1
|
Han B, Zhen F, Sun Y, Sun B, Wang HY, Liu W, Huang J, Liang X, Wang YR, Chen XS, Li SJ, Hu J. Tumor suppressor KEAP1 promotes HSPA9 degradation, controlling mitochondrial biogenesis in breast cancer. Cell Rep 2024; 43:114507. [PMID: 39003742 DOI: 10.1016/j.celrep.2024.114507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/29/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
The oxidative-stress-related protein Kelch-like ECH-associated protein 1 (KEAP1) is a substrate articulator of E3 ubiquitin ligase, which plays an important role in the ubiquitination modification of proteins. However, the function of KEAP1 in breast cancer and its impact on the survival of patients with breast cancer remain unclear. Our study demonstrates that KEAP1, a positive prognostic factor, plays a crucial role in regulating cell proliferation, apoptosis, and cell cycle transition in breast cancer. We investigate the underlying mechanism using human tumor tissues, high-throughput detection technology, and a mouse xenograft tumor model. KEAP1 serves as a key regulator of cellular metabolism, the reprogramming of which is one of the hallmarks of tumorigenesis. KEAP1 has a significant effect on mitochondrial biogenesis and oxidative phosphorylation by regulating HSPA9 ubiquitination and degradation. These results suggest that KEAP1 could serve as a potential biomarker and therapeutic target in the treatment of breast cancer.
Collapse
Affiliation(s)
- Bing Han
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, Heilongjiang Province 150040, China
| | - Fang Zhen
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, Heilongjiang Province 150040, China
| | - Yue Sun
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, Heilongjiang Province 150040, China
| | - Bin Sun
- Research Center for Pharmacoinformatics (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, 157 Baojian Road, Harbin, Heilongjiang Province 150081, China
| | - Hong-Yi Wang
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, Heilongjiang Province 150040, China
| | - Wei Liu
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, Heilongjiang Province 150040, China
| | - Jian Huang
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, Heilongjiang Province 150040, China
| | - Xiao Liang
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang Province 150081, China
| | - Ya-Ru Wang
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, Heilongjiang Province 150040, China
| | - Xue-Song Chen
- Department of Oncology, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang Province 150001, China.
| | - Shui-Jie Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province 150081, China.
| | - Jing Hu
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, 150 Haping Road, Harbin, Heilongjiang Province 150040, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang Province 150081, China.
| |
Collapse
|
2
|
Novotná K, Tenora L, Slusher BS, Rais R. Therapeutic resurgence of 6-diazo-5-oxo-l-norleucine (DON) through tissue-targeted prodrugs. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 100:157-180. [PMID: 39034051 DOI: 10.1016/bs.apha.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
The recognition that rapidly proliferating cancer cells rely heavily on glutamine for their survival and growth has renewed interest in the development of glutamine antagonists for cancer therapy. Glutamine plays a pivotal role as a carbon source for synthesizing lipids and metabolites through the TCA cycle, as well as a nitrogen source for synthesis of amino acid and nucleotides. Numerous studies have explored the significance of glutamine metabolism in cancer, providing a robust rationale for targeting this metabolic pathway in cancer treatment. The glutamine antagonist 6-diazo-5-oxo-l-norleucine (DON) has been explored as an anticancer therapeutic for nearly six decades. Initial investigations revealed remarkable efficacy in preclinical studies and promising outcomes in early clinical trials. However, further advancement of DON was hindered due to dose-limiting gastrointestinal (GI) toxicities as the GI system is highly dependent on glutamine for regulating growth and repair. In an effort to repurpose DON and mitigate gastrointestinal (GI) toxicity concerns, prodrug strategies were utilized. These strategies aimed to enhance the delivery of DON to specific target tissues, such as tumors and the central nervous system (CNS), while sparing DON delivery to normal tissues, particularly the GI tract. When administered at low daily doses, optimized for metabolic inhibition, these prodrugs exhibit remarkable effectiveness without inducing significant toxicity to normal tissues. This approach holds promise for overcoming past challenges associated with DON, offering an avenue for its successful utilization in cancer treatment.
Collapse
Affiliation(s)
- Kateřina Novotná
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, United States; Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic v.v.i., Prague, Czech Republic; Department of Organic Chemistry, Charles University, Faculty of Science, Prague, Czech Republic
| | - Lukáš Tenora
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, United States; Department of Organic Chemistry, Charles University, Faculty of Science, Prague, Czech Republic
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, United States; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States; Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States; Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, United States; Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States; Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States.
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, United States; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States; Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
3
|
Sun S, Qi G, Chen H, He D, Ma D, Bie Y, Xu L, Feng B, Pang Q, Guo H, Zhang R. Ferroptosis sensitization in glioma: exploring the regulatory mechanism of SOAT1 and its therapeutic implications. Cell Death Dis 2023; 14:754. [PMID: 37980334 PMCID: PMC10657441 DOI: 10.1038/s41419-023-06282-1] [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: 06/10/2023] [Revised: 10/30/2023] [Accepted: 11/07/2023] [Indexed: 11/20/2023]
Abstract
Glioma, the most common primary malignant tumor of the central nervous system, lacks effective targeted therapies. This study investigates the role of SOAT1, a key gene involved in cholesterol esterification, in glioma prognosis and its association with ferroptosis. Although the impact of SOAT1 on glioma prognosis has been recognized, its precise mechanism remains unclear. In this study, we demonstrate that inhibiting SOAT1 increases the sensitivity of glioma cells to ferroptosis, both in vitro and in vivo. Mechanistically, SOAT1 positively modulates the expression of SLC40A1, an iron transporter, resulting in enhanced intracellular iron outflow, reduced intracellular iron levels, and subsequent disruption of ferroptosis. Importantly, we find that SOAT1 regulates ferroptosis independently of SREBPs, which are known to be involved in ferroptosis regulation. Furthermore, we identify the involvement of the PI3K-AKT-mTOR signaling pathway in mediating the regulatory effects of SOAT1 on SLC40A1 expression and ferroptosis sensitivity. These findings highlight the contribution of intracellular signaling cascades in the modulation of ferroptosis by SOAT1. We show that inhibiting SOAT1 enhances the efficacy of radiotherapy in gliomas, both in vitro and in vivo, by promoting sensitivity to ferroptosis. This suggests that targeting SOAT1 could potentially improve therapeutic outcomes for glioma patients. In summary, this study uncovers the pivotal role of SOAT1 as a link between cholesterol esterification and ferroptosis in glioma. Our findings underscore the potential of SOAT1 as a promising clinical therapeutic target, providing new avenues for the development of effective treatments for glioma. Further research is warranted to unravel the complete regulatory mechanisms of SOAT1 and explore its clinical applications.
Collapse
Affiliation(s)
- Shicheng Sun
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Guoliang Qi
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Hao Chen
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Dong He
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Dengzhen Ma
- Department of Neurosurgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Yifan Bie
- Department of Radiology, The Second Hospital, Shandong University, Jinan, China
| | - Linzong Xu
- Tumor Research and Therapy Center, Lanzhou University Second Hospital, Lanzhou, 730030, China
- Tumor Research and Therapy Center, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Bin Feng
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Qi Pang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Department of Neurosurgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Hua Guo
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Rui Zhang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
4
|
Geng F, Zhong Y, Su H, Lefai E, Magaki S, Cloughesy TF, Yong WH, Chakravarti A, Guo D. SREBP-1 upregulates lipophagy to maintain cholesterol homeostasis in brain tumor cells. Cell Rep 2023; 42:112790. [PMID: 37436895 PMCID: PMC10528745 DOI: 10.1016/j.celrep.2023.112790] [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: 06/27/2022] [Revised: 05/22/2023] [Accepted: 06/23/2023] [Indexed: 07/14/2023] Open
Abstract
Cholesterol is a structural component of cell membranes. How rapidly growing tumor cells maintain membrane cholesterol homeostasis is poorly understood. Here, we found that glioblastoma (GBM), the most lethal brain tumor, maintains normal levels of membrane cholesterol but with an abundant presence of cholesteryl esters (CEs) in its lipid droplets (LDs). Mechanistically, SREBP-1 (sterol regulatory element-binding protein 1), a master transcription factor that is activated upon cholesterol depletion, upregulates critical autophagic genes, including ATG9B, ATG4A, and LC3B, as well as lysosome cholesterol transporter NPC2. This upregulation promotes LD lipophagy, resulting in the hydrolysis of CEs and the liberation of cholesterol from the lysosomes, thus maintaining plasma membrane cholesterol homeostasis. When this pathway is blocked, GBM cells become quite sensitive to cholesterol deficiency with poor growth in vitro. Our study unravels an SREBP-1-autophagy-LD-CE hydrolysis pathway that plays an important role in maintaining membrane cholesterol homeostasis while providing a potential therapeutic avenue for GBM.
Collapse
Affiliation(s)
- Feng Geng
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital, The Ohio State University, Columbus, OH 43210, USA; Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA; College of Medicine at The Ohio State University, Columbus, OH 43210, USA
| | - Yaogang Zhong
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital, The Ohio State University, Columbus, OH 43210, USA; Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA; College of Medicine at The Ohio State University, Columbus, OH 43210, USA
| | - Huali Su
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital, The Ohio State University, Columbus, OH 43210, USA; Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA; College of Medicine at The Ohio State University, Columbus, OH 43210, USA
| | - Etienne Lefai
- Human Nutrition Unit, French National Research Institute for Agriculture, Food and Environment, University Clermont Auvergne, 63122 Clermont-Ferrand, France
| | - Shino Magaki
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Timothy F Cloughesy
- Department of Neurology (Neuro-Oncology), David Geffen School of Medicine at the University of California, Los Angeles, CA 90095, USA
| | - William H Yong
- Department of Pathology and Laboratory Medicine, School of Medicine at University of California, Irvine, CA 92617, USA
| | - Arnab Chakravarti
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital, The Ohio State University, Columbus, OH 43210, USA; Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA; College of Medicine at The Ohio State University, Columbus, OH 43210, USA
| | - Deliang Guo
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital, The Ohio State University, Columbus, OH 43210, USA; Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA; College of Medicine at The Ohio State University, Columbus, OH 43210, USA; Center of Cancer Metabolism, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA.
| |
Collapse
|
5
|
Chen Y, Tan L, Gao J, Lin C, Wu F, Li Y, Zhang J. Targeting glutaminase 1 (GLS1) by small molecules for anticancer therapeutics. Eur J Med Chem 2023; 252:115306. [PMID: 36996714 DOI: 10.1016/j.ejmech.2023.115306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/16/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023]
Abstract
Glutaminase-1 (GLS1) is a critical enzyme involved in several cellular processes, and its overexpression has been linked to the development and progression of cancer. Based on existing research, GLS1 plays a crucial role in the metabolic activities of cancer cells, promoting rapid proliferation, cell survival, and immune evasion. Therefore, targeting GLS1 has been proposed as a promising cancer therapy strategy, with several GLS1 inhibitors currently under development. To date, several GLS1 inhibitors have been identified, which can be broadly classified into two types: active site and allosteric inhibitors. Despite their pre-clinical effectiveness, only a few number of these inhibitors have advanced to initial clinical trials. Hence, the present medical research emphasizes the need for developing small molecule inhibitors of GLS1 possessing significantly high potency and selectivity. In this manuscript, we aim to summarize the regulatory role of GLS1 in physiological and pathophysiological processes. We also provide a comprehensive overview of the development of GLS1 inhibitors, focusing on multiple aspects such as target selectivity, in vitro and in vivo potency and structure-activity relationships.
Collapse
Affiliation(s)
- Yangyang Chen
- Joint Research Institution of Altitude Health, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lun Tan
- Joint Research Institution of Altitude Health, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jing Gao
- Joint Research Institution of Altitude Health, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Congcong Lin
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Fengbo Wu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Yang Li
- Joint Research Institution of Altitude Health, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Jifa Zhang
- Joint Research Institution of Altitude Health, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
6
|
Wu T, Li C, Zhou J, Han L, Qiang S, Hu Z, Liu J, Li X, Zhao W, Chen X. Primaquine activates Keratin 7 to treat diabetes and its complications. J Diabetes Metab Disord 2022; 21:1731-1741. [PMID: 36404863 PMCID: PMC9672200 DOI: 10.1007/s40200-022-01135-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/19/2022] [Indexed: 10/14/2022]
Abstract
Background The global prevalence of type 2 diabetes mellitus (T2DM) raises the rates of its complications, such as diabetic nephropathy and cardiovascular diseases. To conquer the complications, new strategies to reverse the deterioration of T2DM are urgently needed. In this project, we aimed to examine the hypoglycemic effect of primaquine and explore its specific target. Methods In vitro T2DM insulin resistance model was built in HepG2 cells to screen the potential anti-diabetic chemicals. On the other hand, the potential protein targets were explored by molecular docking. Accordingly, we chose C57BL/6 N mice to establish T2DM model to verify the effect of the chemicals on anti-hyperglycemia and diabetic complications. Results By targeting the Keratin 7 (K7) to activate EGFR/Akt glucose metabolism signaling pathway, primaquine poses a potent hypoglycemic effect. The level of acetyl-CoA is enhanced markedly, supporting that primaquine upregulates the aerobic glycolysis. Moreover, primaquine ameliorates kidney function by reducing the secretion of urinary proteins and creatinine, especially for the urea nitrogen which is significantly decreased compared to no-treatment T2DM mice. Notably, primaquine restores the level of plasma low-density lipoprotein cholesterol (LDL-C) nearly to normal, minimizing the incidence of cardiovascular diseases. Conclusions We find that primaquine may reverse the dysregulated metabolism to prevent diabetic complications by stimulating EGFR/Akt signaling axis, shedding new light on the therapy of T2DM. Graphical abstract Insulin resistance is characterized by reduced p-Akt and glucose metabolism, dominated by anaerobic glycolysis. Primaquine activates the complex made of K7 and EGFR, further stimulating Akt phosphorylation. Then, p-Akt promotes the aerobic glucose metabolism and upregulates Ac-CoA to mobilize TCA cycle, improving insulin sensitivity. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-022-01135-8.
Collapse
Affiliation(s)
- Tongyu Wu
- Department of Pharmacy, Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Chun Li
- Department of Pharmacy, Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Jing Zhou
- Department of Pharmacy, Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Liang Han
- Department of Pharmacy, Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Shaojia Qiang
- Department of Pharmacy, Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Zhuozhou Hu
- Department of Pharmacy, Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Jingjing Liu
- Department of Pharmacy, Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Xiangxiang Li
- Department of Pharmacy, Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Wenyang Zhao
- Department of Pharmacy, Lanzhou University, Lanzhou, 730000 People’s Republic of China
| | - Xinping Chen
- Department of Pharmacy, Lanzhou University, Lanzhou, 730000 People’s Republic of China
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou, 730000 People’s Republic of China
| |
Collapse
|
7
|
Ji Lee E, Babu Duggirala K, Lee Y, Ran Yun M, Jang J, Cyriac R, Eun Jung M, Choi G, Hak Chae C, Chul Cho B, Lee K. Novel Allosteric Glutaminase 1 Inhibitors with Macrocyclic Structure Activity Relationship Analysis. Bioorg Med Chem Lett 2022; 75:128956. [PMID: 36038117 DOI: 10.1016/j.bmcl.2022.128956] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/11/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022]
Abstract
Glutamine-addicted cancer metabolism is recently recognized as novel cancer target especially for KRAS and KEAP1 co-occurring mutations. Selective glutaminase1 (GLS1) inhibition was reported using BPTES which has novel mode of allosteric inhibition. However, BPTES is a highly hydrophobic and symmetric molecule with very poor solubility which results in suboptimal pharmacokinetic parameters and hinders its further development. As an ongoing effort to identify more drug-like GLS1 inhibitors via systematic structure-activity relationship (SAR) analysis of BPTES analogs, we disclose our novel macrocycles for GLS1 inhibition with conclusive SAR analysis on the core, core linker, and wing linker, respectively. Selected molecules resulted in reduction in intracellular glutamate levels in LR (LDK378-resistant) cells which is consistent to cell viability result. Finally, compounds 13 selectively reduced the growth of A549 and H460 cells which have co-occurring mutations including KRAS and KEAP1.
Collapse
Affiliation(s)
- Eun Ji Lee
- Department of Research Support, Yonsei Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Krishna Babu Duggirala
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, South Korea; Medicinal Chemistry & Pharmacology, University of Science & Technology, Daejeon 34113, South Korea
| | - Yujin Lee
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, South Korea; Medicinal Chemistry & Pharmacology, University of Science & Technology, Daejeon 34113, South Korea
| | - Mi Ran Yun
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, South Korea; Yonsei New Ii Han Institute for Integrative Lung Cancer Research, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Jiyoon Jang
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, South Korea; Department of Chemistry, Sungkyunkwan University, Suwon 16419, South Korea
| | - Rajath Cyriac
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, South Korea; Medicinal Chemistry & Pharmacology, University of Science & Technology, Daejeon 34113, South Korea
| | - Myoung Eun Jung
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, South Korea
| | - Gildon Choi
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, South Korea; Medicinal Chemistry & Pharmacology, University of Science & Technology, Daejeon 34113, South Korea
| | - Chong Hak Chae
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, South Korea
| | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Kwangho Lee
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, South Korea; Medicinal Chemistry & Pharmacology, University of Science & Technology, Daejeon 34113, South Korea.
| |
Collapse
|
8
|
Single-Cell FISH Analysis Reveals Distinct Shifts in PKM Isoform Populations during Drug Resistance Acquisition. Biomolecules 2022; 12:biom12081082. [PMID: 36008976 PMCID: PMC9405743 DOI: 10.3390/biom12081082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
The Warburg effect, i.e., the utilization of glycolysis under aerobic conditions, is recognized as a survival advantage of cancer cells. However, how the glycolytic activity is affected during drug resistance acquisition has not been explored at single-cell resolution. Because the relative ratio of the splicing isoform of pyruvate kinase M (PKM), PKM2/PKM1, can be used to estimate glycolytic activity, we utilized a single-molecule fluorescence in situ hybridization (SM-FISH) method to simultaneously quantify the mRNA levels of PKM1 and PKM2. Treatment of HCT116 cells with gefitinib (GE) resulted in two distinct populations of cells. However, as cells developed GE resistance, the GE-sensitive population with reduced PKM2 expression disappeared, and GE-resistant cells (Res) demonstrated enhanced PKM1 expression and a tightly regulated PKM2/PKM1 ratio. Our data suggest that maintaining an appropriate PKM2 level is important for cell survival upon GE treatment, whereas increased PKM1 expression becomes crucial in GE Res. This approach demonstrates the importance of single-cell-based analysis for our understanding of cancer cell metabolic responses to drugs, which could aid in the design of treatment strategies for drug-resistant cancers.
Collapse
|
9
|
Puca F, Yu F, Bartolacci C, Pettazzoni P, Carugo A, Huang-Hobbs E, Liu J, Zanca C, Carbone F, Del Poggetto E, Gumin J, Dasgupta P, Seth S, Srinivasan S, Lang FF, Sulman EP, Lorenzi PL, Tan L, Shan M, Tolstyka ZP, Kachman M, Zhang L, Gao S, Deem AK, Genovese G, Scaglioni PP, Lyssiotis CA, Viale A, Draetta GF. Medium-Chain Acyl-CoA Dehydrogenase Protects Mitochondria from Lipid Peroxidation in Glioblastoma. Cancer Discov 2021; 11:2904-2923. [PMID: 34039636 PMCID: PMC8711129 DOI: 10.1158/2159-8290.cd-20-1437] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 03/25/2021] [Accepted: 05/18/2021] [Indexed: 11/16/2022]
Abstract
Glioblastoma (GBM) is highly resistant to chemotherapies, immune-based therapies, and targeted inhibitors. To identify novel drug targets, we screened orthotopically implanted, patient-derived glioblastoma sphere-forming cells using an RNAi library to probe essential tumor cell metabolic programs. This identified high dependence on mitochondrial fatty acid metabolism. We focused on medium-chain acyl-CoA dehydrogenase (MCAD), which oxidizes medium-chain fatty acids (MCFA), due to its consistently high score and high expression among models and upregulation in GBM compared with normal brain. Beyond the expected energetics impairment, MCAD depletion in primary GBM models induced an irreversible cascade of detrimental metabolic effects characterized by accumulation of unmetabolized MCFAs, which induced lipid peroxidation and oxidative stress, irreversible mitochondrial damage, and apoptosis. Our data uncover a novel protective role for MCAD to clear lipid molecules that may cause lethal cell damage, suggesting that therapeutic targeting of MCFA catabolism may exploit a key metabolic feature of GBM. SIGNIFICANCE: MCAD exerts a protective role to prevent accumulation of toxic metabolic by-products in glioma cells, actively catabolizing lipid species that would otherwise affect mitochondrial integrity and induce cell death. This work represents a first demonstration of a nonenergetic role for dependence on fatty acid metabolism in cancer.This article is highlighted in the In This Issue feature, p. 2659.
Collapse
Affiliation(s)
- Francesca Puca
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Fei Yu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Caterina Bartolacci
- Department of Internal Medicine Division of Hematology & Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Piergiorgio Pettazzoni
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alessandro Carugo
- The Translational Research to Advance Therapeutics and Innovation in Oncology Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Emmet Huang-Hobbs
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jintan Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ciro Zanca
- The Translational Research to Advance Therapeutics and Innovation in Oncology Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Federica Carbone
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Edoardo Del Poggetto
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joy Gumin
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pushan Dasgupta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sahil Seth
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The Translational Research to Advance Therapeutics and Innovation in Oncology Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sanjana Srinivasan
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Erik P Sulman
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Philip L Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mengrou Shan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Zachary P Tolstyka
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Maureen Kachman
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, Michigan
| | - Li Zhang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Sisi Gao
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Angela K Deem
- The Translational Research to Advance Therapeutics and Innovation in Oncology Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Giannicola Genovese
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pier Paolo Scaglioni
- Department of Internal Medicine Division of Hematology & Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan.
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, Michigan
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Andrea Viale
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Giulio F Draetta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
10
|
Rathod B, Chak S, Patel S, Shard A. Tumor pyruvate kinase M2 modulators: a comprehensive account of activators and inhibitors as anticancer agents. RSC Med Chem 2021; 12:1121-1141. [PMID: 34355179 PMCID: PMC8292966 DOI: 10.1039/d1md00045d] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/25/2021] [Indexed: 12/16/2022] Open
Abstract
Pyruvate kinase M2 (PKM2) catalyzes the conversion of phosphoenolpyruvate (PEP) to pyruvate. It plays a central role in the metabolic reprogramming of cancer cells and is expressed in most human tumors. It is essential in indiscriminate proliferation, survival, and tackling apoptosis in cancer cells. This positions PKM2 as a hot target in cancer therapy. Despite its well-known structure and several reported modulators targeting PKM2 as activators or inhibitors, a comprehensive review focusing on such modulators is lacking. Herein we summarize modulators of PKM2, the assays used to detect their potential, the preferable tense (T) and relaxed (R) states in which the enzyme resides, lacunae in existing modulators, and several strategies that may lead to effective anticancer drug development targeting PKM2.
Collapse
Affiliation(s)
- Bhagyashri Rathod
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad Opposite Air Force Station Gandhinagar Gujarat 382355 India
| | - Shivam Chak
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad Opposite Air Force Station Gandhinagar Gujarat 382355 India
| | - Sagarkumar Patel
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad Opposite Air Force Station Gandhinagar Gujarat 382355 India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad Opposite Air Force Station Gandhinagar Gujarat 382355 India
| |
Collapse
|
11
|
Xi H, He Z, Lv C. FOXG1 improves mitochondrial function and promotes the progression of nasopharyngeal carcinoma. Mol Med Rep 2021; 24:651. [PMID: 34278485 PMCID: PMC8299199 DOI: 10.3892/mmr.2021.12290] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 05/14/2021] [Indexed: 12/28/2022] Open
Abstract
Forkhead‑box gene 1 (FOXG1) has been reported to serve an important role in various malignancies, but its effects on nasopharyngeal cancer (NPC) remain unknown. Thus, the present study aimed to investigate the specific regulatory relationship between FOXG1 and NPC progression. Tumor tissues and matching para‑carcinoma tissues were obtained from patients with NPC. Small interfering (si)RNA‑FOXG1 and pcDNA3.1‑FOXG1 were transfected into SUNE‑1 and C666‑1 cells to knockdown and overexpress FOXG1 expression, respectively. FOXG1 expression was detected using reverse transcription‑quantitative PCR and immunohistochemistry. Cell proliferation was detected using MTT and 5‑ethynyl‑20‑deoxyuridine assays. Transwell invasion assay, wound healing assay and flow cytometry were used to detect cell invasion, migration and apoptosis, respectively. Western blotting was conducted to detect the expression levels of mitochondrial markers (succinate dehydrogenase complex flavoprotein subunit A, heat shock protein 60 and pyruvate dehydrogenase), epithelial‑mesenchymal transition (EMT) related proteins (N‑cadherin, Snail and E‑cadherin) and apoptosis‑related proteins [Bax, Bcl‑2, poly(ADP‑ribose) polymerase 1 (PARP), cleaved PARP, cleaved caspase‑3, cleaved caspase‑8, cleaved caspase‑9, caspase‑3, caspase‑8 and caspase‑9]. The mitochondrial membrane potential was detected via flow cytometry, while the ATP/ADP ratio was determined using the ADP/ATP ratio assay kit. The present results demonstrated that FOXG1 expression was upregulated in NPC tissues and cells, and was associated with distant metastasis and TNM stage. Moreover, knockdown of FOXG1 inhibited the proliferation, migration, invasion, EMT and mitochondrial function of SUNE‑1 cells, as well as promoted cell apoptosis, while the opposite results were observed in C666‑1 cells. In conclusion, FOXG1 enhanced proliferation, migration and invasion, induced EMT and improved mitochondrial function in NPC cells. The current findings provide an adequate theoretical basis for the treatment of NPC.
Collapse
Affiliation(s)
- Huajun Xi
- Department of Otolaryngology and Stomatology, Shouguang People's Hospital, Shouguang, Shandong 262700, P.R. China
| | - Zhengxiang He
- Department of Otolaryngology and Maxillofacial Surgery, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| | - Cao Lv
- Department of Otolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| |
Collapse
|
12
|
Moraes JA, Encarnação C, Franco VA, Xavier Botelho LG, Rodrigues GP, Ramos-Andrade I, Barja-Fidalgo C, Renovato-Martins M. Adipose Tissue-Derived Extracellular Vesicles and the Tumor Microenvironment: Revisiting the Hallmarks of Cancer. Cancers (Basel) 2021; 13:3328. [PMID: 34283044 PMCID: PMC8268128 DOI: 10.3390/cancers13133328] [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: 05/31/2021] [Revised: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are crucial elements that sustain the communication between tumor cells and their microenvironment, and have emerged as a widespread mechanism of tumor formation and metastasis. In obesity, the adipose tissue becomes hypertrophic and hyperplastic, triggering increased production of pro-inflammatory adipokines, such as tumor necrosis factor α, interleukin 6, interleukin 1, and leptin. Furthermore, obese adipose tissue undergoes dysregulation in the cargo content of the released EVs, resulting in an increased content of pro-inflammatory proteins, fatty acids, and oncogenic microRNAs. These alterations drive obesity-associated inflammatory responses both locally and systemically. After being ignored for a long time, adipose tissues have recently received considerable attention as a major player in tumor microenvironment-linked obesity and cancer. The role of adipose tissue in the establishment and progression of cancer is reinforced by its high plasticity and inflammatory content. Such a relationship may be established by direct contact between adipocytes and cancer cells within the microenvironment or systemically, via EV-mediated cell-to-cell communication. Here, we highlight cues evidencing the influence of adipose tissue-derived EVs on the hallmarks of cancer, which are critical for tumor malignancy.
Collapse
Affiliation(s)
- João Alfredo Moraes
- Redox Biology Laboratory, Programa de Pesquisa em Farmacologia e Inflamação, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil;
| | - Carol Encarnação
- Laboratory of Inflammation and Metabolism, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, 24210-201 Niterói, Brazil; (C.E.); (V.A.F.); (L.G.X.B.); (G.P.R.)
| | - Victor Aguiar Franco
- Laboratory of Inflammation and Metabolism, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, 24210-201 Niterói, Brazil; (C.E.); (V.A.F.); (L.G.X.B.); (G.P.R.)
| | - Luiz Gabriel Xavier Botelho
- Laboratory of Inflammation and Metabolism, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, 24210-201 Niterói, Brazil; (C.E.); (V.A.F.); (L.G.X.B.); (G.P.R.)
| | - Gabriella Pacheco Rodrigues
- Laboratory of Inflammation and Metabolism, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, 24210-201 Niterói, Brazil; (C.E.); (V.A.F.); (L.G.X.B.); (G.P.R.)
| | - Isadora Ramos-Andrade
- Laboratory of Cellular and Molecular Pharmacology, Departamento de Biologia Celular, IBRAG, Universidade do Estado do Rio de Janeiro, 20550-170 Rio de Janeiro, Brazil; (I.R.-A.); (C.B.-F.)
| | - Christina Barja-Fidalgo
- Laboratory of Cellular and Molecular Pharmacology, Departamento de Biologia Celular, IBRAG, Universidade do Estado do Rio de Janeiro, 20550-170 Rio de Janeiro, Brazil; (I.R.-A.); (C.B.-F.)
| | - Mariana Renovato-Martins
- Laboratory of Inflammation and Metabolism, Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, 24210-201 Niterói, Brazil; (C.E.); (V.A.F.); (L.G.X.B.); (G.P.R.)
| |
Collapse
|
13
|
Zhao S, Zhang Y, Pei M, Wu L, Li J. miR-145 inhibits mitochondrial function of ovarian cancer by targeting ARL5B. J Ovarian Res 2021; 14:8. [PMID: 33419459 PMCID: PMC7796643 DOI: 10.1186/s13048-020-00762-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/26/2020] [Indexed: 12/21/2022] Open
Abstract
Metabolic reprogramming refers to the transformation of the whole metabolic network including glycolysis and mitochondrial metabolism, mainly manifested in Warburg effect and mitochondrial metabolic reprogramming. The roles of miR-145 in glycolysis have been established in ovarian cancer cells. Howerer, its roles in mitochondrial metabolic reprogramming are still unclear. This study aims to identify whether miR-145 regulates mitochondrial metabolic reprogramming in ovarian cancer cells. First, functional experiment showed that overexpression of miR-145 inhibited mitochondrial function in ovarian cancer cells, evident by the decreased mtDNA copy numbers, ATP level, mitochondrial membrane potential, and the expression levels of mitochondrial markers. Mechanistically, miR-145 inhibited mitochondrial function by targeting ARL5B directly. Futhermore, miR-145 overexpression decreased ARL5B expression in ovarian cancer tissue subcutaneous tumors of nude mice. In conclusion, we have highlighted that miR-145 inhibits mitochondrial function and achieves this by targeting ARL5B directly for the first time. The results provides a more adequate theoretical basis for understanding the molecular pathology of ovarian cancer, and provides the necessary basic data for miR-145 as a potential diagnosis and treatment target for ovarian cancer.
Collapse
Affiliation(s)
- Shuo Zhao
- Department of SICU, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yun Zhang
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, 710061, Xi'an, Shaanxi, China
| | - Meili Pei
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lei Wu
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jie Li
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, 710061, Xi'an, Shaanxi, China.
| |
Collapse
|
14
|
Diospyros kaki and Citrus unshiu Mixture Improves Disorders of Lipid Metabolism in Nonalcoholic Fatty Liver Disease. Can J Gastroenterol Hepatol 2020; 2020:8812634. [PMID: 33425805 PMCID: PMC7775147 DOI: 10.1155/2020/8812634] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 12/02/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has been a major cause of a chronic liver disease over recent decades and increasing worldwide in parallel with the remarkable growth of obesity. In the present study, we investigate the ameliorative effects of PCM, a combination of Diospyros kaki fruit and Citrus unshiu peel mixture, on high-fat diet- (HFD-) induced NAFLD and clarify the potential mechanisms. PCM in HFD-fed mice was orally administered at a dose of 50 or 100 mg/kg subsequently for 2 months. Thereafter, lipid metabolism parameters and fat synthesis-related genes in the mouse liver were evaluated. Subsequently, body weight changes, liver weight, serum liver function and lipid profiles, and liver pathology were examined, and the relative levels of fatty acid synthesis and β-oxidation gene expression were evaluated by western blot. Serum AST, ALT, and TG levels in the HFD control mice were significantly higher than those of normal mice. Compared with HFD control mice, PCM supplementation increased phosphorylation of AMP-activated protein kinase (AMPK). Peroxisome proliferator-activated receptor (PPAR) α was significantly increased by PCM administration. Continuously, the activation of PPARα significantly elevated carnitine palmitoyltransferase 1 (CPT-1), a key enzyme in fatty acid β-oxidation, and mitochondrial uncoupling protein 2 (UCP-2), thermogenic regulatory genes, in PCM-treated mice compared with those of HFD control mice. Moreover, PCM inhibits lipogenesis and cholesterol synthesis via suppression of sterol regulatory element binding protein-1 (SREBP-1) and SREBP-2 and its target genes such as acetyl-CoA carboxylase (ACC), fatty acid synthase (FAS), stearoyl-CoA desaturase-1 (SCD-1), and 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR). Taken together, these effects were mediated through activation of AMPK. In the conclusion, PCM improved liver damage in HFD-fed mice and attenuated NAFLD by the activation of PPARα and the inhibition of SREBPs expression via AMPK-dependent pathways.
Collapse
|
15
|
Wu X, Geng F, Cheng X, Guo Q, Zhong Y, Cloughesy TF, Yong WH, Chakravarti A, Guo D. Lipid Droplets Maintain Energy Homeostasis and Glioblastoma Growth via Autophagic Release of Stored Fatty Acids. iScience 2020; 23:101569. [PMID: 33083736 PMCID: PMC7549116 DOI: 10.1016/j.isci.2020.101569] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/18/2020] [Accepted: 09/14/2020] [Indexed: 12/18/2022] Open
Abstract
Recently, lipid metabolism reprogramming has been further evidenced in malignancies via the observation of large amounts of lipid droplets (LDs) in human tumors, including in glioblastoma (GBM), the most lethal primary brain tumor. However, the role played by LDs in tumor cells remains unknown. Here, we show that triglycerides (TG), the major components of LDs, serve as a critical energy reservoir to support GBM cell survival. TG/LDs rapidly diminished in GBM cells upon glucose reduction, whereas inhibiting fatty acid oxidation or autophagy resulted in the accumulation of TG/LDs and strongly potentiated GBM cell death. Immunofluorescence imaging and time-lapse videos showed that LDs are hydrolyzed by autophagy to release free fatty acids that mobilize into mitochondria for energy production. Our study demonstrates that autophagy-mediated hydrolysis of TG/LDs maintains energy homeostasis and GBM survival upon glucose reduction, suggesting that limiting TG/LDs utilization might be necessary upon treating GBM. TG/LDs function as energy reservoir for GBM tumors TG/LDs are hydrolyzed by autophagy to maintain GBM survival when glucose levels decrease TG/LD hydrolysis releases fatty acids that enter into mitochondria for energy production Inhibiting autophagy causes TG/LD accumulation and GBM cell death
Collapse
Affiliation(s)
- Xiaoning Wu
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine at the Ohio State University, Columbus, OH 43210, USA
| | - Feng Geng
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine at the Ohio State University, Columbus, OH 43210, USA
| | - Xiang Cheng
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine at the Ohio State University, Columbus, OH 43210, USA
| | - Qiang Guo
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine at the Ohio State University, Columbus, OH 43210, USA
| | - Yaogang Zhong
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine at the Ohio State University, Columbus, OH 43210, USA
| | - Timothy F Cloughesy
- Department of Neurology (Neuro-Oncology), David Geffen School of Medicine at the University of California, Los Angeles, CA 90095, USA
| | - William H Yong
- Department of Pathology and Laboratory Medicine (Neuropathology), David Geffen School of Medicine at the University of California, Los Angeles, CA 90095, USA
| | - Arnab Chakravarti
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine at the Ohio State University, Columbus, OH 43210, USA
| | - Deliang Guo
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine at the Ohio State University, Columbus, OH 43210, USA.,Center for Cancer Metabolism, James Comprehensive Cancer Center at the Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
16
|
Nguyen T, Kirsch BJ, Asaka R, Nabi K, Quinones A, Tan J, Antonio MJ, Camelo F, Li T, Nguyen S, Hoang G, Nguyen K, Udupa S, Sazeides C, Shen YA, Elgogary A, Reyes J, Zhao L, Kleensang A, Chaichana KL, Hartung T, Betenbaugh MJ, Marie SK, Jung JG, Wang TL, Gabrielson E, Le A. Uncovering the Role of N-Acetyl-Aspartyl-Glutamate as a Glutamate Reservoir in Cancer. Cell Rep 2020; 27:491-501.e6. [PMID: 30970252 PMCID: PMC6472703 DOI: 10.1016/j.celrep.2019.03.036] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/14/2019] [Accepted: 03/08/2019] [Indexed: 11/13/2022] Open
Abstract
N-acetyl-aspartyl-glutamate (NAAG) is a peptide-based neurotransmitter that has been extensively studied in many neurological diseases. In this study, we show a specific role of NAAG in cancer. We found that NAAG is more abundant in higher grade cancers and is a source of glutamate in cancers expressing glutamate carboxypeptidase II (GCPII), the enzyme that hydrolyzes NAAG to glutamate and N-acetyl-aspartate (NAA). Knocking down GCPII expression through genetic alteration or pharmacological inhibition of GCPII results in a reduction of both glutamate concentrations and cancer growth. Moreover, targeting GCPII in combination with glutaminase inhibition accentuates these effects. These findings suggest that NAAG serves as an important reservoir to provide glutamate to cancer cells through GCPII when glutamate production from other sources is limited. Thus, GCPII is a viable target for cancer therapy, either alone or in combination with glutaminase inhibition. Nguyen et al. show that NAAG is more abundant in higher grade cancers and a source of glutamate in cancers expressing GCPII, the enzyme that hydrolyzes NAAG to glutamate and NAA. The results suggest that GCPII is a viable target for cancer therapy, either alone or in combination with glutaminase inhibition.
Collapse
Affiliation(s)
- Tu Nguyen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Brian James Kirsch
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA
| | - Ryoichi Asaka
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Karim Nabi
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Addison Quinones
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jessica Tan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | - Felipe Camelo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ting Li
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Stephanie Nguyen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Giang Hoang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kiet Nguyen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sunag Udupa
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Christos Sazeides
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yao-An Shen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Amira Elgogary
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Juvenal Reyes
- Department of Radiation Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Liang Zhao
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Andre Kleensang
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Kaisorn Lee Chaichana
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Thomas Hartung
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; University of Konstanz, 78464 Konstanz, Germany
| | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA
| | - Suely K Marie
- Department of Neurology, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Jin G Jung
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tian-Li Wang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Edward Gabrielson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Anne Le
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
17
|
Seow P, Narayanan V, Romelean RJ, Wong JHD, Win MT, Chandran H, Chinna K, Rahmat K, Ramli N. Lipid Fraction Derived From MRI In- and Opposed-Phase Sequence as a Novel Biomarker for Predicting Survival Outcome of Glioma. Acad Radiol 2020; 27:180-187. [PMID: 31155487 DOI: 10.1016/j.acra.2019.04.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/03/2019] [Accepted: 04/08/2019] [Indexed: 12/29/2022]
Abstract
RATIONALE AND PURPOSE Our study evaluated the capability of magnetic resonance imaging in- and opposed-phase (IOP) derived lipid fraction as a novel prognostic biomarker of survival outcome in glioma. MATERIALS AND METHODS We analyzed 46 histologically proven glioma (WHO grades II-IV) patients using standard 3T magnetic resonance imaging brain tumor protocol and IOP sequence. Lipid fraction was derived from the IOP sequence signal-loss ratio. The lipid fraction of solid nonenhancing region of glioma was analyzed, using a three-group analysis approach based on volume under surface of receiver-operating characteristics to stratify the prognostic factors into three groups of low, medium, and high lipid fraction. The survival outcome was evaluated, using Kaplan-Meier survival analysis and Cox regression model. RESULTS Significant differences were seen between the three groups (low, medium, and high lipid fraction groups) stratified by the optimal cut-off point for overall survival (OS) (p ≤ 0.01) and time to progression (p ≤ 0.01) for solid nonenhancing region. The group with high lipid fraction had five times higher risk of poor survival and earlier time to progression compared to the low lipid fraction group. The OS plot stratified by lipid fraction also had a strong correlation with OS plot stratified by WHO grade (R = 0.61, p < 0.01), implying association to underlying histopathological changes. CONCLUSION The lipid fraction of solid nonenhancing region showed potential for prognostication of glioma. This method will be a useful adjunct in imaging protocol for treatment stratification and as a prognostic tool in glioma patients.
Collapse
Affiliation(s)
- Pohchoo Seow
- Department of Biomedical Imaging, Faculty of Medicine, University of Malaya, Jalan Universiti, Kuala Lumpur 50603, Malaysia; Faculty of Medicine, University of Malaya Research Imaging Centre, University of Malaya, Kuala Lumpur, Malaysia
| | - Vairavan Narayanan
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Ronie J Romelean
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Jeannie Hsiu Ding Wong
- Department of Biomedical Imaging, Faculty of Medicine, University of Malaya, Jalan Universiti, Kuala Lumpur 50603, Malaysia; Faculty of Medicine, University of Malaya Research Imaging Centre, University of Malaya, Kuala Lumpur, Malaysia
| | - Myint Tun Win
- Department of Biomedical Imaging, Faculty of Medicine, University of Malaya, Jalan Universiti, Kuala Lumpur 50603, Malaysia
| | - Hari Chandran
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Karuthan Chinna
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Malaysia
| | - Kartini Rahmat
- Department of Biomedical Imaging, Faculty of Medicine, University of Malaya, Jalan Universiti, Kuala Lumpur 50603, Malaysia; Faculty of Medicine, University of Malaya Research Imaging Centre, University of Malaya, Kuala Lumpur, Malaysia
| | - Norlisah Ramli
- Department of Biomedical Imaging, Faculty of Medicine, University of Malaya, Jalan Universiti, Kuala Lumpur 50603, Malaysia; Faculty of Medicine, University of Malaya Research Imaging Centre, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
18
|
Hoang G, Udupa S, Le A. Application of metabolomics technologies toward cancer prognosis and therapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 347:191-223. [PMID: 31451214 DOI: 10.1016/bs.ircmb.2019.07.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Altered metabolism is one of the defining features of cancer. Since the discovery of the Warburg effect in 1924, research into the metabolic aspects of cancer has been reinvigorated over the past decade. Metabolomics is an invaluable tool for gaining insights into numerous biochemical processes including those related to cancer metabolism and metabolic aspects of other diseases. The combination of untargeted and targeted metabolomics approaches has greatly facilitated the discovery of many cancer biomarkers with prognostic potential. Using mass spectrometry-based stable isotope-resolved metabolomics (SIRM) with isotopic labeling, a powerful tool used in pathway analysis, researchers have discovered novel cancer metabolic pathways and metabolic targets for therapeutic application. Metabolomics technologies provide invaluable metabolic insights reflecting cancer progression in coordination with genomics and proteomics aspects. The systematic study of metabolite levels in the metabolome and their dynamics within a biological organism has been, in recent years, applied across a wide range of fields. Metabolomics technologies have been applied to both early clinical trials and pre-clinical research in several essential aspects of human health. This chapter will give an overview of metabolomics technologies and their application in the discovery of novel pathways using isotopic labeled and non-labeled metabolomics.
Collapse
Affiliation(s)
- Giang Hoang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Biomedical Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD, United States
| | - Sunag Udupa
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Anne Le
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
19
|
Li J, Li X, Wu L, Pei M, Li H, Jiang Y. miR‐145 inhibits glutamine metabolism through c‐myc/GLS1 pathways in ovarian cancer cells. Cell Biol Int 2019; 43:921-930. [PMID: 31115975 DOI: 10.1002/cbin.11182] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/08/2019] [Accepted: 05/19/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Jie Li
- Department of PathologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'an 710061 Shaanxi China
| | - Xu Li
- Center for Translational MedicineThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'an 710061 Shaanxi China
| | - Lei Wu
- Department of Gynecology and ObstetricsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'an 710061 Shaanxi China
| | - Meili Pei
- Department of Gynecology and ObstetricsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'an 710061 Shaanxi China
| | - Huijin Li
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational MedicineXi'an Medical UniversityXi'an 710061 Shaanxi China
| | - Yu Jiang
- Department of Gynecology and ObstetricsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'an 710061 Shaanxi China
| |
Collapse
|
20
|
Civita P, Franceschi S, Aretini P, Ortenzi V, Menicagli M, Lessi F, Pasqualetti F, Naccarato AG, Mazzanti CM. Laser Capture Microdissection and RNA-Seq Analysis: High Sensitivity Approaches to Explain Histopathological Heterogeneity in Human Glioblastoma FFPE Archived Tissues. Front Oncol 2019; 9:482. [PMID: 31231613 PMCID: PMC6568189 DOI: 10.3389/fonc.2019.00482] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/21/2019] [Indexed: 12/21/2022] Open
Abstract
Laser capture microdissection (LCM) coupled with RNA-seq is a powerful tool to identify genes that are differentially expressed in specific histological tumor subtypes. To better understand the role of single tumor cell populations in the complex heterogeneity of glioblastoma, we paired microdissection and NGS technology to study intra-tumoral differences into specific histological regions and cells of human GBM FFPE tumors. We here isolated astrocytes, neurons and endothelial cells in 6 different histological contexts: tumor core astrocytes, pseudopalisading astrocytes, perineuronal astrocytes in satellitosis, neurons with satellitosis, tumor blood vessels, and normal blood vessels. A customized protocol was developed for RNA amplification, library construction, and whole transcriptome analysis of each single portion. We first validated our protocol comparing the obtained RNA expression pattern with the gene expression levels of RNA-seq raw data experiments from the BioProject NCBI database, using Spearman's correlation coefficients calculation. We found a good concordance for pseudopalisading and tumor core astrocytes compartments (0.5 Spearman correlation) and a high concordance for perineuronal astrocytes, neurons, normal, and tumor endothelial cells compartments (0.7 Spearman correlation). Then, Principal Component Analysis and differential expression analysis were employed to find differences between tumor compartments and control tissue and between same cell types into distinct tumor contexts. Data consistent with the literature emerged, in which multiple therapeutic targets significant for glioblastoma (such as Integrins, Extracellular Matrix, transmembrane transport, and metabolic processes) play a fundamental role in the disease progression. Moreover, specific cellular processes have been associated with certain cellular subtypes within the tumor. Our results are promising and suggest a compelling method for studying glioblastoma heterogeneity in FFPE samples and its application in both prospective and retrospective studies.
Collapse
Affiliation(s)
| | | | | | - Valerio Ortenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, Pisa University Hospital, Pisa, Italy
| | | | | | | | - Antonio Giuseppe Naccarato
- Department of Translational Research and New Technologies in Medicine and Surgery, Pisa University Hospital, Pisa, Italy
| | | |
Collapse
|
21
|
Hu X, Matsumoto K, Jung RS, Weston TA, Heizer PJ, He C, Sandoval NP, Allan CM, Tu Y, Vinters HV, Liau LM, Ellison RM, Morales JE, Baufeld LJ, Bayley NA, He L, Betsholtz C, Beigneux AP, Nathanson DA, Gerhardt H, Young SG, Fong LG, Jiang H. GPIHBP1 expression in gliomas promotes utilization of lipoprotein-derived nutrients. eLife 2019; 8:e47178. [PMID: 31169500 PMCID: PMC6594755 DOI: 10.7554/elife.47178] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/05/2019] [Indexed: 12/25/2022] Open
Abstract
GPIHBP1, a GPI-anchored protein of capillary endothelial cells, binds lipoprotein lipase (LPL) within the subendothelial spaces and shuttles it to the capillary lumen. GPIHBP1-bound LPL is essential for the margination of triglyceride-rich lipoproteins (TRLs) along capillaries, allowing the lipolytic processing of TRLs to proceed. In peripheral tissues, the intravascular processing of TRLs by the GPIHBP1-LPL complex is crucial for the generation of lipid nutrients for adjacent parenchymal cells. GPIHBP1 is absent from the capillaries of the brain, which uses glucose for fuel; however, GPIHBP1 is expressed in the capillaries of mouse and human gliomas. Importantly, the GPIHBP1 in glioma capillaries captures locally produced LPL. We use NanoSIMS imaging to show that TRLs marginate along glioma capillaries and that there is uptake of TRL-derived lipid nutrients by surrounding glioma cells. Thus, GPIHBP1 expression in gliomas facilitates TRL processing and provides a source of lipid nutrients for glioma cells.
Collapse
Affiliation(s)
- Xuchen Hu
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Ken Matsumoto
- VIB-KU Leuven Center for Cancer Biology (CCB)LeuvenBelgium
| | - Rachel S Jung
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Thomas A Weston
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Patrick J Heizer
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Cuiwen He
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Norma P Sandoval
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Christopher M Allan
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Yiping Tu
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Harry V Vinters
- Department of Pathology and Laboratory Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Jonsson Comprehensive Cancer Center (JCCC), David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Rochelle M Ellison
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Jazmin E Morales
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Lynn J Baufeld
- Department of Molecular and Medical Pharmacology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Ahmanson Translational Imaging Division, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Nicholas A Bayley
- Department of Molecular and Medical Pharmacology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Ahmanson Translational Imaging Division, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck LaboratoryUppsala UniversityUppsalaSweden
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck LaboratoryUppsala UniversityUppsalaSweden
- Integrated Cardio Metabolic Centre (ICMC)Karolinska InstitutetHuddingeSweden
| | - Anne P Beigneux
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - David A Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Ahmanson Translational Imaging Division, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Holger Gerhardt
- VIB-KU Leuven Center for Cancer Biology (CCB)LeuvenBelgium
- Max Delbrück Center for Molecular MedicineBerlinGermany
| | - Stephen G Young
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Department of Human Genetics, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Loren G Fong
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Haibo Jiang
- Department of Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- School of Molecular SciencesUniversity of Western AustraliaPerthAustralia
| |
Collapse
|
22
|
Choung S, Kim JM, Joung KH, Lee ES, Kim HJ, Ku BJ. Epidermal growth factor receptor inhibition attenuates non-alcoholic fatty liver disease in diet-induced obese mice. PLoS One 2019; 14:e0210828. [PMID: 30735525 PMCID: PMC6368280 DOI: 10.1371/journal.pone.0210828] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 01/02/2019] [Indexed: 12/31/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the main causes of chronic liver disease. NAFLD begins with excessive lipid accumulation in the liver and progresses to nonalcoholic steatohepatitis (NASH) and cirrhosis. NAFLD is closely linked to dysregulated hepatic lipid metabolism. Although recent studies have reported that epidermal growth factor receptor (EGFR) signaling regulates lipid metabolism, the roles of EGFR and EGFR inhibitors as modulators of lipid metabolism are largely unknown. Here, we investigated whether inhibiting EGFR using the EGFR tyrosine kinase inhibitor (TKI) PD153035 improves NAFLD. Our results demonstrate that EGFR was activated in liver tissues from high fat diet (HFD)-induced NAFLD mice. Inhibiting EGFR using PD153035 significantly reduced phosphatidylinositol-3-kinase/protein kinase B signaling and sterol responsive elementary binding protein 1 and 2 expression, which prevented HFD-induced hepatic steatosis and hypercholesterolemia by reducing de novo lipogenesis and cholesterol synthesis and enhancing fatty acid oxidation. Additionally, inhibiting EGFR improved HFD-induced glucose intolerance. In conclusion, these results indicate that EGFR plays an important role in NAFLD and is a potential therapeutic target.
Collapse
Affiliation(s)
- Sorim Choung
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Ji Min Kim
- Department of Internal Medicine, College of Medicine, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Kyong Hye Joung
- Department of Internal Medicine, College of Medicine, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Eaum Seok Lee
- Department of Internal Medicine, College of Medicine, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Hyun Jin Kim
- Department of Internal Medicine, College of Medicine, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Bon Jeong Ku
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
- Department of Internal Medicine, College of Medicine, Chungnam National University School of Medicine, Daejeon, South Korea
| |
Collapse
|
23
|
Xie Q, Wu J, Du Z, Di N, Yan R, Pang H, Jin T, Zhang H, Wu Y, Zhang Y, Yao Z, Feng X. DCE-MRI in Human Gliomas: A Surrogate for Assessment of Invasive Hypoxia Marker HIF-1Α Based on MRI-Neuronavigation Stereotactic Biopsies. Acad Radiol 2019; 26:179-187. [PMID: 29754996 DOI: 10.1016/j.acra.2018.04.015] [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] [Received: 02/04/2018] [Revised: 03/31/2018] [Accepted: 04/12/2018] [Indexed: 12/15/2022]
Abstract
RATIONALE AND OBJECTIVES The purpose of this study was to correlate dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) parameters with data from a specific marker of hypoxia, hypoxia-inducible factor 1α (HIF-1α), in human gliomas on a point-to-point basis by using coregistered magnetic resonance imaging and frameless stereotactic biopsies. MATERIALS AND METHODS Thirty-four patients with treatment-naive gliomas underwent DCE, axial T1-weighted, T2-weighted, T2-weighted fluid acquisition of inversion recovery, and three-dimensional T1-weighted brain volume with gadolinium contrast enhancement sequences on a 3.0-T magnetic resonance scanner before stereotactic surgery. Quantitative perfusion indices such as endothelial transfer constant, fractional extravascular extracellular space volume, fractional plasma volume, and reflux rate were measured at corresponding stereotactic biopsy sites. Each sample was considered an independent measurement, and its histology grade was diagnosed. HIF-1α expression was quantified from the point-to-point biopsy tissues. Analyses of receiver operating characteristic curves were done for HIF-1α to discriminate different grades of glioma. To look for correlations between immunohistochemical parameters and DCE indices, Spearman's correlation coefficient was used. RESULTS Seventy biopsy samples from 34 subjects were included in the analysis. Mean immunoreactivity scores of HIF-1α were 2.75 ± 1.11 for grade II (n = 24), 6.20 ± 2.33 for grade III (n = 20), and 10.46 ± 2.42 for grade IV (n = 26). HIF-1α showed very good-to-excellent accuracy in discriminating grade II from III, III from IV, and II from IV (area under the curve = 0.838, 0.862, and 0.994, respectively). Endothelial transfer constant and fractional extravascular extracellular space volume showed a significantly positive correlation with HIF-1α expression (r = 0.686, P < .001; r = 0.549, P < .001, respectively). CONCLUSION Our study demonstrated HIF-1α to be a significant predictor of different grades of gliomas with high sensitivity and specificity. DCE-MRI is a useful, noninvasive imaging tool for quantitative evaluation of HIF-1α, and its parameters may be used as a surrogate for HIF-1α expression.
Collapse
|
24
|
Seow P, Wong JHD, Ahmad-Annuar A, Mahajan A, Abdullah NA, Ramli N. Quantitative magnetic resonance imaging and radiogenomic biomarkers for glioma characterisation: a systematic review. Br J Radiol 2018; 91:20170930. [PMID: 29902076 PMCID: PMC6319852 DOI: 10.1259/bjr.20170930] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 05/25/2018] [Accepted: 06/07/2018] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE: The diversity of tumour characteristics among glioma patients, even within same tumour grade, is a big challenge for disease outcome prediction. A possible approach for improved radiological imaging could come from combining information obtained at the molecular level. This review assembles recent evidence highlighting the value of using radiogenomic biomarkers to infer the underlying biology of gliomas and its correlation with imaging features. METHODS: A literature search was done for articles published between 2002 and 2017 on Medline electronic databases. Of 249 titles identified, 38 fulfilled the inclusion criteria, with 14 articles related to quantifiable imaging parameters (heterogeneity, vascularity, diffusion, cell density, infiltrations, perfusion, and metabolite changes) and 24 articles relevant to molecular biomarkers linked to imaging. RESULTS: Genes found to correlate with various imaging phenotypes were EGFR, MGMT, IDH1, VEGF, PDGF, TP53, and Ki-67. EGFR is the most studied gene related to imaging characteristics in the studies reviewed (41.7%), followed by MGMT (20.8%) and IDH1 (16.7%). A summary of the relationship amongst glioma morphology, gene expressions, imaging characteristics, prognosis and therapeutic response are presented. CONCLUSION: The use of radiogenomics can provide insights to understanding tumour biology and the underlying molecular pathways. Certain MRI characteristics that show strong correlations with EGFR, MGMT and IDH1 could be used as imaging biomarkers. Knowing the pathways involved in tumour progression and their associated imaging patterns may assist in diagnosis, prognosis and treatment management, while facilitating personalised medicine. ADVANCES IN KNOWLEDGE: Radiogenomics can offer clinicians better insight into diagnosis, prognosis, and prediction of therapeutic responses of glioma.
Collapse
Affiliation(s)
| | | | - Azlina Ahmad-Annuar
- Department of Biomedical Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Abhishek Mahajan
- Department of Radiodiagnosis and Imaging, Tata Memorial Hospital, Mumbai, India
| | - Nor Aniza Abdullah
- Department of Computer System and Technology, University of Malaya, Kuala Lumpur, Malaysia
| | | |
Collapse
|
25
|
Chi KC, Tsai WC, Wu CL, Lin TY, Hueng DY. An Adult Drosophila Glioma Model for Studying Pathometabolic Pathways of Gliomagenesis. Mol Neurobiol 2018; 56:4589-4599. [DOI: 10.1007/s12035-018-1392-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 10/11/2018] [Indexed: 11/28/2022]
|
26
|
Xu X, Meng Y, Li L, Xu P, Wang J, Li Z, Bian J. Overview of the Development of Glutaminase Inhibitors: Achievements and Future Directions. J Med Chem 2018; 62:1096-1115. [PMID: 30148361 DOI: 10.1021/acs.jmedchem.8b00961] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
It has been demonstrated that glutamine metabolism has become the main energy and building blocks supply for the growth and viability of a potentially large subset of malignant tumors. The glutamine metabolism often depends upon mitochondrial glutaminase (GLS) activity, which converts glutamine to glutamate and serves as a significant role for bioenergetic processes. Thus, recently, the GLS has become a key target for small molecule therapeutic intervention. Numerous medicinal chemistry studies are currently aimed at the design of novel and potent inhibitors for GLS, however, to date, only one compound (named CB-839) have entered clinical trials for the treatment of advanced solid tumors and hematological malignancies. The perspective summarizes the progress in the discovery and development of GLS inhibitors, including the potential binding site, biochemical techniques for inhibitor identification, and approaches for identifying small-molecule inhibitors, as well as future therapeutic perspectives in glutamine metabolism are also put forward in order to provide reference and rational for the drug discovery of novel and potent glutamine metabolism modulators.
Collapse
Affiliation(s)
- Xi Xu
- Department of Medicinal Chemistry , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , P. R. China
| | - Ying Meng
- Department of Medicinal Chemistry , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , P. R. China
| | - Lei Li
- Department of Medicinal Chemistry , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , P. R. China
| | - Pengfei Xu
- Department of Medicinal Chemistry , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , P. R. China
| | - Jubo Wang
- Department of Medicinal Chemistry , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , P. R. China
| | - Zhiyu Li
- Department of Medicinal Chemistry , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , P. R. China.,Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 21009 , P. R. China
| | - Jinlei Bian
- Department of Medicinal Chemistry , China Pharmaceutical University , 24 Tongjiaxiang , Nanjing 210009 , P. R. China.,Jiangsu Key Laboratory of Drug Design and Optimization , China Pharmaceutical University , Nanjing 21009 , P. R. China
| |
Collapse
|
27
|
Cheng C, Geng F, Cheng X, Guo D. Lipid metabolism reprogramming and its potential targets in cancer. Cancer Commun (Lond) 2018; 38:27. [PMID: 29784041 PMCID: PMC5993136 DOI: 10.1186/s40880-018-0301-4] [Citation(s) in RCA: 473] [Impact Index Per Article: 67.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/12/2018] [Indexed: 12/13/2022] Open
Abstract
Reprogramming of lipid metabolism is a newly recognized hallmark of malignancy. Increased lipid uptake, storage and lipogenesis occur in a variety of cancers and contribute to rapid tumor growth. Lipids constitute the basic structure of membranes and also function as signaling molecules and energy sources. Sterol regulatory element-binding proteins (SREBPs), a family of membrane-bound transcription factors in the endoplasmic reticulum, play a central role in the regulation of lipid metabolism. Recent studies have revealed that SREBPs are highly up-regulated in various cancers and promote tumor growth. SREBP cleavage-activating protein is a key transporter in the trafficking and activation of SREBPs as well as a critical glucose sensor, thus linking glucose metabolism and de novo lipid synthesis. Targeting altered lipid metabolic pathways has become a promising anti-cancer strategy. This review summarizes recent progress in our understanding of lipid metabolism regulation in malignancy, and highlights potential molecular targets and their inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Chunming Cheng
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH, 43210, USA
| | - Feng Geng
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH, 43210, USA
| | - Xiang Cheng
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH, 43210, USA
| | - Deliang Guo
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH, 43210, USA.
| |
Collapse
|
28
|
Wang G, Li Y, Yang Z, Xu W, Yang Y, Tan X. ROS mediated EGFR/MEK/ERK/HIF-1α Loop Regulates Glucose metabolism in pancreatic cancer. Biochem Biophys Res Commun 2018; 500:873-878. [PMID: 29702094 DOI: 10.1016/j.bbrc.2018.04.177] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 04/22/2018] [Indexed: 11/26/2022]
Abstract
To investigate the glycometabolism associated mechanism in invasion and metastasis of pancreatic cancer, We screened out genes involved in anaerobic glycolysis headed by HIF-1α,using pre-established a pair of pancreatic cancer cell lines. In this study, we further detected the glucose metabolism state not only in the cells but all also in two groups of patients with different SUVmax on 18F-FDG PET/CT. The data suggests that ROS mediated EGFR/MEK/ERK/HIF-1α loop is activated in high glucose metabolic samples both in vitro and in vivo: The increasing of HIF-1α expression is controlled by activation of EGFR/MEK/ERK pathway in hypoxia condition, HIF-1α inhibits excessive release of ROS, the reduction of ROS further activates EGFR to form a positive feedback loop. This difference is closely related to invasion and metastasis capacity of pancreatic cancer, and can be rescued by separate or combined inhibition of EGFR or HIF-1α in various degree. These results indicate a new clue to develop therapy of pancreatic cancer by regulating the glucose metabolism.
Collapse
Affiliation(s)
- Gang Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China; Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Yifeng Li
- 101K, The First Clinical Department of China Medical University, Shenyang, 110122, China.
| | - Zeyu Yang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Weina Xu
- Department of Radiology and Nuclear Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Yifan Yang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Xiaodong Tan
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
29
|
The mechanisms of malic enzyme 2 in the tumorigenesis of human gliomas. Oncotarget 2018; 7:41460-41472. [PMID: 27166188 PMCID: PMC5173072 DOI: 10.18632/oncotarget.9190] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 04/23/2016] [Indexed: 01/10/2023] Open
Abstract
The high level of resistance of glioblastoma multiforme (GBM) to currently used chemotherapies and other conventional therapies, its invasive characteristics and the presence of stem-like cells are the major factors that make the treatment of GBM difficult. Recent studies have demonstrated that the homeostasis of energy metabolism, glycolysis and mitochondrial oxidation of glucose are important for GBM cell growth and chemo-resistance. However, it is not clear which specific gene(s) are involved in the homeostasis of energy metabolism and invasiveness of GBM cells. We performed a preliminary analysis of data obtained from Gene Expression Omnibus profiles and determined that malic enzyme 2 (ME2) expression was positively associated with WHO grade in human primary gliomas. Hence, we evaluated the detailed working mechanisms of ME2 in human GBM cell processes, including proliferation, cell cycle, invasion, migration, ROS, and ATP production. Our data demonstrated that ME2 was involved in GBM growth, invasion and migration. ME2 has two cofactors, NAD+ or NADP+, which are used to produce NADH and NADPH for ATP production and ROS clearance, respectively. If the catalytic activity of ME2 is determined to be critical for its roles in GBM growth, invasion and migration, small molecule inhibitors of ME2 may be valuable drugs for GBM therapy. We hope that our current data provides a candidate treatment strategy for GBM.
Collapse
|
30
|
Chen M, Chai W, Song T, Ma M, Lian XY, Zhang Z. Anti-glioma Natural Products Downregulating Tumor Glycolytic Enzymes from Marine Actinomycete Streptomyces sp. ZZ406. Sci Rep 2018; 8:72. [PMID: 29311676 PMCID: PMC5758648 DOI: 10.1038/s41598-017-18484-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/12/2017] [Indexed: 12/15/2022] Open
Abstract
Marine natural products are important resources for discovering novel anticancer drugs. In this study, an extract prepared from the culture of a sea anemone-derived actinomycete Streptomyces sp. ZZ406 in soluble starch and casein-related liquid medium was found to have activity in inhibiting the proliferation of glioma cells and reducing the production of lactate in glioma cells. Chemical investigation of this active crude extract resulted in the isolation of four new compounds and seven known ones. Structures of the new compounds were determined by a combination of extensive NMR analyses, HRESIMS and MS-MS data, electronic circular dichroism calculation, chemical degradation, and Marfey's method. New compound 1 showed potent activity against the proliferation of different glioma cells with IC50 values of 4.7 to 8.1 μM, high selectivity index (>12.3 to 21.3), and good stability in human liver microsomes. Western blot analysis revealed that compound 1 remarkably downregulated the expressions of several important glioma glycolytic enzymes. The data from this study suggested that compound 1 might have potential as a novel anti-glioma agent to be further investigated.
Collapse
Affiliation(s)
- Mengxuan Chen
- Ocean College, Zhoushan Campus, Zhejiang University, Zhoushan, 316021, China
| | - Weiyun Chai
- Ocean College, Zhoushan Campus, Zhejiang University, Zhoushan, 316021, China
| | - Tengfei Song
- Ocean College, Zhoushan Campus, Zhejiang University, Zhoushan, 316021, China
| | - Mingzhu Ma
- Ocean College, Zhoushan Campus, Zhejiang University, Zhoushan, 316021, China
| | - Xiao-Yuan Lian
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Zhizhen Zhang
- Ocean College, Zhoushan Campus, Zhejiang University, Zhoushan, 316021, China.
| |
Collapse
|
31
|
Anna I, Bartosz P, Lech P, Halina A. Novel strategies of Raman imaging for brain tumor research. Oncotarget 2017; 8:85290-85310. [PMID: 29156720 PMCID: PMC5689610 DOI: 10.18632/oncotarget.19668] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 04/29/2017] [Indexed: 01/07/2023] Open
Abstract
Raman diagnostics and imaging have been shown to be an effective tool for the analysis and discrimination of human brain tumors from normal structures. Raman spectroscopic methods have potential to be applied in clinical practice as they allow for identification of tumor margins during surgery. In this study, we investigate medulloblastoma (grade IV WHO) (n= 5), low-grade astrocytoma (grades I-II WHO) (n =4), ependymoma (n=3) and metastatic brain tumors (n= 1) and the tissue from the negative margins used as normal controls. We compare a high grade medulloblastoma, low grade astrocytoma and non-tumor samples from human central nervous system (CNS) tissue. Based on the properties of the Raman vibrational features and Raman images we provide a real–time feedback method that is label-free to monitor tumor metabolism that reveals reprogramming of biosynthesis of lipids, proteins, DNA and RNA. Our results indicate marked metabolic differences between low and high grade brain tumors. We discuss molecular mechanisms causing these metabolic changes, particularly lipid alterations in malignant medulloblastoma and low grade gliomas that may shed light on the mechanisms driving tumor recurrence thereby revealing new approaches for the treatment of malignant glioma. We have found that the high-grade tumors of central nervous system (medulloblastoma) exhibit enhanced level of β-sheet conformation and down-regulated level of α-helix conformation when comparing against normal tissue. We have found that almost all tumors studied in the paper have increased Raman signals of nucleic acids. This increase can be interpreted as increased DNA/RNA turnover in brain tumors. We have shown that the ratio of Raman intensities I2930/I2845 at 2930 and 2845 cm-1 is a good source of information on the ratio of lipid and protein contents. We have found that the ratio reflects the different lipid and protein contents of cancerous brain tissue compared to the non-tumor tissue. We found that levels of the saturated fatty acids were significantly reduced in the high grade medulloblastoma samples compared with non-tumor brain samples and low grade astrocytoma. Differences were also noted in the n-6/n-3 polyunsaturated fatty acids (PUFA) content between medulloblastoma and non-tumor brain samples. The content of the oleic acid (OA) was significantly smaller in almost all brain high grade brain tumors than that observed in the control samples. It indicates that the fatty acid composition of human brain tumors differs from that found in non-tumor brain tissue. The iodine number NI for the normal brain tissue is 60. For comparison OA has 87, docosahexaenoic acid (DHA) 464, α-linolenic acid (ALA) 274. The high grade tumors have the iodine numbers between that for palmitic acid, stearic acid, arachidic acid (NI=0) and oleic acid (NI=87). Most low grade tumors have NI similar to that of OA. The iodine number for arachidonic acid (AA) (NI=334) is much higher than those observed for all studied samples.
Collapse
Affiliation(s)
- Imiela Anna
- Lodz University of Technology, Institute of Applied Radiation Chemistry, Laboratory of Laser Molecular Spectroscopy, 93-590 Lodz, Poland
| | - Polis Bartosz
- Polish Mother's Memorial Hospital Research Institute, Department of Neurosurgery and Neurotraumatology, 3-338 Lodz, Poland
| | - Polis Lech
- Polish Mother's Memorial Hospital Research Institute, Department of Neurosurgery and Neurotraumatology, 3-338 Lodz, Poland
| | - Abramczyk Halina
- Lodz University of Technology, Institute of Applied Radiation Chemistry, Laboratory of Laser Molecular Spectroscopy, 93-590 Lodz, Poland
| |
Collapse
|
32
|
Beyer S, Fleming J, Meng W, Singh R, Haque SJ, Chakravarti A. The Role of miRNAs in Angiogenesis, Invasion and Metabolism and Their Therapeutic Implications in Gliomas. Cancers (Basel) 2017; 9:cancers9070085. [PMID: 28698530 PMCID: PMC5532621 DOI: 10.3390/cancers9070085] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/01/2017] [Accepted: 07/03/2017] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding, endogenous RNA molecules that function in gene silencing by post-transcriptional regulation of gene expression. The dysregulation of miRNA plays a pivotal role in cancer tumorigenesis, including the development and progression of gliomas. Their small size, stability and ability to target multiple oncogenes have simultaneously distinguished miRNAs as attractive candidates for biomarkers and novel therapeutic targets for glioma patients. In this review, we summarize the most frequently cited miRNAs known to contribute to gliomagenesis and progression by regulating the defining hallmarks of gliomas, including angiogenesis, invasion, and cell metabolism. We also discuss their promising potential as prognostic and predictive biomarkers and novel therapeutic targets, in addition to the challenges that must be overcome before their translation from bench to bedside.
Collapse
Affiliation(s)
- Sasha Beyer
- Department of Radiation Oncology, the Ohio State University Comprehensive Cancer Center & Arthur, G. James Cancer Hospital, Columbus, OH 43012, USA.
| | - Jessica Fleming
- Department of Radiation Oncology, the Ohio State University Comprehensive Cancer Center & Arthur, G. James Cancer Hospital, Columbus, OH 43012, USA.
| | - Wei Meng
- Department of Radiation Oncology, the Ohio State University Comprehensive Cancer Center & Arthur, G. James Cancer Hospital, Columbus, OH 43012, USA.
| | - Rajbir Singh
- Department of Radiation Oncology, the Ohio State University Comprehensive Cancer Center & Arthur, G. James Cancer Hospital, Columbus, OH 43012, USA.
| | - S Jaharul Haque
- Department of Radiation Oncology, the Ohio State University Comprehensive Cancer Center & Arthur, G. James Cancer Hospital, Columbus, OH 43012, USA.
| | - Arnab Chakravarti
- Department of Radiation Oncology, the Ohio State University Comprehensive Cancer Center & Arthur, G. James Cancer Hospital, Columbus, OH 43012, USA.
| |
Collapse
|
33
|
Association between epidermal growth factor receptor amplification and ADP-ribosylation factor 1 methylation in human glioblastoma. Cell Oncol (Dordr) 2017. [PMID: 28631186 DOI: 10.1007/s13402-017-0329-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
PURPOSE Glioblastoma (GB) is the most frequent and most malignant primary brain tumor in adults. Previously, it has been found that both genetic and epigenetic factors may play critical roles in its etiology and prognosis. In addition, it has been found that the epidermal growth factor receptor gene (EGFR) is frequently over-expressed and amplified in primary GBs. Here, we assessed the promoter methylation status of 10 genes relevant to GB and explored associations between these findings and the EGFR gene amplification status. METHODS Tumor samples were obtained from 36 patients with primary GBs. In addition, 6 control specimens were included from patients who were operated for diseases other than brain tumors. The amplification status of the EGFR gene, and its deletion mutant EGFRvIII, were evaluated using FISH and MLPA, respectively. The IDH1/2 gene mutation status was verified using Sanger sequencing. A commercial DNA methylation kit was used to assess the promoter methylation status of 10 pre-selected genes. Metabolic profiles were measured using HR-MAS NMR spectroscopy. The EGFR and ARF1 mRNA expression levels were quantified using qRT-PCR. RESULTS Of the 10 genes analyzed, we found that only ARF1 promoter hypermethylation was significantly associated with EGFR gene amplification. ARF1 is a GTPase that is involved in vesicle trafficking and the Golgi apparatus. Subsequent tumor metabolism measurements revealed a positive association between EGFR amplification and different membrane precursors and methyl-donor metabolites. Finally, we found that EGFR gene amplifications were associated with distinct tumor infiltration patterns, thus representing a putative novel functional association between EGFR gene amplification and ARF1 gene promoter methylation in GB. CONCLUSIONS The results reported here provide a basis for a new hypotheses connecting EGFR gene amplification in GB cells with ARF1 gene promoter methylation, vesicle trafficking, membrane turnover and tumor metabolism. The mechanism(s) underlying these connections and their functional consequences remain to be established.
Collapse
|
34
|
Geng F, Guo D. Lipid droplets, potential biomarker and metabolic target in glioblastoma. INTERNAL MEDICINE REVIEW (WASHINGTON, D.C. : ONLINE) 2017; 3:10.18103/imr.v3i5.443. [PMID: 29034362 PMCID: PMC5639724 DOI: 10.18103/imr.v3i5.443] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lipid droplets (LDs) are subcellular organelles that store large amounts of the neutral lipids, triglycerides (TG) and/or cholesteryl esters (CE). LDs are commonly formed in adipocytes, liver cells and macrophages, and their formation has been shown to be associated with the progression of metabolic diseases, i.e., obesity, fatty liver and atherosclerosis. Interestingly, LDs are also found in some tumor tissues. We recently showed that LDs are prevalent in glioblastoma (GBM), the most deadly brain tumor, but are not detectable in low-grade gliomas and normal brain tissues, suggesting that LDs may serve as a novel diagnostic biomarker for GBM. This short review will briefly introduce LD biology, summarize recent observations about LDs in several types of cancer tissues, and discuss LD formation in GBM. Moreover, we will highlight the role of SOAT1 (sterol-O transferase 1), a key enzyme regulating CE synthesis and LD formation in GBM, in the regulation of SREBP (sterol regulatory-element binding protein) activation. The therapeutic potential of LDs and SOAT1 will be discussed.
Collapse
Affiliation(s)
- Feng Geng
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH 43210, USA
| | - Deliang Guo
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH 43210, USA
| |
Collapse
|
35
|
Role of ketogenic metabolic therapy in malignant glioma: A systematic review. Crit Rev Oncol Hematol 2017; 112:41-58. [DOI: 10.1016/j.critrevonc.2017.02.016] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/30/2017] [Accepted: 02/14/2017] [Indexed: 12/22/2022] Open
|
36
|
Ru P, Guo D. microRNA-29 mediates a novel negative feedback loop to regulate SCAP/SREBP-1 and lipid metabolism. RNA & DISEASE 2017; 4. [PMID: 28664184 DOI: 10.14800/rd.1525] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The membrane-bound transcription factors, SREBPs (sterol regulatory element-binding proteins), play a central role in regulating lipid metabolism. The transcriptional activation of SREBPs requires the key protein SCAP (SREBP-cleavage activating protein) to translocate their precursors from the endoplasmic reticulum to the Golgi for subsequent proteolytic activation, a process tightly regulated by a cholesterol-mediated negative feedback loop. Our previous work showed that the SCAP/SREBP-1 pathway is significantly upregulated in human glioblastoma (GBM), the most deadly brain cancer, and that glucose-mediated N-glycosylation of SCAP is a prerequisite step for SCAP/SREBP trafficking. More recently, we demonstrated that microRNA-29 (miR-29) mediates a previously unrecognized negative feedback loop in SCAP/SREBP-1 signaling to control lipid metabolism. We found that SREBP-1, functioning as a transcription factor, promotes the expression of the miR-29 family members, miR-29a, -29b and -29c. In turn, the miR-29 isoforms reversely repress the expression of SCAP and SREBP-1. Moreover, treatment with miR-29 mimics effectively suppressed GBM tumor growth by inhibiting SCAP/SREBP-1 and de novo lipid synthesis. These findings, recently published in Cell Reports, strongly suggest that delivery of miR-29 in vivo may be a promising approach to treat cancer and metabolic diseases by suppressing SCAP/SREBP-1-regulated lipid metabolism.
Collapse
Affiliation(s)
- Peng Ru
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH 43210, USA
| | - Deliang Guo
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, OH 43210, USA
| |
Collapse
|
37
|
Ye X, Anjum K, Song T, Wang W, Liang Y, Chen M, Huang H, Lian XY, Zhang Z. Antiproliferative cyclodepsipeptides from the marine actinomycete Streptomyces sp. P11-23B downregulating the tumor metabolic enzymes of glycolysis, glutaminolysis, and lipogenesis. PHYTOCHEMISTRY 2017; 135:151-159. [PMID: 28049552 PMCID: PMC7111624 DOI: 10.1016/j.phytochem.2016.12.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 12/07/2016] [Accepted: 12/13/2016] [Indexed: 05/16/2023]
Abstract
Two cyclodepsipeptides and a known cyclodepsipeptide valinomycin were isolated from a culture of the marine actinomycete Streptomyces sp. P11-23B. Their structures were established based on NMR, HRESIMS, and MS-MS spectroscopic interpretation as well as by chemical degradation. Both streptodepsipeptides P11A and P11B inhibited proliferation of different glioma cell lines, with IC50 values ranging from 0.1 μM to 1.4 μM. Streptodepsipeptide P11A was found to block the cell cycle at the G0/G1 phase and induce apoptosis in glioma cells. Further investigation demonstrated that streptodepsipeptide P11A downregulated expression of HK2, PFKFB3, PKM2, GLS, and FASN, important tumor metabolic enzymes. Data from this study suggested that targeting multiple tumor metabolic regulators might be one anti-glioma mechanism of streptodepsipeptide P11A. A possible mechanism for this class of streptodepsipeptides is reported herein.
Collapse
Affiliation(s)
- Xuewei Ye
- Ocean College, Zhoushan Campus, Zhejiang University, Zhoushan 316021, China
| | - Komal Anjum
- Ocean College, Zhoushan Campus, Zhejiang University, Zhoushan 316021, China
| | - Tengfei Song
- Ocean College, Zhoushan Campus, Zhejiang University, Zhoushan 316021, China
| | - Wenling Wang
- Ocean College, Zhoushan Campus, Zhejiang University, Zhoushan 316021, China
| | - Ying Liang
- Ocean College, Zhoushan Campus, Zhejiang University, Zhoushan 316021, China
| | - Mengxuan Chen
- Ocean College, Zhoushan Campus, Zhejiang University, Zhoushan 316021, China
| | - Haocai Huang
- Ocean College, Zhoushan Campus, Zhejiang University, Zhoushan 316021, China
| | - Xiao-Yuan Lian
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Zhizhen Zhang
- Ocean College, Zhoushan Campus, Zhejiang University, Zhoushan 316021, China.
| |
Collapse
|
38
|
Cheng C, Guo JY, Geng F, Wu X, Cheng X, Li Q, Guo D. Analysis of SCAP N-glycosylation and Trafficking in Human Cells. J Vis Exp 2016. [PMID: 27911384 DOI: 10.3791/54709] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Elevated lipogenesis is a common characteristic of cancer and metabolic diseases. Sterol regulatory element-binding proteins (SREBPs), a family of membrane-bound transcription factors controlling the expression of genes important for the synthesis of cholesterol, fatty acids and phospholipids, are frequently upregulated in these diseases. In the process of SREBP nuclear translocation, SREBP-cleavage activating protein (SCAP) plays a central role in the trafficking of SREBP from the endoplasmic reticulum (ER) to the Golgi and in subsequent proteolysis activation. Recently, we uncovered that glucose-mediated N-glycosylation of SCAP is a prerequisite condition for the exit of SCAP/SREBP from the ER and movement to the Golgi. N-glycosylation stabilizes SCAP and directs SCAP/SREBP trafficking. Here, we describe a protocol for the isolation of membrane fractions in human cells and for the preparation of the samples for the detection of SCAP N-glycosylation and total protein by using western blot. We further provide a method to monitor SCAP trafficking by using confocal microscopy. This protocol is appropriate for the investigation of SCAP N-glycosylation and trafficking in mammalian cells.
Collapse
Affiliation(s)
- Chunming Cheng
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center and College of Medicine
| | - Jeffrey Yunhua Guo
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center and College of Medicine
| | - Feng Geng
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center and College of Medicine
| | - Xiaoning Wu
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center and College of Medicine
| | - Xiang Cheng
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center and College of Medicine
| | - Qiyue Li
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center and College of Medicine
| | - Deliang Guo
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center and College of Medicine;
| |
Collapse
|
39
|
Geng F, Cheng X, Wu X, Yoo JY, Cheng C, Guo JY, Mo X, Ru P, Hurwitz B, Kim SH, Otero J, Puduvalli V, Lefai E, Ma J, Nakano I, Horbinski C, Kaur B, Chakravarti A, Guo D. Inhibition of SOAT1 Suppresses Glioblastoma Growth via Blocking SREBP-1-Mediated Lipogenesis. Clin Cancer Res 2016; 22:5337-5348. [PMID: 27281560 PMCID: PMC5093025 DOI: 10.1158/1078-0432.ccr-15-2973] [Citation(s) in RCA: 219] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 05/27/2016] [Indexed: 12/17/2022]
Abstract
PURPOSE Elevated lipogenesis regulated by sterol regulatory element-binding protein-1 (SREBP-1), a transcription factor playing a central role in lipid metabolism, is a novel characteristic of glioblastoma (GBM). The aim of this study was to identify effective approaches to suppress GBM growth by inhibition of SREBP-1. As SREBP activation is negatively regulated by endoplasmic reticulum (ER) cholesterol, we sought to determine whether suppression of sterol O-acyltransferase (SOAT), a key enzyme converting ER cholesterol to cholesterol esters (CE) to store in lipid droplets (LDs), effectively suppressed SREBP-1 and blocked GBM growth. EXPERIMENTAL DESIGN The presence of LDs in glioma patient tumor tissues was analyzed using immunofluorescence, immunohistochemistry, and electronic microscopy. Western blotting and real-time PCR were performed to analyze protein levels and gene expression of GBM cells, respectively. Intracranial GBM xenografts were used to determine the effects of genetically silencing SOAT1 and SREBP-1 on tumor growth. RESULTS Our study unraveled that cholesterol esterification and LD formation are signature of GBM, and human patients with glioma possess elevated LDs that correlate with GBM progression and poor survival. We revealed that SOAT1 is highly expressed in GBM and functions as a key player in controlling the cholesterol esterification and storage in GBM. Targeting SOAT1 suppresses GBM growth and prolongs survival in xenograft models via inhibition of SREBP-1-regulated lipid synthesis. CONCLUSIONS Cholesterol esterification and storage in LDs are novel characteristics of GBM, and inhibiting SOAT1 to block cholesterol esterification is a promising therapeutic strategy to treat GBM by suppressing SREBP-1. Clin Cancer Res; 22(21); 5337-48. ©2016 AACR.
Collapse
Affiliation(s)
- Feng Geng
- Department of Radiation Oncology, James Comprehensive Cancer Center & Arthur G James Cancer Hospital, The Ohio State Medical Center, Columbus, Ohio
| | - Xiang Cheng
- Department of Radiation Oncology, James Comprehensive Cancer Center & Arthur G James Cancer Hospital, The Ohio State Medical Center, Columbus, Ohio
| | - Xiaoning Wu
- Department of Radiation Oncology, James Comprehensive Cancer Center & Arthur G James Cancer Hospital, The Ohio State Medical Center, Columbus, Ohio
| | - Ji Young Yoo
- Department of Neurosurgery, James Comprehensive Cancer Center & Arthur G James Cancer Hospital, The Ohio State Medical Center, Columbus, Ohio
| | - Chunming Cheng
- Department of Radiation Oncology, James Comprehensive Cancer Center & Arthur G James Cancer Hospital, The Ohio State Medical Center, Columbus, Ohio
| | - Jeffrey Yunhua Guo
- Department of Radiation Oncology, James Comprehensive Cancer Center & Arthur G James Cancer Hospital, The Ohio State Medical Center, Columbus, Ohio
| | - Xiaokui Mo
- Center for Biostatistics, Department of Biomedical Informatics, James Comprehensive Cancer Center & Arthur G James Cancer Hospital, The Ohio State Medical Center, Columbus, Ohio
| | - Peng Ru
- Department of Radiation Oncology, James Comprehensive Cancer Center & Arthur G James Cancer Hospital, The Ohio State Medical Center, Columbus, Ohio
| | - Brian Hurwitz
- Department of Neurosurgery, James Comprehensive Cancer Center & Arthur G James Cancer Hospital, The Ohio State Medical Center, Columbus, Ohio
| | - Sung-Hak Kim
- Department of Neurosurgery at University Alabama at Birmingham, Alabama
| | - Jose Otero
- Department of Pathology, James Comprehensive Cancer Center & Arthur G James Cancer Hospital, The Ohio State Medical Center, Columbus, Ohio
| | - Vinay Puduvalli
- Department of Neurosurgery, James Comprehensive Cancer Center & Arthur G James Cancer Hospital, The Ohio State Medical Center, Columbus, Ohio
| | - Etienne Lefai
- CarMeN Laboratory, INSERM U1060, INRA 1397, Faculté de Médecine Lyon Sud, University de Lyon, Oullins, France
| | - Jianjie Ma
- Department of Surgery, James Comprehensive Cancer Center & Arthur G James Cancer Hospital, The Ohio State Medical Center, Columbus, Ohio
| | - Ichiro Nakano
- Department of Neurosurgery at University Alabama at Birmingham, Alabama
| | - Craig Horbinski
- Departments of Pathology and Neurosurgery at Northwestern University, Chicago, Illinois
| | - Balveen Kaur
- Department of Neurosurgery, James Comprehensive Cancer Center & Arthur G James Cancer Hospital, The Ohio State Medical Center, Columbus, Ohio
| | - Arnab Chakravarti
- Department of Radiation Oncology, James Comprehensive Cancer Center & Arthur G James Cancer Hospital, The Ohio State Medical Center, Columbus, Ohio
| | - Deliang Guo
- Department of Radiation Oncology, James Comprehensive Cancer Center & Arthur G James Cancer Hospital, The Ohio State Medical Center, Columbus, Ohio.
| |
Collapse
|
40
|
New Metabolites and Bioactive Actinomycins from Marine-Derived Streptomyces sp. ZZ338. Mar Drugs 2016; 14:md14100181. [PMID: 27727167 PMCID: PMC5082329 DOI: 10.3390/md14100181] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 09/20/2016] [Accepted: 09/21/2016] [Indexed: 11/16/2022] Open
Abstract
An extract prepared from the culture of a marine-derived actinomycete Streptomyces sp. ZZ338 was found to have significant antimicrobial and antiproliferative activities. A chemical investigation of this active extract resulted in the isolation of three known bioactive actinomycins (1–3) and two new metabolites (4 and 5). The structures of the isolated compounds were identified as actinomycins D (1), V (2), X0β (3), 2-acetylamino-3-hydroxyl-4-methyl-benzoic acid methyl ester (4), and N-1S-(4-methylaminophenylmethyl)-2-oxo-propyl acetamide (5) based on their nuclear magnetic resonance (NMR) and high resolution electrospray ionization mass spectroscopy (HRESIMS) data as well as their optical rotation. This class of new compound 5 had never before been found from a natural resource. Three known actinomycins showed activities in inhibiting the proliferation of glioma cells and the growth of methicillin-resistant Staphylococcus aureus, Escherichia coli, and Candida albicans and are responsible for the activity of the crude extract. Actinomycin D (1) was also found to downregulate several glioma metabolic enzymes of glycolysis, glutaminolysis, and lipogenesis, suggesting that targeting multiple tumor metabolic regulators might be a new anti-glioma mechanism of actinomycin D. This is the first report of such a possible mechanism for the class of actinomycins.
Collapse
|
41
|
Rais R, Jančařík A, Tenora L, Nedelcovych M, Alt J, Englert J, Rojas C, Le A, Elgogary A, Tan J, Monincová L, Pate K, Adams R, Ferraris D, Powell J, Majer P, Slusher BS. Discovery of 6-Diazo-5-oxo-l-norleucine (DON) Prodrugs with Enhanced CSF Delivery in Monkeys: A Potential Treatment for Glioblastoma. J Med Chem 2016; 59:8621-33. [PMID: 27560860 DOI: 10.1021/acs.jmedchem.6b01069] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The glutamine antagonist 6-diazo-5-oxo-l-norleucine (DON, 1) has shown robust anticancer efficacy in preclinical and clinical studies, but its development was halted due to marked systemic toxicities. Herein we demonstrate that DON inhibits glutamine metabolism and provides antitumor efficacy in a murine model of glioblastoma, although toxicity was observed. To enhance DON's therapeutic index, we utilized a prodrug strategy to increase its brain delivery and limit systemic exposure. Unexpectedly, simple alkyl ester-based prodrugs were ineffective due to chemical instability cyclizing to form a unique diazo-imine. However, masking both DON's amine and carboxylate functionalities imparted sufficient chemical stability for biological testing. While these dual moiety prodrugs exhibited rapid metabolism in mouse plasma, several provided excellent stability in monkey and human plasma. The most stable compound (5c, methyl-POM-DON-isopropyl-ester) was evaluated in monkeys, where it achieved 10-fold enhanced cerebrospinal fluid to plasma ratio versus DON. This strategy may provide a path to DON utilization in glioblastoma multiforme patients.
Collapse
Affiliation(s)
| | - Andrej Jančařík
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, v.v.i. , Flemingovo n. 2, 166 10 Prague 6, Czech Republic
| | - Lukáš Tenora
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, v.v.i. , Flemingovo n. 2, 166 10 Prague 6, Czech Republic
| | | | | | | | | | | | | | | | - Lenka Monincová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, v.v.i. , Flemingovo n. 2, 166 10 Prague 6, Czech Republic
| | | | | | - Dana Ferraris
- Department of Chemistry, McDaniel College , Westminster, Maryland 21157, United States
| | | | - Pavel Majer
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, v.v.i. , Flemingovo n. 2, 166 10 Prague 6, Czech Republic
| | | |
Collapse
|
42
|
Ru P, Hu P, Geng F, Mo X, Cheng C, Yoo JY, Cheng X, Wu X, Guo JY, Nakano I, Lefai E, Kaur B, Chakravarti A, Guo D. Feedback Loop Regulation of SCAP/SREBP-1 by miR-29 Modulates EGFR Signaling-Driven Glioblastoma Growth. Cell Rep 2016; 16:1527-1535. [PMID: 27477273 DOI: 10.1016/j.celrep.2016.07.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 06/06/2016] [Accepted: 07/06/2016] [Indexed: 01/06/2023] Open
Abstract
Dysregulated lipid metabolism is a characteristic of malignancies. Sterol regulatory element binding protein 1 (SREBP-1), a transcription factor playing a central role in lipid metabolism, is highly activated in malignancies. Here, we unraveled a link between miR-29 and the SCAP (SREBP cleavage-activating protein)/SREBP-1 pathway in glioblastoma (GBM) growth. Epidermal growth factor receptor (EGFR) signaling enhances miR-29 expression in GBM cells via upregulation of SCAP/SREBP-1, and SREBP-1 activates miR-29 expression via binding to specific sites in its promoter. In turn, miR-29 inhibits SCAP and SREBP-1 expression by interacting with their 3' UTRs. miR-29 transfection suppressed lipid synthesis and GBM cell growth, which were rescued by the addition of fatty acids or N-terminal SREBP-1 expression. Xenograft studies showed that miR-29 mimics significantly inhibit GBM growth and prolong the survival of GBM-bearing mice. Our study reveals a previously unrecognized negative feedback loop in SCAP/SREBP-1 signaling mediated by miR-29 and suggests that miR-29 treatment may represent an effective means to target GBM.
Collapse
Affiliation(s)
- Peng Ru
- Department of Radiation Oncology, James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Peng Hu
- Department of Radiation Oncology, James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Feng Geng
- Department of Radiation Oncology, James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Xiaokui Mo
- Center for Biostatistics, Department of Bioinformatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Chunming Cheng
- Department of Radiation Oncology, James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Ji Young Yoo
- Department of Neurosurgery, James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Xiang Cheng
- Department of Radiation Oncology, James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Xiaoning Wu
- Department of Radiation Oncology, James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jeffrey Yunhua Guo
- Department of Radiation Oncology, James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Department of Neurosurgery, James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Ichiro Nakano
- Department of Neurosurgery and Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Etienne Lefai
- CarMeN Laboratory, INSERM U1060, INRA 1397, Faculté de Médecine Lyon Sud BP 12, Université de Lyon, 69921 Oullins Cedex, France
| | - Balveen Kaur
- Department of Neurosurgery, James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Arnab Chakravarti
- Department of Radiation Oncology, James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Deliang Guo
- Department of Radiation Oncology, James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
43
|
Cimini A, d'Angelo M, Benedetti E, D'Angelo B, Laurenti G, Antonosante A, Cristiano L, Di Mambro A, Barbarino M, Castelli V, Cinque B, Cifone MG, Ippoliti R, Pentimalli F, Giordano A. Flavopiridol: An Old Drug With New Perspectives? Implication for Development of New Drugs. J Cell Physiol 2016; 232:312-322. [PMID: 27171480 DOI: 10.1002/jcp.25421] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/10/2016] [Indexed: 12/30/2022]
Abstract
Glioblastoma, the most common brain tumor, is characterized by high proliferation rate, invasion, angiogenesis, and chemo- and radio-resistance. One of most remarkable feature of glioblastoma is the switch toward a glycolytic energetic metabolism that leads to high glucose uptake and consumption and a strong production of lactate. Activation of several oncogene pathways like Akt, c-myc, and ras induces glycolysis and angiogenesis and acts to assure glycolysis prosecution, tumor proliferation, and resistance to therapy. Therefore, the high glycolytic flux depends on the overexpression of glycolysis-related genes resulting in an overproduction of pyruvate and lactate. Metabolism of glioblastoma thus represents a key issue for cancer research. Flavopiridol is a synthetic flavonoid that inhibits a wide range of Cyclin-dependent kinase, that has been demonstrate to inactivate glycogen phosphorylase, decreasing glucose availability for glycolysis. In this work the study of glucose metabolism upon flavopiridol treatment in the two different glioblastoma cell lines. The results obtained point towards an effect of flavopiridol in glycolytic cells, thus suggesting a possible new use of this compound or flavopiridol-derived formulations in combination with anti-proliferative agents in glioblastoma patients. J. Cell. Physiol. 232: 312-322, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy. .,National Institute for Nuclear Physics (INFN), Gran Sasso National Laboratory (LNGS), Assergi, Italy. .,Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, Pennsylvania.
| | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Barbara D'Angelo
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, Pennsylvania
| | - Giulio Laurenti
- Institute of Metabolism and System Research, University of Birmingham, Birmingham, United Kingdom
| | - Andrea Antonosante
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Loredana Cristiano
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Antonella Di Mambro
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Marcella Barbarino
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Benedetta Cinque
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Maria Grazia Cifone
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Francesca Pentimalli
- Department of Experimental Oncology, National Institute of Tumors "G. Pascale", Naples, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, Pennsylvania. .,Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy.
| |
Collapse
|
44
|
DUSP28 links regulation of Mucin 5B and Mucin 16 to migration and survival of AsPC-1 human pancreatic cancer cells. Tumour Biol 2016; 37:12193-12202. [PMID: 27230679 DOI: 10.1007/s13277-016-5079-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 05/15/2016] [Indexed: 12/20/2022] Open
Abstract
The prognosis of pancreatic cancer has not improved despite considerable and continuous effort. Dual-specificity phosphatase 28 (DUSP28) is highly expressed in human pancreatic cancers and exerts critical effects. However, knowledge of its function in pancreatic cancers is extremely limited. Here, we demonstrate the peculiar role of DUSP28 in pancreatic cancers. Analysis using the Gene Expression Omnibus public microarray database indicated higher DUSP28, MUC1, MUC4, MUC5B, MUC16 and MUC20 messenger RNA (mRNA) levels in pancreatic cancers compared with normal pancreas tissues. DUSP28 expression in human pancreatic cancer correlated positively with those of MUC1, MUC4, MUC5B, MUC16 and MUC20. In contrast, there were no significant correlations between DUSP28 and mucins in normal pancreas tissues. Decreased DUSP28 expression resulted in down-regulation of MUC5B and MUC16 at both the mRNA and protein levels; furthermore, transfection with small interfering RNA (siRNA) for MUC5B and MUC16 inhibited the migration and survival of AsPC-1 cells. In addition, transfection of siRNA for MUC5B and MUC16 resulted in a significant decrease in phosphorylation of FAK and ERK1/2 compared with transfection with scrambled-siRNA. These results collectively indicate unique links between DUSP28 and MUC5B/MUC16 and their roles in pancreatic cancer; moreover, they strongly support a rationale for targeting DUSP28 to inhibit development of malignant pancreatic cancer.
Collapse
|
45
|
Özcan E, Çakır T. Reconstructed Metabolic Network Models Predict Flux-Level Metabolic Reprogramming in Glioblastoma. Front Neurosci 2016; 10:156. [PMID: 27147948 PMCID: PMC4834348 DOI: 10.3389/fnins.2016.00156] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/26/2016] [Indexed: 12/12/2022] Open
Abstract
Developments in genome scale metabolic modeling techniques and omics technologies have enabled the reconstruction of context-specific metabolic models. In this study, glioblastoma multiforme (GBM), one of the most common and aggressive malignant brain tumors, is investigated by mapping GBM gene expression data on the growth-implemented brain specific genome-scale metabolic network, and GBM-specific models are generated. The models are used to calculate metabolic flux distributions in the tumor cells. Metabolic phenotypes predicted by the GBM-specific metabolic models reconstructed in this work reflect the general metabolic reprogramming of GBM, reported both in in-vitro and in-vivo experiments. The computed flux profiles quantitatively predict that major sources of the acetyl-CoA and oxaloacetic acid pool used in TCA cycle are pyruvate dehydrogenase from glycolysis and anaplerotic flux from glutaminolysis, respectively. Also, our results, in accordance with recent studies, predict a contribution of oxidative phosphorylation to ATP pool via a slightly active TCA cycle in addition to the major contributor aerobic glycolysis. We verified our results by using different computational methods that incorporate transcriptome data with genome-scale models and by using different transcriptome datasets. Correct predictions of flux distributions in glycolysis, glutaminolysis, TCA cycle and lipid precursor metabolism validate the reconstructed models for further use in future to simulate more specific metabolic patterns for GBM.
Collapse
Affiliation(s)
- Emrah Özcan
- Computational Systems Biology Group, Department of Bioengineering, Gebze Technical University Gebze, Turkey
| | - Tunahan Çakır
- Computational Systems Biology Group, Department of Bioengineering, Gebze Technical University Gebze, Turkey
| |
Collapse
|
46
|
Bhattacharya B, Mohd Omar MF, Soong R. The Warburg effect and drug resistance. Br J Pharmacol 2016; 173:970-9. [PMID: 26750865 PMCID: PMC4793921 DOI: 10.1111/bph.13422] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 12/17/2015] [Accepted: 12/22/2015] [Indexed: 12/15/2022] Open
Abstract
: The Warburg effect describes the increased utilization of glycolysis rather than oxidative phosphorylation by tumour cells for their energy requirements under physiological oxygen conditions. This effect has been the basis for much speculation on the survival advantage of tumour cells, tumourigenesis and the microenvironment of tumours. More recently, studies have begun to reveal how the Warburg effect could influence drug efficacy and how our understanding of tumour energetics could be exploited to improve drug development. In particular, evidence is emerging demonstrating how better modelling of the tumour metabolic microenvironment could lead to a better prediction of drug efficacy and the identification of new combination strategies. This review will provide details of the current understanding of the complex interplay between glucose metabolism and pharmacology and discuss opportunities for utilizing the Warburg effect in future drug development.
Collapse
Affiliation(s)
| | | | - Richie Soong
- Cancer Science Institute of SingaporeNational University of SingaporeSingapore
- Department of PathologyNational University of SingaporeSingapore
| |
Collapse
|
47
|
Abstract
We recently uncovered that glucose is a critical activator of sterol regulatory element-binding proteins (SREBPs). Glucose promotes SREBP-cleavage activating protein (SCAP)/SREBP complex trafficking from the ER to the Golgi and subsequent SREBP activation via N-glycosylation of SCAP. Our study also demonstrated that SCAP plays a critical role in tumor growth.
Collapse
Affiliation(s)
- Deliang Guo
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center, Columbus, Ohio 43210
| |
Collapse
|
48
|
Fogal V, Babic I, Chao Y, Pastorino S, Mukthavaram R, Jiang P, Cho YJ, Pingle SC, Crawford JR, Piccioni DE, Kesari S. Mitochondrial p32 is upregulated in Myc expressing brain cancers and mediates glutamine addiction. Oncotarget 2015; 6:1157-70. [PMID: 25528767 PMCID: PMC4359224 DOI: 10.18632/oncotarget.2708] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/08/2014] [Indexed: 12/22/2022] Open
Abstract
Metabolic reprogramming is a key feature of tumorigenesis that is controlled by oncogenes. Enhanced utilization of glucose and glutamine are the best-established hallmarks of tumor metabolism. The oncogene c-Myc is one of the major players responsible for this metabolic alteration. However, the molecular mechanisms involved in Myc-induced metabolic reprogramming are not well defined. Here we identify p32, a mitochondrial protein known to play a role in the expression of mitochondrial respiratory chain complexes, as a critical player in Myc-induced glutamine addiction. We show that p32 is a direct transcriptional target of Myc and that high level of Myc in malignant brain cancers correlates with high expression of p32. Attenuation of p32 expression reduced growth rate of glioma cells expressing Myc and impaired tumor formation in vivo. Loss of p32 in glutamine addicted glioma cells induced resistance to glutamine deprivation and imparted sensitivity to glucose withdrawal. Finally, we provide evidence that p32 expression contributes to Myc-induced glutamine addiction of cancer cells. Our findings suggest that Myc promotes the expression of p32, which is required to maintain sufficient respiratory capacity to sustain glutamine metabolism in Myc transformed cells.
Collapse
Affiliation(s)
- Valentina Fogal
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Ivan Babic
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Ying Chao
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Sandra Pastorino
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Rajesh Mukthavaram
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Pengfei Jiang
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, University of California San Diego, La Jolla, CA
| | | | - Sandeep C Pingle
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - John R Crawford
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, University of California San Diego, La Jolla, CA.,Division of Neuro-Oncology, Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - David E Piccioni
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, University of California San Diego, La Jolla, CA.,Division of Neuro-Oncology, Department of Neurosciences, University of California, San Diego, La Jolla, CA
| | - Santosh Kesari
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, University of California San Diego, La Jolla, CA.Division of Neuro-Oncology, Department of Neurosciences, University of California, San Diego, La Jolla, CA.,Translational Neuro-Oncology Laboratories, Moores Cancer Center, University of California San Diego, La Jolla, CA.Division of Neuro-Oncology, Department of Neurosciences, University of California, San Diego, La Jolla, CA
| |
Collapse
|
49
|
Cheng C, Ru P, Geng F, Liu J, Yoo JY, Wu X, Cheng X, Euthine V, Hu P, Guo JY, Lefai E, Kaur B, Nohturfft A, Ma J, Chakravarti A, Guo D. Glucose-Mediated N-glycosylation of SCAP Is Essential for SREBP-1 Activation and Tumor Growth. Cancer Cell 2015; 28:569-581. [PMID: 26555173 PMCID: PMC4643405 DOI: 10.1016/j.ccell.2015.09.021] [Citation(s) in RCA: 201] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 08/04/2015] [Accepted: 09/27/2015] [Indexed: 12/17/2022]
Abstract
Tumorigenesis is associated with increased glucose consumption and lipogenesis, but how these pathways are interlinked is unclear. Here, we delineate a pathway in which EGFR signaling, by increasing glucose uptake, promotes N-glycosylation of sterol regulatory element-binding protein (SREBP) cleavage-activating protein (SCAP) and consequent activation of SREBP-1, an ER-bound transcription factor with central roles in lipid metabolism. Glycosylation stabilizes SCAP and reduces its association with Insig-1, allowing movement of SCAP/SREBP to the Golgi and consequent proteolytic activation of SREBP. Xenograft studies reveal that blocking SCAP N-glycosylation ameliorates EGFRvIII-driven glioblastoma growth. Thus, SCAP acts as key glucose-responsive protein linking oncogenic signaling and fuel availability to SREBP-dependent lipogenesis. Targeting SCAP N-glycosylation may provide a promising means of treating malignancies and metabolic diseases.
Collapse
Affiliation(s)
- Chunming Cheng
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Peng Ru
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Feng Geng
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Junfeng Liu
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Ji Young Yoo
- Department of Neurosurgery, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Xiaoning Wu
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Xiang Cheng
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Vanessa Euthine
- CarMeN Laboratory, INSERM U1060, INRA 1397, Faculté de Médecine Lyon Sud BP 12, Université de Lyon, 69921 Oullins Cedex, France
| | - Peng Hu
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Jeffrey Yunhua Guo
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Etienne Lefai
- CarMeN Laboratory, INSERM U1060, INRA 1397, Faculté de Médecine Lyon Sud BP 12, Université de Lyon, 69921 Oullins Cedex, France
| | - Balveen Kaur
- Department of Neurosurgery, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Axel Nohturfft
- Vascular Biology Research Centre, St. George's University of London, London SW17 0RE, UK
| | - Jianjie Ma
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
| | - Arnab Chakravarti
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Deliang Guo
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA.
| |
Collapse
|
50
|
Çoban G, Mohan S, Kural F, Wang S, O'Rourke DM, Poptani H. Prognostic Value of Dynamic Susceptibility Contrast-Enhanced and Diffusion-Weighted MR Imaging in Patients with Glioblastomas. AJNR Am J Neuroradiol 2015; 36:1247-52. [PMID: 25836728 DOI: 10.3174/ajnr.a4284] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 12/14/2014] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Prediction of survival in patients with glioblastomas is important for individualized treatment planning. This study aimed to assess the prognostic utility of presurgical dynamic susceptibility contrast and diffusion-weighted imaging for overall survival in patients with glioblastoma. MATERIALS AND METHODS MR imaging data from pathologically proved glioblastomas between June 2006 to December 2013 in 58 patients (mean age, 62.7 years; age range, 22-89 years) were included in this retrospective study. Patients were divided into long survival (≥15 months) and short survival (<15 months) groups, depending on overall survival time. Patients underwent dynamic susceptibility contrast perfusion and DWI before surgery and were treated with chemotherapy and radiation therapy. The maximum relative cerebral blood volume and minimum mean diffusivity values were measured from the enhancing part of the tumor. RESULTS Maximum relative cerebral blood volume values in patients with short survival were significantly higher compared with those who demonstrated long survival (P < .05). No significant difference was observed in the minimum mean diffusivity between short and long survivors. Receiver operator curve analysis demonstrated that a maximum relative cerebral blood volume cutoff value of 5.79 differentiated patients with low and high survival with an area under the curve of 0.93, sensitivity of 0.89, and specificity of 0.90 (P < .001), while a minimum mean diffusivity cutoff value of 8.35 × 10(-4)mm(2)/s had an area under the curve of 0.55, sensitivity of 0.71, and specificity of 0.47 (P > .05) in separating the 2 groups. CONCLUSIONS Maximum relative cerebral blood volume may be used as a prognostic marker of overall survival in patients with glioblastomas.
Collapse
Affiliation(s)
- G Çoban
- From the Department of Radiology (G.Ç., F.K.), Baskent University School of Medicine, Ankara, Turkey Departments of Radiology (G.Ç., S.M., F.K., S.W., H.P.)
| | - S Mohan
- Departments of Radiology (G.Ç., S.M., F.K., S.W., H.P.)
| | - F Kural
- From the Department of Radiology (G.Ç., F.K.), Baskent University School of Medicine, Ankara, Turkey Departments of Radiology (G.Ç., S.M., F.K., S.W., H.P.)
| | - S Wang
- Departments of Radiology (G.Ç., S.M., F.K., S.W., H.P.)
| | - D M O'Rourke
- Neurosurgery (D.M.O.), University of Pennsylvania, Philadelphia, Pennsylvania
| | - H Poptani
- Departments of Radiology (G.Ç., S.M., F.K., S.W., H.P.)
| |
Collapse
|