed. Using the RNA-seq analysis approach, we examined the effects of CNPs (200 μg/mL) on gene expression in zebrafish embryos exposed from 4 to 96 h-post-fertilization (hpf). We observed that CNPs-treated embryos exhibited increased malformations and decreased hatching. A total of 236 differentially expressed genes were detected by transcriptome analyses, which were associated with phototransduction, amino acid metabolism, steroid and steroid hormone biosynthesis. Transcriptome results were verified by real-time fluorescence quantitative PCR (RT-qPCR). Our results indicated that CNPs exposure was most likely to lead to differential gene changes in steroid and hormone biosynthesis pathways, thus inducing developmental toxicity such as delayed incubation of zebrafish embryos, increased malformation rate and multiple malformation phenotypes.
Collapse
Affiliation(s)
- Leilei Shi
- School of Public Health, Baotou Medical College, Baotou, Inner Mongolia 014040, PR China
| | - Yaru Qian
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 10012, PR China
| | - Qian Shen
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 10012, PR China
| | - Yanan He
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 10012, PR China
| | - Yuqiao Jia
- School of Public Health, Baotou Medical College, Baotou, Inner Mongolia 014040, PR China.
| | - Feifei Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 10012, PR China.
| |
Collapse
|
15
|
Shi H, Wang H, Zhang C, Lu Y, Yao J, Chen Z, Xing G, Wei Q, Cao X. Mutations in OSBPL2 cause hearing loss associated with primary cilia defects via Sonic Hedgehog signaling. JCI Insight 2022; 7:149626. [PMID: 35041619 PMCID: PMC8876550 DOI: 10.1172/jci.insight.149626] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 01/12/2022] [Indexed: 11/17/2022] Open
Abstract
Defective primary cilia cause a range of diseases called ciliopathies, which include hearing loss (HL). Variants in the human oxysterol-binding protein like 2 (OSBPL2/ORP2) are responsible for autosomal dominant nonsyndromic HL (DFNA67). However, the pathogenesis of OSBPL2 deficiency has not been fully elucidated. In this study, we show that the Osbpl2-KO mice exhibited progressive HL and abnormal cochlear development with defective cilia. Further research revealed that OSBPL2 was located at the base of the kinocilia in hair cells (HCs) and primary cilia in supporting cells (SCs) and functioned in the maintenance of ciliogenesis by regulating the homeostasis of PI(4,5)P2 (phosphatidylinositol 4,5-bisphosphate) on the cilia membrane. OSBPL2 deficiency led to a significant increase of PI(4,5)P2 on the cilia membrane, which could be partially rescued by the overexpression of INPP5E. In addition, smoothened and GL13, the key molecules in the Sonic Hedgehog (Shh) signaling pathway, were detected to be downregulated in Osbpl2-KO HEI-OC1 cells. Our findings revealed that OSBPL2 deficiency resulted in ciliary defects and abnormal Shh signaling transduction in auditory cells, which helped to elucidate the underlying mechanism of OSBPL2 deficiency in HL.
Collapse
Affiliation(s)
- Hairong Shi
- Department of Medical Genetics, Nanjing Medical Univeristy, Nanjing, China
| | - Hongshun Wang
- Department of Medical Genetics, Nanjing Medical Univeristy, Nanjing, China
| | - Cheng Zhang
- Department of Medical Genetics, Nanjing Medical Univeristy, Nanjing, China
| | - Yajie Lu
- Department of Medical Genetics, Nanjing Medical Univeristy, Nanjing, China
| | - Jun Yao
- Department of Medical Genetics, Nanjing Medical Univeristy, Nanjing, China
| | - Zhibin Chen
- Department of Otolaryngology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guangqian Xing
- Department of Otolaryngology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qinjun Wei
- Department of Medical Genetics, Nanjing Medical Univeristy, Nanjing, China
| | - Xin Cao
- Department of Medical Genetics, Nanjing Medical Univeristy, Nanjing, China
| |
Collapse
|
16
|
Zhou F, Sun X. Cholesterol Metabolism: A Double-Edged Sword in Hepatocellular Carcinoma. Front Cell Dev Biol 2021; 9:762828. [PMID: 34869352 PMCID: PMC8635701 DOI: 10.3389/fcell.2021.762828] [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: 08/22/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) represents a leading cause of cancer-related deaths globally. The rising incidence of metabolic syndrome and its hepatic manifestation, nonalcoholic fatty liver disease (NAFLD), have emerged as the fastest-growing cause of HCC in recent years. Cholesterol, a major lipid component of the cell membrane and lipoprotein particles, is primarily produced and metabolized by the liver. Numerous studies have revealed an increased cholesterol biosynthesis and uptake, reduced cholesterol exportation and excretion in HCC, which all contribute to lipotoxicity, inflammation, and fibrosis, known HCC risk factors. In contrast, some clinical studies have shown that higher cholesterol is associated with a reduced risk of HCC. These contradictory observations imply that the relationship between cholesterol and HCC is far more complex than initially anticipated. Understanding the role of cholesterol and deciphering the underlying molecular events in HCC development is highly relevant to developing new therapies. Here, we discuss the current understanding of cholesterol metabolism in the pathogenesis of NAFLD-associated HCC, and the underlying mechanisms, including the roles of cholesterol in the disruption of normal function of specific cell types and signaling transduction. We also review the clinical progression in evaluating the association of cholesterol with HCC. The therapeutic effects of lowering cholesterol will also be summarized. We also interpret reasons for the contradictory observations from different preclinical and human studies of the roles of cholesterol in HCC, aiming to provide a critical assessment of the potential of cholesterol as a therapeutic target.
Collapse
Affiliation(s)
- Fangli Zhou
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoli Sun
- Department of Pharmacology, Mays Cancer Center, Transplant Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
17
|
Liu Y, Fang L, Liu W. High SQLE Expression and Gene Amplification Correlates with Poor Prognosis in Head and Neck Squamous Cell Carcinoma. Cancer Manag Res 2021; 13:4709-4723. [PMID: 34163246 PMCID: PMC8213972 DOI: 10.2147/cmar.s305719] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/18/2021] [Indexed: 12/24/2022] Open
Abstract
Objective Squalene epoxidase (SQLE) is considered a metabolic oncogene, but its biological function and prognostic value in head and neck squamous cell carcinoma (HNSCC) remain unclear. We aimed to evaluate the role of SQLE in the occurrence and development of HNSCC through bioinformatics analysis, and validation experiments. Methods Transcriptomic, genomic, and clinical data from The Cancer Genome Atlas were used for pan-cancer analysis. SQLE expression in HNSCC was evaluated using Gene Expression Omnibus datasets and immunohistochemistry. The biological significance of SQLE in the tumor microenvironment (TME) of HNSCC was determined using TISCH, HuRI, LinkedOmics, and TIMER 2.0. The prognostic value of SQLE in HNSCC was analyzed using univariate Cox regression and Kaplan–Meier survival curves. Effect of SQLE on the Cal27 HNSCC cell line was evaluated using cell counting kit 8, wound healing, and EdU assays. Results SQLE was overexpressed and amplified in various cancers, including HNSCC. High SQLE expression promoted cell proliferation, associated with T stage in HNSCC patients. Copy number amplification and DNA demethylation contributed to high SQLE expression in HNSCC, which was associated with poor prognosis. SQLE was related to HNSCC TME, and its mRNA expression/copy number alterations were negatively correlated with the infiltration of CD8+ T cells, follicular helper T cells, and regulatory T cell infiltration and mast cell activation and positively correlated with the infiltration of M0 macrophages and resting mast cells in HNSCC. Conclusion SQLE was identified as a prognostic biomarker and a potential pharmaceutical target for HNSCC.
Collapse
Affiliation(s)
- Yang Liu
- Department of Oral Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, People's Republic of China
| | - Lijun Fang
- Department of Oral Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, People's Republic of China
| | - Weixian Liu
- Department of Oral Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, People's Republic of China
| |
Collapse
|
18
|
Wang T, Zhang T, Tang Y, Wang H, Wei Q, Lu Y, Yao J, Qu Y, Cao X. Oxysterol-binding protein-like 2 contributes to the developmental progression of preadipocytes by binding to β-catenin. Cell Death Discov 2021; 7:109. [PMID: 34001864 PMCID: PMC8129138 DOI: 10.1038/s41420-021-00503-2] [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: 04/10/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
Oxysterol-binding protein-like 2 (OSBPL2), also known as oxysterol-binding protein-related protein (ORP) 2, is a member of lipid transfer protein well-known for its role in regulating cholesterol homeostasis. A recent study reported that OSBPL2/ORP2 localizes to lipid droplets (LDs) and is associated with energy metabolism and obesity. However, the function of OSBPL2/ORP2 in adipocyte differentiation is poorly understood. Here, we report that OSBPL2/ORP2 contributes to the developmental progression of preadipocytes. We found that OSBPL2/ORP2 binds to β-catenin, a key effector in the Wnt signaling pathway that inhibits adipogenesis. This complex plays a role in regulating the protein level of β-catenin only in preadipocytes, not in mature adipocytes. Our data further indicated that OSBPL2/ORP2 mediates the transport of β-catenin into the nucleus and thus regulates target genes related to adipocyte differentiation. Deletion of OSBPL2/ORP2 markedly reduces β-catenin both in the cytoplasm and in the nucleus, promotes preadipocytes maturation, and ultimately leads to obesity-related characteristics. Altogether, we provide novel insight into the function of OSBPL2/ORP2 in the developmental progression of preadipocytes and suggest OSBPL2/ORP2 may be a potential therapeutic target for the treatment of obesity-related diseases.
Collapse
Affiliation(s)
- Tianming Wang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Tianyu Zhang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Youzhi Tang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Hongshun Wang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Qinjun Wei
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Yajie Lu
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Jun Yao
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China
| | - Yuan Qu
- Jiangsu Cancer Hospital, Nanjing Medical University, Nanjing, China
| | - Xin Cao
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing, China. .,Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
19
|
Zheng YQ, Jin MF, Suo GH, Wu YJ, Sun YX, Ni H. Proteomics for Studying the Effects of Ketogenic Diet Against Lithium Chloride/Pilocarpine Induced Epilepsy in Rats. Front Neurosci 2020; 14:562853. [PMID: 33132826 PMCID: PMC7550537 DOI: 10.3389/fnins.2020.562853] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
The ketogenic diet (KD) demonstrates antiepileptogenic and neuroprotective efficacy, but the precise mechanisms are unclear. Here we explored the mechanism through systematic proteomics analysis of the lithium chloride-pilocarpine rat model. Sprague-Dawley rats (postnatal day 21, P21) were randomly divided into control (Ctr), seizure (SE), and KD treatment after seizure (SE + KD) groups. Tandem mass tag (TMT) labeling and liquid chromatography-tandem mass spectroscopy (LC-MS/MS) were utilized to assess changes in protein abundance in the hippocampus. A total of 5,564 proteins were identified, of which 110 showed a significant change in abundance between the SE and Ctr groups (18 upregulated and 92 downregulated), 278 between SE + KD and SE groups (218 upregulated and 60 downregulated), and 180 between Ctr and SE + KD groups (121 upregulated and 59 downregulated) (all p < 0.05). Seventy-nine proteins showing a significant change in abundance between SE and Ctr groups were reciprocally regulated in the SD + KD group compared to the SE group (i.e., the seizure-induced change was reversed by KD). Of these, five (dystrobrevin, centromere protein V, oxysterol-binding protein, tetraspanin-2, and progesterone receptor membrane component 2) were verified by parallel reaction monitoring. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis indicated that proteins of the synaptic vesicle cycle pathway were enriched both among proteins differing in abundance between SE and Ctr groups as well as between SE + KD and SE groups. This comprehensive proteomics analyze of KD-treated epilepsy by quantitative proteomics revealed novel molecular mechanisms of KD antiepileptogenic efficacy and potential treatment targets.
Collapse
Affiliation(s)
- Yu-Qin Zheng
- Division of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China.,Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
| | - Mei-Fang Jin
- Division of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Gui-Hai Suo
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
| | - You-Jia Wu
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
| | - Yu-Xiao Sun
- Division of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Hong Ni
- Division of Brain Science, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
20
|
Koponen A, Pan G, Kivelä AM, Ralko A, Taskinen JH, Arora A, Kosonen R, Kari OK, Ndika J, Ikonen E, Cho W, Yan D, Olkkonen VM. ORP2, a cholesterol transporter, regulates angiogenic signaling in endothelial cells. FASEB J 2020; 34:14671-14694. [DOI: 10.1096/fj.202000202r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 07/22/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Annika Koponen
- Minerva Foundation Institute for Medical ResearchBiomedicum 2U Helsinki Finland
| | - Guoping Pan
- Department of Biology Jinan University Guangzhou China
| | - Annukka M. Kivelä
- Minerva Foundation Institute for Medical ResearchBiomedicum 2U Helsinki Finland
| | - Arthur Ralko
- Department of Chemistry University of Illinois at Chicago Chicago IL USA
| | - Juuso H. Taskinen
- Minerva Foundation Institute for Medical ResearchBiomedicum 2U Helsinki Finland
| | - Amita Arora
- Minerva Foundation Institute for Medical ResearchBiomedicum 2U Helsinki Finland
| | - Riikka Kosonen
- Minerva Foundation Institute for Medical ResearchBiomedicum 2U Helsinki Finland
| | - Otto K. Kari
- Drug Research Program Division of Pharmaceutical Biosciences Faculty of Pharmacy University of Helsinki Helsinki Finland
| | - Joseph Ndika
- Human Microbiome Research Faculty of Medicine University of Helsinki Helsinki Finland
| | - Elina Ikonen
- Minerva Foundation Institute for Medical ResearchBiomedicum 2U Helsinki Finland
- Department of Anatomy Faculty of Medicine University of Helsinki Helsinki Finland
| | - Wonhwa Cho
- Department of Chemistry University of Illinois at Chicago Chicago IL USA
| | - Daoguang Yan
- Department of Biology Jinan University Guangzhou China
| | - Vesa M. Olkkonen
- Minerva Foundation Institute for Medical ResearchBiomedicum 2U Helsinki Finland
- Department of Anatomy Faculty of Medicine University of Helsinki Helsinki Finland
| |
Collapse
|
21
|
Chua NK, Coates HW, Brown AJ. Squalene monooxygenase: a journey to the heart of cholesterol synthesis. Prog Lipid Res 2020; 79:101033. [DOI: 10.1016/j.plipres.2020.101033] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/21/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023]
|
22
|
Wang T, Wei Q, Liang L, Tang X, Yao J, Lu Y, Qu Y, Chen Z, Xing G, Cao X. OSBPL2 Is Required for the Binding of COPB1 to ATGL and the Regulation of Lipid Droplet Lipolysis. iScience 2020; 23:101252. [PMID: 32650117 PMCID: PMC7348002 DOI: 10.1016/j.isci.2020.101252] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/03/2020] [Accepted: 06/04/2020] [Indexed: 12/22/2022] Open
Abstract
The accumulation of giant lipid droplets (LDs) increases the risk of metabolic disorders including obesity and insulin resistance. The lipolysis process involves the activation and transfer of lipase, but the molecular mechanism is not completely understood. The translocation of ATGL, a critical lipolysis lipase, from the ER to the LD surface is mediated by an energy catabolism complex. Oxysterol-binding protein-like 2 (OSBPL2/ORP2) is one of the lipid transfer proteins that regulates intracellular cholesterol homeostasis. A recent study has proven that Osbpl2−/− pigs exhibit hypercholesterolemia and obesity phenotypes with an increase in adipocytes. In this study, we identified that OSBPL2 links the endoplasmic reticulum (ER) with LDs, binds to COPB1, and mediates ATGL transport. We provide important insights into the function of OSBPL2, indicating that it is required for the regulation of lipid droplet lipolysis. LD lipolysis is impaired in OSBPL2/osbpl2b-mutant HepG2 cells and zebrafish OSBPL2 interacts with COPB1, a subunit of the COPI complex located on LDs Altered COPI complexes on LDs may perturb the trafficking of lipolysis lipase ATGL
Collapse
Affiliation(s)
- Tianming Wang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing 211166, China
| | - Qinjun Wei
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; The Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Lihong Liang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing 211166, China
| | - Xujun Tang
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing 211166, China
| | - Jun Yao
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Yajie Lu
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China
| | - Yuan Qu
- Jiangsu Cancer Hospital, Nanjing 210009, China
| | - Zhibin Chen
- Department of Otolaryngology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Guangqian Xing
- Department of Otolaryngology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xin Cao
- Department of Medical Genetics, School of Basic Medical Science, Nanjing Medical University, Nanjing 211166, China; Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing 211166, China; The Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
23
|
Song Z, Lv S, Wu H, Qin L, Cao H, Zhang B, Ren S. Identification of foam cell biomarkers by microarray analysis. BMC Cardiovasc Disord 2020; 20:211. [PMID: 32375652 PMCID: PMC7201525 DOI: 10.1186/s12872-020-01495-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/23/2020] [Indexed: 01/12/2023] Open
Abstract
Background Lipid infiltration and inflammatory response run through the occurrence of atherosclerosis. Differentiation into macrophages and foam cell formation are the key steps of AS. Aim of this study was that the differential gene expression between foam cells and macrophages was analyzed to search the key links of foam cell generation, so as to explore the pathogenesis of atherosclerosis and provide targets for the early screening and prevention of coronary artery disease (CAD). Methods The gene expression profiles of GSE9874 were downloaded from Gene Expression Omnibus (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE9874) on GPL96 [HG-U133A] Affymetrix Human Genome U133. A total of 22,383 genes were analyzed for differentially expression genes (DEGs) by Bayes package. GO enrichment analysis and KEGG pathway analysis for DEGs were performed using KOBAS 3.0 software (Peking University, Beijing, China). STRING software (STRING 10.0; European Molecular Biology Laboratory, Heidelberg, Germany) was used to analyze the protein-protein interaction (PPI) of DEGs. Results A total of 167 DEGs between macrophages and foam cells were identified. Compared with macrophages, 102 genes were significantly upregulated and 65 genes were significantly downregulated (P < 0.01, fold-change > 1) in foam cells. DEGs were mainly enrich in ‘sterol biosynthetic and metabolic process’, ‘cholesterol metabolic and biosynthetic process’ by GO enrichment analysis. The results of KEGG pathway analysis showed all differential genes are involved in biological processes through 143 KEGG pathways. A PPI network of the DEGs was constructed and 10 outstanding genes of the PPI network was identified by using Cytoscape, which include HMGCR, SREBF2, LDLR, HMGCS1, FDFT1, LPL, DHCR24, SQLE, ABCA1 and FDPS. Conclusion: Lipid metabolism related genes and molecular pathways were the key to the transformation of macrophages into foam cells. Therefore, lipid metabolism disorder is the key to turn macrophages into foam cells, which plays a major role in CAD.
Collapse
Affiliation(s)
- Zikai Song
- Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Shijie Lv
- Department of Orthopedics, Jilin Province FAW General Hospital, Changchun, Jilin Province, China
| | - Haidi Wu
- Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Ling Qin
- Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Hongyan Cao
- Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Bo Zhang
- Department of Pediatric Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Shuping Ren
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
24
|
Moriyama R, Iwamoto K, Hagiwara T, Yoshida S, Kato T, Kato Y. AMP-activated protein kinase activation reduces the transcriptional activity of the murine luteinizing hormone β-subunit gene. J Reprod Dev 2019; 66:97-104. [PMID: 31813919 PMCID: PMC7175385 DOI: 10.1262/jrd.2019-143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Malnutrition is one of the factors that induces reproductive disorders. However, the underlying biological processes are unclear. AMP-activated protein kinase (AMPK) is an enzyme that plays crucial role as a cellular energy sensor. In the present study, we examined the effects of AMPK activation on the transcription of the murine gonadotropin subunit genes Cga, Lhb, and Fshb, and the gonadotropin-releasing hormone receptor Gnrh-r. Real-time PCR and transcription assay using LβT2 cells demonstrated that 5-amino-imidazole carboxamide riboside (AICAR), a cell-permeable AMP analog, repressed the expression of Lhb. Next, we examined deletion mutants of the upstream region of Lhb and found that the upstream regulatory region of Lhb (-2527 to -2198 b) was responsible for the repression by AICAR. Furthermore, putative transcription factors (SP1, STAT5a, and TEF) that might mediate transcriptional control of the Lhb repression induced by AICAR were identified. In addition, it was confirmed that both AICAR and a competitive inhibitor of glucose metabolism, 2-deoxy-D-glucose, induced AMPK phosphorylation in LβT2 cells. Therefore, the upstream region of Lhb is one of the target sites for glucoprivation inducing AMPK activation. In addition, AMPK plays a role in repressing Lhb expression through the distal -2527 to -2198 b region.
Collapse
Affiliation(s)
- Ryutaro Moriyama
- Laboratory of Environmental Physiology, Department of Life Science, School of Science and Engineering, Kindai University, Osaka 577-8502, Japan
| | - Koichi Iwamoto
- Laboratory of Environmental Physiology, Department of Life Science, School of Science and Engineering, Kindai University, Osaka 577-8502, Japan
| | - Teruki Hagiwara
- Laboratory of Molecular and Cellular Biology, Department of Life Science, School of Science and Engineering, Kindai University, Osaka 577-8502, Japan
| | - Saishu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo 105-8461, Japan.,Laboratory of Molecular Biology and Gene Regulation, Department of Life Science, School of Agriculture, Meiji University, Kanagawa 214-8571, Japan
| | - Takako Kato
- Laboratory of Molecular Biology and Gene Regulation, Department of Life Science, School of Agriculture, Meiji University, Kanagawa 214-8571, Japan
| | - Yukio Kato
- Laboratory of Molecular Biology and Gene Regulation, Department of Life Science, School of Agriculture, Meiji University, Kanagawa 214-8571, Japan
| |
Collapse
|