151
|
Jiang T, Zhang G, Lou Z. Role of the Sterol Regulatory Element Binding Protein Pathway in Tumorigenesis. Front Oncol 2020; 10:1788. [PMID: 33014877 PMCID: PMC7506081 DOI: 10.3389/fonc.2020.01788] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 08/11/2020] [Indexed: 12/15/2022] Open
Abstract
Metabolic changes are a major feature of tumors, including various metabolic forms, such as energy, lipid, and amino acid metabolism. Sterol regulatory element binding proteins (SREBPs) are important modules in regulating lipid metabolism and play an essential role in metabolic diseases. In the previous decades, the regulatory range of SREBPs has been markedly expanded. It was found that SREBPs also played a critical role in tumor development. SREBPs are involved in energy supply, lipid supply, immune environment and inflammatory environment shaping in tumor cells, and as a protective umbrella to support the malignant proliferation of tumor cells. Natural medicine and traditional Chinese medicine, as an important part of drug therapy, demonstrates the multifaceted effects of SREBPs regulation. This review summarizes the core processes in the involvement of SREBPs in tumors and provides a comprehensive understanding of the pathways through which natural drugs target the SREBP pathway and regulate tumor progression.
Collapse
Affiliation(s)
- Tao Jiang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Guangji Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhaohuan Lou
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
152
|
Isocitrate dehydrogenase 1 mutation enhances 24(S)-hydroxycholesterol production and alters cholesterol homeostasis in glioma. Oncogene 2020; 39:6340-6353. [PMID: 32855525 DOI: 10.1038/s41388-020-01439-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/10/2020] [Accepted: 08/17/2020] [Indexed: 01/08/2023]
Abstract
Isocitrate dehydrogenase (IDH) mutation is the most important initiating event in gliomagenesis, and the increasing evidence shows that IDH mutation is associated with the metabolic reprogramming in the tumor. Dysregulated cholesterol metabolism is a hallmark of tumor cells, but the cholesterol homeostasis in IDH-mutated glioma is still unknown. In this study, we found that astrocyte-specific mutant IDH1(R132H) knockin reduced the cholesterol contents and damaged the structure of myelin in mouse brains. In U87 and U251 cells, the expression of mutant IDH1 consistently reduced the cholesterol levels. Furthermore, we found that IDH1 mutation enhanced the production of 24(S)-hydroxycholesterol (24-OHC), which is not only the metabolite of cholesterol elimination, but also functions as an endogenous ligand for the liver X receptors (LXRs). In IDH1-mutant glioma cells, the elevated 24-OHC activated LXRs, which consequently accelerated the low-density lipoprotein receptor (LDLR) degradation by upregulating the inducible degrader of the LDLR (IDOL). The reduced LDLR expressions in IDH1-mutant glioma cells abated the uptakes of low-density lipoprotein (LDL) to decrease the cholesterol influx. In addition, the activated LXRs also promoted the cholesterol efflux by elevating the ATP-binding cassette transporter A1 (ABCA1), ABCG1, and apolipoprotein E (ApoE) in both IDH1-mutant astrocytes and glioma cells. As a feedback, the reduced cholesterol levels stimulated the cholesterol biosynthesis, which made IDH1-mutated glioma cells more sensitive to atorvastatin, an inhibitor of 3-hydroxy-3-methylglutaryl-CoA reductase. The altered cholesterol homeostasis regulated by mutant IDH provides a pivotal therapeutical strategy for the IDH-mutated gliomas.
Collapse
|
153
|
Pontini L, Marinozzi M. Shedding light on the roles of liver X receptors in cancer by using chemical probes. Br J Pharmacol 2020; 178:3261-3276. [PMID: 32673401 DOI: 10.1111/bph.15200] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/01/2020] [Accepted: 07/06/2020] [Indexed: 12/19/2022] Open
Abstract
Nuclear receptors, liver X receptor-α (LXRα; NR1H3) and liver X receptor-β (LXRβ; NR1H2), are considered master regulators of lipid homeostasis. During the last couple of decades, their pivotal roles in several physiological and pathological processes ranging from energy supply, immunity, cardiovascular, neurodegenerative disorders and cancer have been highlighted. In this review, the main results achieved during more recent years about our understanding of the LXR involvement in cancer has been mainly obtained using small-molecule chemical probes. Remarkably, all these probes, albeit having different structure and biological properties, have a well demonstrated anti-tumoral activity arising from LXR modulation, indicating a high potential of LXR targeting for the treatment of cancer. LINKED ARTICLES: This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc.
Collapse
Affiliation(s)
- Lorenzo Pontini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Maura Marinozzi
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| |
Collapse
|
154
|
Chandra NC. Atherosclerosis and carcinoma: Two facets of dysfunctional cholesterol homeostasis. J Biochem Mol Toxicol 2020; 34:e22595. [PMID: 32761975 DOI: 10.1002/jbt.22595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 07/04/2020] [Accepted: 07/23/2020] [Indexed: 12/11/2022]
Abstract
Although cholesterol is an essential and necessary component for biological systems; inappropriate accumulation of cholesterol in blood vessels and intracellular territory is also detrimental to living things. On one hand, cholesterol is the acting precursor of many metabolic regulators, a component of the structural veracity and scaffold fluidity of biomembranes, an insulator of electrical transmission in nerves and many more; on the other hand, its deposition in blood vessels induces atherosclerotic plaque and cardiovascular complications with the consequences of heart attack and stroke. It is also an emerging fact that cholesterol is a prelate in the cell nucleus for cell proliferation and any oddity in this venture may be the cause of tumorigenesis. Hence, cholesterol homeostasis is a very crucial element in issues of health management. Cholesterol is now a global target for maintaining quality health, particularly to control the two giants of the present world health tragedy: atherosclerosis and carcinoma, which appear to be the two facets of dysfunctional cholesterol homeostasis.
Collapse
Affiliation(s)
- Nimai C Chandra
- Department of Biochemistry, All India Institute of Medical Sciences, Patna, India
| |
Collapse
|
155
|
Cheng X, Geng F, Pan M, Wu X, Zhong Y, Wang C, Tian Z, Cheng C, Zhang R, Puduvalli V, Horbinski C, Mo X, Han X, Chakravarti A, Guo D. Targeting DGAT1 Ameliorates Glioblastoma by Increasing Fat Catabolism and Oxidative Stress. Cell Metab 2020; 32:229-242.e8. [PMID: 32559414 PMCID: PMC7415721 DOI: 10.1016/j.cmet.2020.06.002] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 04/15/2020] [Accepted: 06/01/2020] [Indexed: 12/21/2022]
Abstract
Glioblastoma (GBM), a mostly lethal brain tumor, acquires large amounts of free fatty acids (FAs) to promote cell growth. But how the cancer avoids lipotoxicity is unknown. Here, we identify that GBM upregulates diacylglycerol-acyltransferase 1 (DGAT1) to store excess FAs into triglycerides and lipid droplets. Inhibiting DGAT1 disrupted lipid homeostasis and resulted in excessive FAs moving into mitochondria for oxidation, leading to the generation of high levels of reactive oxygen species (ROS), mitochondrial damage, cytochrome c release, and apoptosis. Adding N-acetyl-cysteine or inhibiting FA shuttling into mitochondria decreased ROS and cell death induced by DGAT1 inhibition. We show in xenograft models that targeting DGAT1 blocked lipid droplet formation, induced tumor cell apoptosis, and markedly suppressed GBM growth. Together, our study demonstrates that DGAT1 upregulation protects GBM from oxidative damage and maintains lipid homeostasis by facilitating storage of excess FAs. Targeting DGAT1 could be a promising therapeutic approach for GBM.
Collapse
Affiliation(s)
- Xiang Cheng
- 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
| | - Meixia Pan
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Xiaoning Wu
- 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
| | - Chunyan Wang
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Zhihua Tian
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine at The Ohio State University, Columbus, OH 43210, USA
| | - Chunming Cheng
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine at The Ohio State University, Columbus, OH 43210, USA
| | - Rui Zhang
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine at The Ohio State University, Columbus, OH 43210, USA
| | - Vinay Puduvalli
- Division of Neuro-Oncology, Department of Neurology, The Ohio State University, Columbus, OH 43210, USA
| | - Craig Horbinski
- Departments of Pathology and Neurosurgery, Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA
| | - Xiaokui Mo
- Biostatistic Center and Department of Bioinformatics, College of Medicine at The Ohio State University, Columbus, OH 43210, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, 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
|
156
|
Zheng Y, Jin J, Gao Y, Luo C, Wu X, Liu J. Phospholipase Cε Regulates Prostate Cancer Lipid Metabolism and Proliferation by Targeting AMP-Activated Protein Kinase (AMPK)/Sterol Regulatory Element-Binding Protein 1 (SREBP-1) Signaling Pathway. Med Sci Monit 2020; 26:e924328. [PMID: 32696762 PMCID: PMC7392057 DOI: 10.12659/msm.924328] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Metabolic reprogramming is a common characteristic of numerous kinds of tumors, including prostate cancer (PCa). Tumor metabolism such as lipid metabolism provides sufficient lipids for tumor cell division and rapid growing as well as a vital source for formation of new cellular membranes. Phospholipase Cε (PLCε) is an oncogene that can drive proliferation, progression, and lipid metabolism of tumors, but its effect in lipid metabolism of PCa is not clear. MATERIAL AND METHODS Benign prostatic hyperplasia (BPH) and PCa tissue specimens were assessed for SREBP-1, FASN, and PLCε by immunohistochemistry, and PLCε was knocked-down by a lentiviral short hairpin RNA. The mRNA and protein level expression of related factors were tested by qPCR and Western blot analyses. Cell proliferation was assessed by clone formation, CCK-8, and Ki-67 assays. Nile red and oil red O staining were performed to detect endogenous lipid levels. Immunofluorescence was used to localize the protein of SREBP-1. Finally, a tumor xenograft assay of nude mice was performed to assess the role of PLCε in prostate tumor generation. RESULTS We found that overexpression of PLCε indicates low PFS in PCa and is involved in metastasis of PCa, and that the PLCε/AMPK/SREBP-1 signaling network promotes the progression of PCa through lipid metabolism in vivo and in vitro. CONCLUSIONS This study is the first to discover the lethal role of PLCε in lipid metabolism and malignant behavior of PCa, elucidation PCa occurrence and progression.
Collapse
Affiliation(s)
- Yongbo Zheng
- Department of Urology Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Jiajia Jin
- Chongqing University Cancer Hospital, Chongqing, China (mainland)
| | - Yingying Gao
- Department of Laboratory Diagnosis, Jiamusi University, Jiamusi, Heilongjiang, China (mainland)
| | - Chunli Luo
- College of Laboratory Medicine, Chongqing Medical University, Chongqing, China (mainland)
| | - Xiaohou Wu
- Department of Urology Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Jiayu Liu
- Department of Urology Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland)
| |
Collapse
|
157
|
Cholesterol metabolism: New functions and therapeutic approaches in cancer. Biochim Biophys Acta Rev Cancer 2020; 1874:188394. [PMID: 32698040 DOI: 10.1016/j.bbcan.2020.188394] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/08/2020] [Accepted: 07/12/2020] [Indexed: 02/05/2023]
Abstract
Cholesterol and its metabolites (precursors and derivatives) play an important role in cancer. In recent years, numerous studies have reported the functions of cholesterol metabolism in the regulation of tumor biological processes, especially oncogenic signaling pathways, ferroptosis, and tumor microenvironment. Preclinical studies have over the years indicated the inhibitory effects of blocking cholesterol synthesis and uptake on tumor formation and growth. Besides, some new cholesterol metabolic molecules such as SOAT1, SQLE, and NPC1 have recently emerged as promising drug targets for cancer treatment. Here, we systematically review the roles of cholesterol and its metabolites, and the latest advances in cancer therapy targeting cholesterol metabolism.
Collapse
|
158
|
Inhibition of HDAC1/2 Along with TRAP1 Causes Synthetic Lethality in Glioblastoma Model Systems. Cells 2020; 9:cells9071661. [PMID: 32664214 PMCID: PMC7407106 DOI: 10.3390/cells9071661] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 02/07/2023] Open
Abstract
The heterogeneity of glioblastomas, the most common primary malignant brain tumor, remains a significant challenge for the treatment of these devastating tumors. Therefore, novel combination treatments are warranted. Here, we showed that the combined inhibition of TRAP1 by gamitrinib and histone deacetylases (HDAC1/HDAC2) through romidepsin or panobinostat caused synergistic growth reduction of established and patient-derived xenograft (PDX) glioblastoma cells. This was accompanied by enhanced cell death with features of apoptosis and activation of caspases. The combination treatment modulated the levels of pro- and anti-apoptotic Bcl-2 family members, including BIM and Noxa, Mcl-1, Bcl-2 and Bcl-xL. Silencing of Noxa, BAK and BAX attenuated the effects of the combination treatment. At the metabolic level, the combination treatment led to an enhanced reduction of oxygen consumption rate and elicited an unfolded stress response. Finally, we tested whether the combination treatment of gamitrinib and panobinostat exerted therapeutic efficacy in PDX models of glioblastoma (GBM) in mice. While single treatments led to mild to moderate reduction in tumor growth, the combination treatment suppressed tumor growth significantly stronger than single treatments without induction of toxicity. Taken together, we have provided evidence that simultaneous targeting of TRAP1 and HDAC1/2 is efficacious to reduce tumor growth in model systems of glioblastoma.
Collapse
|
159
|
Ruiz CF, Montal ED, Haley JA, Bott AJ, Haley JD. SREBP1 regulates mitochondrial metabolism in oncogenic KRAS expressing NSCLC. FASEB J 2020; 34:10574-10589. [PMID: 32568455 DOI: 10.1096/fj.202000052r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/08/2020] [Accepted: 05/27/2020] [Indexed: 12/18/2022]
Abstract
Cancer cells require extensive metabolic reprograming in order to provide the bioenergetics and macromolecular precursors needed to sustain a malignant phenotype. Mutant KRAS is a driver oncogene that is well-known for its ability to regulate the ERK and PI3K signaling pathways. However, it is now appreciated that KRAS can promote the tumor growth via upregulation of anabolic metabolism. We recently reported that oncogenic KRAS promotes a gene expression program of de novo lipogenesis in non-small cell lung cancer (NSCLC). To define the mechanism(s) responsible, we focused on the lipogenic transcription factor SREBP1. We observed that KRAS increases SREBP1 expression and genetic knockdown of SREBP1 significantly inhibited the cell proliferation of mutant KRAS-expressing cells. Unexpectedly, lipogenesis was not significantly altered in cells subject to SREBP1 knockdown. Carbon tracing metabolic studies showed a significant decrease in oxidative phosphorylation and RNA-seq data revealed a significant decrease in mitochondrial encoded subunits of the electron transport chain (ETC). Taken together, these data support a novel role, distinct from lipogenesis, of SREBP1 on mitochondrial function in mutant KRAS NSCLC.
Collapse
Affiliation(s)
- Christian F Ruiz
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Emily D Montal
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John A Haley
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Alex J Bott
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - John D Haley
- Department of Pathology, Stony Brook University School of Medicine, Stony Brook, NY, USA
| |
Collapse
|
160
|
Floeth M, Elges S, Gerss J, Schwöppe C, Kessler T, Herold T, Wardelmann E, Berdel WE, Lenz G, Mikesch JH, Hartmann W, Schliemann C, Angenendt L. Low-density lipoprotein receptor (LDLR) is an independent adverse prognostic factor in acute myeloid leukaemia. Br J Haematol 2020; 192:494-503. [PMID: 32511755 DOI: 10.1111/bjh.16853] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 05/15/2020] [Indexed: 01/23/2023]
Abstract
The low-density lipoprotein receptor (LDLR) is a membrane receptor that mediates the endocytosis of low-density lipoprotein (LDL). Uptake of LDL has been proposed to contribute to chemotherapy resistance of acute myeloid leukaemia (AML) cell lines in vitro. In the present study, we analysed LDLR expression and survival using bone marrow biopsies from 187 intensively treated patients with AML. Here, increasing LDLR expression was associated with decreasing overall (58·4%, 44·2%, and 24·4%; P = 0·0018), as well as event-free survival (41·7%, 18·1%, and 14·3%; P = 0·0077), and an increasing cumulative incidence of relapse (33·9%, 55·1%, and 71·4%; P = 0·0011). Associations of LDLR expression with survival were confirmed in 557 intensively treated patients from two international validation cohorts. In the analytic and validation cohorts, LDLR expression remained associated with outcome in multivariable regression analyses including the European LeukemiaNet genetic risk classification. Thus, LDLR predicts outcome of patients with AML beyond existing risk factors. Furthermore, we found low expression levels of LDLR in most healthy tissues, suggesting it as a promising target for antibody-based pharmacodelivery approaches in AML.
Collapse
Affiliation(s)
- Matthias Floeth
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Sandra Elges
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Joachim Gerss
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
| | | | - Torsten Kessler
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Tobias Herold
- Laboratory for Leukemia Diagnostics, Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Eva Wardelmann
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Wolfgang E Berdel
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Georg Lenz
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | | | - Wolfgang Hartmann
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Münster, Germany
| | | | - Linus Angenendt
- Department of Medicine A, University Hospital Münster, Münster, Germany
| |
Collapse
|
161
|
Wu M, Li X, Wang S, Yang S, Zhao R, Xing Y, Liu L. Polydatin for treating atherosclerotic diseases: A functional and mechanistic overview. Biomed Pharmacother 2020; 128:110308. [PMID: 32480216 DOI: 10.1016/j.biopha.2020.110308] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/17/2020] [Accepted: 05/19/2020] [Indexed: 12/23/2022] Open
Abstract
With the advancement of science and technology, the living standards of human beings have continuously improved, but the incidence and mortality from atherosclerosis worldwide have also increased by year. Although interventional surgery and the continuous development of new drugs have significant therapeutic effects, their side effects cannot be ignored. Polydatin, an active ingredient isolated from the natural medicine Polygonum cuspidatum, has been shown to have a prominent role in the treatment of cardiovascular diseases. Polydatin treats atherosclerosis mainly from three aspects: anti-inflammatory, regulating lipid metabolism and anti-oxidative stress. This article will review the pharmacological mechanism of polydatin in anti-atherosclerosis, the biological characteristics of Polygonum cuspidatum, the toxicology and pharmacokinetics of polydatin and will provide ideas for further research.
Collapse
Affiliation(s)
- Min Wu
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoya Li
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Beijing University of Chinese Medicine, Beijing, China
| | - Songzi Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shengjie Yang
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ran Zhao
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Beijing University of Chinese Medicine, Beijing, China
| | - Yanwei Xing
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Longtao Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
162
|
Transcriptomic Analysis of Glioma Based on IDH Status Identifies ACAA2 as a Prognostic Factor in Lower Grade Glioma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1086792. [PMID: 32280672 PMCID: PMC7115055 DOI: 10.1155/2020/1086792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/05/2020] [Indexed: 01/01/2023]
Abstract
Background Glioma is the most common and lethal tumor in the central nervous system (CNS). More than 70% of WHO grade II/III gliomas were found to harbor isocitrate dehydrogenase (IDH) mutations which generated targetable metabolic vulnerabilities. Focusing on the metabolic vulnerabilities, some targeted therapies, such as NAMPT, have shown significant effects in preclinical and clinical trials. Methods We explored the TCGA as well as CGGA database and analyzed the RNA-seq data of lower grade gliomas (LGG) with the method of weighted correlation network analysis (WGCNA). Differential expressed genes were screened, and coexpression relationships were grouped together by performing average linkage hierarchical clustering on the topological overlap. Clinical data were used to conduct Kaplan–Meier analysis. Results In this study, we identified ACAA2 as a prognostic factor in IDH mutation lower grade glioma with the method of weighted correlation network analysis (WGCNA). The difference of ACAA2 gene expressions between the IDH wild-type (IDH-WT) group and the IDH mutant (IDH-MUT) group suggested that there may be different potential targeted therapies based on the fatty acid metabolic vulnerabilities, which promoted the personalized treatment for LGG patients.
Collapse
|
163
|
Saxena N, Chandra NC. Cholesterol: A Prelate in Cell Nucleus and its Serendipity. Curr Mol Med 2020; 20:692-707. [PMID: 32282300 DOI: 10.2174/1566524020666200413112030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 11/22/2022]
Abstract
Cholesterol is a chameleon bio-molecule in cellular multiplex. It acts as a prelate in almost every cellular compartment with its site specific characteristics viz. regulation of structural veracity and scaffold fluidity of bio-membranes, insulation of electrical transmission in nerves, controlling of genes by making steroid endocrines, acting as precursors of metabolic regulators and many more with its emerging prophecy in the cell nucleus to drive new cell formation. Besides the crucial legacy in cellular functionality, cholesterol is ostracized as a member of LDL particle, which has been proved responsible to clog blood vessels. LDL particles get deposited in the blood vessels because of their poor clearance owing to the non-functioning LDL receptor on the vessel wall and surrounding tissues. Blocking of blood vessel promotes heart attack and stroke. On the other hand, cholesterol has been targeted as pro-cancerous molecule. At this phase again cholesterol is biphasic. Although cholesterol is essential to construct nuclear membrane and its lipid-rafts; in cancer tumour cells, cholesterol is not under the control of intracellular feedback regulation and gets accumulated within cell nucleus by crossing nuclear membrane and promoting cell proliferation. In precancerous stage, the immune cells also die because of the lack of requisite concentration of intracellular and intranuclear cholesterol pool. The existence of cholesterol within the cell nucleus has been found in the nuclear membrane, epichromosomal location and nucleoplasm. The existence of cholesterol in the microdomain of nuclear raft has been reported to be linked with gene transcription, cell proliferation and apoptosis. Hydrolysis of cholesterol esters in chromosomal domain is linked with new cell generation. Apparently, Cholesterol is now a prelate in cell nucleus too ------ A serendipity in cellular haven.
Collapse
Affiliation(s)
- Nimisha Saxena
- Department of Biochemistry, KDMCH & Research Center, Akbarpur, Mathura - 281406, India
| | - Nimai Chand Chandra
- Department of Biochemistry, All India Institute of Medical Sciences, Phulwarisharif, Patna - 801507, India
| |
Collapse
|
164
|
Chen H, Chen Z, Zhang Z, Li Y, Zhang S, Jiang F, Wei J, Ding P, Zhou H, Gu Q, Xu J. Discovery of new LXRβ agonists as glioblastoma inhibitors. Eur J Med Chem 2020; 194:112240. [PMID: 32248003 DOI: 10.1016/j.ejmech.2020.112240] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/24/2022]
Abstract
Discovery and optimization of selective liver X receptor β (LXRβ) agonists are challenging due to the high homology of LXRα and LXRβ in the ligand-binding domain. There is only one different residue (Val versus Ile) at the ligand-binding pocket of LXRs. With machine learning methods, we identified pan LXR agonists with a novel scaffold (spiro[pyrrolidine-3,3'-oxindole]). Then, we figured out the mechanism of LXR isoform selectivity from co-crystal structures. Based on the mechanism and the new scaffold, LXRβ selective agonists were designed and synthesized. This led to the discovery of LXRβ agonists 4-7rr, 4-13 and 4-13rr with IC50 values ranging from 1.78 to 6.36 μM against glioblastoma in vitro. Treatment with 50 mg/kg/day of 4-13 for 15 days significantly reduced tumor growth using an in vivo xenograft glioblastoma model.
Collapse
Affiliation(s)
- Hao Chen
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Research Center for Drug Discovery at School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Ziyang Chen
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Research Center for Drug Discovery at School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Zizhen Zhang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Research Center for Drug Discovery at School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Yali Li
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Research Center for Drug Discovery at School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Shushu Zhang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Research Center for Drug Discovery at School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Fuqiang Jiang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Research Center for Drug Discovery at School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Junkang Wei
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Research Center for Drug Discovery at School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Peng Ding
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Research Center for Drug Discovery at School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Huihao Zhou
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Research Center for Drug Discovery at School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.
| | - Qiong Gu
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Research Center for Drug Discovery at School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.
| | - Jun Xu
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Research Center for Drug Discovery at School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.
| |
Collapse
|
165
|
Li D, Li S, Xue AZ, Smith Callahan LA, Liu Y. Expression of SREBP2 and cholesterol metabolism related genes in TCGA glioma cohorts. Medicine (Baltimore) 2020; 99:e18815. [PMID: 32195924 PMCID: PMC7220679 DOI: 10.1097/md.0000000000018815] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Diffuse gliomas are the most common primary brain tumors. The Cancer Genome Atlas (TCGA) database provides correlative evidence between altered molecular pathways and gliomas. Dysregulated cholesterol homeostasis emerges as a potential indicator of the pathogenesis of gliomas.Mining large cohorts from the TCGA together with database from the Chinese Glioma Genome Atlas (CGGA) for confirmation, we compared gene expression of cholesterol synthesis master regulator SREBP2 and its regulatory networks in low grade glioma (LGG) and glioblastoma (GBM).Our analysis shows that expression of SREBP2 and related genes is lower in GBM than in LGG, indicating that cholesterol metabolism processes, including de novo synthesis, cholesterol uptakes, and cholesterol conversion and efflux, are suppressed in GBM.Overall, our data suggests that SREBP2 transcript could serve as a potential prognosis marker or therapeutic target in diffuse glioma including GBM.
Collapse
Affiliation(s)
- Dali Li
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School
- Center for Stem Cell and Regenerative Medicine, the Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Shenglan Li
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School
- Center for Stem Cell and Regenerative Medicine, the Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Allen Z. Xue
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School
- Center for Stem Cell and Regenerative Medicine, the Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Laura A. Smith Callahan
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School
- Center for Stem Cell and Regenerative Medicine, the Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Ying Liu
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School
- Center for Stem Cell and Regenerative Medicine, the Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
166
|
Garcia-Bermudez J, Williams RT, Guarecuco R, Birsoy K. Targeting extracellular nutrient dependencies of cancer cells. Mol Metab 2020; 33:67-82. [PMID: 31926876 PMCID: PMC7056928 DOI: 10.1016/j.molmet.2019.11.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Cancer cells rewire their metabolism to meet the energetic and biosynthetic demands of their high proliferation rates and environment. Metabolic reprogramming of cancer cells may result in strong dependencies on nutrients that could be exploited for therapy. While these dependencies may be in part due to the nutrient environment of tumors, mutations or expression changes in metabolic genes also reprogram metabolic pathways and create addictions to extracellular nutrients. SCOPE OF REVIEW This review summarizes the major nutrient dependencies of cancer cells focusing on their discovery and potential mechanisms by which metabolites become limiting for tumor growth. We further detail available therapeutic interventions based on these metabolic features and highlight opportunities for restricting nutrient availability as an anti-cancer strategy. MAJOR CONCLUSIONS Strategies to limit nutrients required for tumor growth using dietary interventions or nutrient degrading enzymes have previously been suggested for cancer therapy. The best clinical example of exploiting cancer nutrient dependencies is the treatment of leukemia with l-asparaginase, a first-line chemotherapeutic that depletes serum asparagine. Despite the success of nutrient starvation in blood cancers, it remains unclear whether this approach could be extended to other solid tumors. Systematic studies to identify nutrient dependencies unique to individual tumor types have the potential to discover targets for therapy.
Collapse
Affiliation(s)
- Javier Garcia-Bermudez
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| | - Robert T Williams
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Rohiverth Guarecuco
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Kıvanç Birsoy
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
167
|
Zhou Y, Wang L, Wang C, Wu Y, Chen D, Lee TH. Potential implications of hydrogen peroxide in the pathogenesis and therapeutic strategies of gliomas. Arch Pharm Res 2020; 43:187-203. [PMID: 31956964 DOI: 10.1007/s12272-020-01205-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 01/05/2020] [Indexed: 12/15/2022]
Abstract
Glioma is the most common type of primary brain tumor, and it has a high mortality rate. Currently, there are only a few therapeutic approaches for gliomas, and their effects are unsatisfactory. Therefore, uncovering the pathogenesis and exploring more therapeutic strategies for the treatment of gliomas are urgently needed to overcome the ongoing challenges. Cellular redox imbalance has been shown to be associated with the initiation and progression of gliomas. Among reactive oxygen species (ROS), hydrogen peroxide (H2O2) is considered the most suitable for redox signaling and is a potential candidate as a key molecule that determines the fate of cancer cells. In this review, we discuss the potential cellular and molecular roles of H2O2 in gliomagenesis and explore the potential implications of H2O2 in radiotherapy and chemotherapy and in the ongoing challenges of current glioma treatment. Moreover, we evaluate H2O2 as a potential redox sensor and potential driver molecule of nanocatalytic therapeutic strategies for glioma treatment.
Collapse
Affiliation(s)
- Ying Zhou
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, 1 Xuefu North Road, Fuzhou, 350122, Fujian, China.,Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Provincial Universities and Colleges, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Long Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, 1 Xuefu North Road, Fuzhou, 350122, Fujian, China
| | - Chaojia Wang
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, 150040, Heilongjiang, China
| | - Yilin Wu
- The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Dongmei Chen
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, 1 Xuefu North Road, Fuzhou, 350122, Fujian, China
| | - Tae Ho Lee
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, 1 Xuefu North Road, Fuzhou, 350122, Fujian, China.
| |
Collapse
|
168
|
Han M, Wang S, Yang N, Wang X, Zhao W, Saed HS, Daubon T, Huang B, Chen A, Li G, Miletic H, Thorsen F, Bjerkvig R, Li X, Wang J. Therapeutic implications of altered cholesterol homeostasis mediated by loss of CYP46A1 in human glioblastoma. EMBO Mol Med 2020; 12:e10924. [PMID: 31777202 PMCID: PMC6949512 DOI: 10.15252/emmm.201910924] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/19/2019] [Accepted: 10/23/2019] [Indexed: 01/04/2023] Open
Abstract
Dysregulated cholesterol metabolism is a hallmark of many cancers, including glioblastoma (GBM), but its role in disease progression is not well understood. Here, we identified cholesterol 24-hydroxylase (CYP46A1), a brain-specific enzyme responsible for the elimination of cholesterol through the conversion of cholesterol into 24(S)-hydroxycholesterol (24OHC), as one of the most dramatically dysregulated cholesterol metabolism genes in GBM. CYP46A1 was significantly decreased in GBM samples compared with normal brain tissue. A reduction in CYP46A1 expression was associated with increasing tumour grade and poor prognosis in human gliomas. Ectopic expression of CYP46A1 suppressed cell proliferation and in vivo tumour growth by increasing 24OHC levels. RNA-seq revealed that treatment of GBM cells with 24OHC suppressed tumour growth through regulation of LXR and SREBP signalling. Efavirenz, an activator of CYP46A1 that is known to penetrate the blood-brain barrier, inhibited GBM growth in vivo. Our findings demonstrate that CYP46A1 is a critical regulator of cellular cholesterol in GBM and that the CYP46A1/24OHC axis is a potential therapeutic target.
Collapse
Affiliation(s)
- Mingzhi Han
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Department of BiomedicineUniversity of BergenBergenNorway
| | - Shuai Wang
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
| | - Ning Yang
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
| | - Xu Wang
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
| | - Wenbo Zhao
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
| | | | - Thomas Daubon
- INSERM U1029Institut Nationale de la Santé et de la Recherche MédicalePessacFrance
- University BordeauxPessacFrance
| | - Bin Huang
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
| | - Anjing Chen
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- School of MedicineShandong UniversityJinanChina
| | - Gang Li
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
| | - Hrvoje Miletic
- Department of BiomedicineUniversity of BergenBergenNorway
- Department of PathologyHaukeland University HospitalBergenNorway
| | - Frits Thorsen
- Department of BiomedicineUniversity of BergenBergenNorway
- Department of BiomedicineThe Molecular Imaging CenterUniversity of BergenBergenNorway
| | - Rolf Bjerkvig
- Department of BiomedicineUniversity of BergenBergenNorway
- NorLux Neuro‐Oncology LaboratoryDepartment of OncologyLuxembourg Institute of HealthLuxembourg CityLuxembourg
| | - Xingang Li
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
| | - Jian Wang
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Department of BiomedicineUniversity of BergenBergenNorway
| |
Collapse
|
169
|
Snaebjornsson MT, Janaki-Raman S, Schulze A. Greasing the Wheels of the Cancer Machine: The Role of Lipid Metabolism in Cancer. Cell Metab 2020; 31:62-76. [PMID: 31813823 DOI: 10.1016/j.cmet.2019.11.010] [Citation(s) in RCA: 521] [Impact Index Per Article: 130.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 10/27/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022]
Abstract
Altered lipid metabolism is among the most prominent metabolic alterations in cancer. Enhanced synthesis or uptake of lipids contributes to rapid cancer cell growth and tumor formation. Lipids are a highly complex group of biomolecules that not only constitute the structural basis of biological membranes but also function as signaling molecules and an energy source. Here, we summarize recent evidence implicating altered lipid metabolism in different aspects of the cancer phenotype and discuss potential strategies by which targeting lipid metabolism could provide a therapeutic window for cancer treatment.
Collapse
Affiliation(s)
- Marteinn Thor Snaebjornsson
- Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074 Würzburg, Germany; Division of Tumor Metabolism and Microenvironment, German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany.
| | - Sudha Janaki-Raman
- Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074 Würzburg, Germany.
| | - Almut Schulze
- Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074 Würzburg, Germany; Division of Tumor Metabolism and Microenvironment, German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany.
| |
Collapse
|
170
|
Bi J, Chowdhry S, Wu S, Zhang W, Masui K, Mischel PS. Altered cellular metabolism in gliomas - an emerging landscape of actionable co-dependency targets. Nat Rev Cancer 2020; 20:57-70. [PMID: 31806884 DOI: 10.1038/s41568-019-0226-5] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/31/2019] [Indexed: 12/18/2022]
Abstract
Altered cellular metabolism is a hallmark of gliomas. Propelled by a set of recent technological advances, new insights into the molecular mechanisms underlying glioma metabolism are rapidly emerging. In this Review, we focus on the dynamic nature of glioma metabolism and how it is shaped by the interaction between tumour genotype and brain microenvironment. Recent advances integrating metabolomics with genomics are discussed, yielding new insight into the mechanisms that drive glioma pathogenesis. Studies that shed light on interactions between the tumour microenvironment and tumour genotype are highlighted, providing important clues as to how gliomas respond to and adapt to their changing tissue and biochemical contexts. Finally, a road map for the discovery of potential new glioma drug targets is suggested, with the goal of translating these new insights about glioma metabolism into clinical benefits for patients.
Collapse
Affiliation(s)
- Junfeng Bi
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Sudhir Chowdhry
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Sihan Wu
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Wenjing Zhang
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Kenta Masui
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Paul S Mischel
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA.
- Department of Pathology, UCSD School of Medicine, La Jolla, CA, USA.
- Moores Cancer Center, UCSD School of Medicine, La Jolla, CA, USA.
| |
Collapse
|
171
|
Xiang DB, Zhang KQ, Zeng YL, Yan QZ, Shi Z, Tuo QH, Lin LM, Xia BH, Wu P, Liao DF. Curcumin: From a controversial "panacea" to effective antineoplastic products. Medicine (Baltimore) 2020; 99:e18467. [PMID: 31914018 PMCID: PMC6959860 DOI: 10.1097/md.0000000000018467] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Curcumin, a controversial "panacea," has been broadly studied. Its bioactivities including antioxidant, anti-inflammatory, and especially antineoplastic activities have been documented. However, due to its extensive bioactivities, some scientists hold a skeptical point of view toward curcumin and described curcumin as a "deceiver" to chemists. The objective of this study was to explore curcumin's another possibility as a potential supplementary leading compound to cancer treatments. METHODS Literature searches were conducted using electronic databases. Search terms such as "curcumin," "curcumin analogues," and so on were used. The literatures were collected and summarized. In this article, reported targets of curcumin are reviewed. The limitations of a curcumin as a therapeutic anticancer product including low bioavailability and poor targeting are mentioned. Furthermore, modified curcumin analogues and antitumor mechanisms are listed and discussed in the aspects of cell death and tumor microenvironment including angiogenesis, tissue hypoxia status, and energy metabolism. RESULTS Several possible modification strategies were presented by analyzing the relationships between the antitumor activity of curcumin analogues and their structural characteristics, including the introduction of hydrophilic group, shortening of redundant hydrocarbon chain, the introduction of extra chemical group, and so on. CONCLUSIONS From our perspective, after structural modification curcumin could be more effective complementary product for cancer therapies by the enhancement of targeting abilities and the improvement of bioavailability.
Collapse
Affiliation(s)
- De-Biao Xiang
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| | - Kai-Qiang Zhang
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| | - Ya-Ling Zeng
- Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Qing-Zi Yan
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| | - Zhe Shi
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| | - Qin-Hui Tuo
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| | - Li-Mei Lin
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| | - Bo-Hou Xia
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| | - Ping Wu
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| | - Duan-Fang Liao
- Division of Stem Cell Regulation and Application, Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province
| |
Collapse
|
172
|
Lipoprotein Drug Delivery Vehicles for Cancer: Rationale and Reason. Int J Mol Sci 2019; 20:ijms20246327. [PMID: 31847457 PMCID: PMC6940806 DOI: 10.3390/ijms20246327] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/26/2019] [Accepted: 12/04/2019] [Indexed: 12/11/2022] Open
Abstract
Lipoproteins are a family of naturally occurring macromolecular complexes consisting amphiphilic apoproteins, phospholipids, and neutral lipids. The physiological role of mammalian plasma lipoproteins is to transport their apolar cargo (primarily cholesterol and triglyceride) to their respective destinations through a highly organized ligand-receptor recognition system. Current day synthetic nanoparticle delivery systems attempt to accomplish this task; however, many only manage to achieve limited results. In recent years, many research labs have employed the use of lipoprotein or lipoprotein-like carriers to transport imaging agents or drugs to tumors. The purpose of this review is to highlight the pharmacologic, clinical, and molecular evidence for utilizing lipoprotein-based formulations and discuss their scientific rationale. To accomplish this task, evidence of dynamic drug interactions with circulating plasma lipoproteins are presented. This is followed by epidemiologic and molecular data describing the association between cholesterol and cancer.
Collapse
|
173
|
Wu A, Grela E, Wójtowicz K, Filipczak N, Hamon Y, Luchowski R, Grudziński W, Raducka-Jaszul O, Gagoś M, Szczepaniak A, Chimini G, Gruszecki WI, Trombik T. ABCA1 transporter reduces amphotericin B cytotoxicity in mammalian cells. Cell Mol Life Sci 2019; 76:4979-4994. [PMID: 31134303 PMCID: PMC6881254 DOI: 10.1007/s00018-019-03154-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 05/07/2019] [Accepted: 05/16/2019] [Indexed: 01/20/2023]
Abstract
Amphotericin B (AmB) belongs to a group of polyene antibiotics commonly used in the treatment of systemic mycotic infections. A widely accepted mechanism of action of AmB is based on the formation of an oligomeric pore structure within the plasma membrane (PM) by interaction with membrane sterols. Although AmB binds preferentially to ergosterol, it can also bind to cholesterol in the mammalian PM and cause severe cellular toxicity. The lipid content and its lateral organization at the cell PM appear to be significant for AmB binding. Several ATP-binding cassette (ABC) transporters, including ABCA1, play a crucial role in lipid translocation, cholesterol redistribution and efflux. Here, we demonstrate that cells expressing ABCA1 are more resistant to AmB treatment, while cells lacking ABCA1 expression or expressing non-active ABCA1MM mutant display increased sensitivity. Further, a FLIM analysis of AmB-treated cells reveals a fraction of the antibiotic molecules, characterized by relatively high fluorescence lifetimes (> 6 ns), involved in formation of bulk cholesterol-AmB structures at the surface of ABCA1-expressing cells. Finally, lowering the cellular cholesterol content abolishes resistance of ABCA1-expressing cells to AmB. Therefore, we propose that ABCA1-mediated cholesterol efflux from cells induces formation of bulk cholesterol-AmB structures at the cell surface, preventing AmB cytotoxicity.
Collapse
Affiliation(s)
- A Wu
- Faculty of Biotechnology, University of Wroclaw, 50-383, Wrocław, Poland
| | - E Grela
- Department of Biophysics, Institute of Physics, Maria Curie-Sklodowska University, 20-031, Lublin, Poland
| | - K Wójtowicz
- Faculty of Biotechnology, University of Wroclaw, 50-383, Wrocław, Poland
| | - N Filipczak
- Faculty of Biotechnology, University of Wroclaw, 50-383, Wrocław, Poland
| | - Y Hamon
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
| | - R Luchowski
- Department of Biophysics, Institute of Physics, Maria Curie-Sklodowska University, 20-031, Lublin, Poland
| | - W Grudziński
- Department of Biophysics, Institute of Physics, Maria Curie-Sklodowska University, 20-031, Lublin, Poland
| | - O Raducka-Jaszul
- Faculty of Biotechnology, University of Wroclaw, 50-383, Wrocław, Poland
| | - M Gagoś
- Department of Cell Biology, Maria Curie-Skłodowska University, 20-033, Lublin, Poland
| | - A Szczepaniak
- Faculty of Biotechnology, University of Wroclaw, 50-383, Wrocław, Poland
| | - G Chimini
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
| | - W I Gruszecki
- Department of Biophysics, Institute of Physics, Maria Curie-Sklodowska University, 20-031, Lublin, Poland
| | - T Trombik
- Faculty of Biotechnology, University of Wroclaw, 50-383, Wrocław, Poland.
| |
Collapse
|
174
|
Shao W, Zhu W, Lin J, Luo M, Lin Z, Lu L, Jia H, Qin L, Lu M, Chen J. Liver X Receptor Agonism Sensitizes a Subset of Hepatocellular Carcinoma to Sorafenib by Dual-Inhibiting MET and EGFR. Neoplasia 2019; 22:1-9. [PMID: 31751859 PMCID: PMC6911865 DOI: 10.1016/j.neo.2019.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/08/2019] [Accepted: 08/12/2019] [Indexed: 01/01/2023] Open
Abstract
Sorafenib is the first approved systemic therapy for advanced hepatocellular carcinoma (HCC) and is the first-line choice in clinic. Sustained activation of receptor tyrosine kinases (RTKs) is associated with low efficacy of sorafenib in HCC. Activation of liver X receptor (LXR) has been reported to inhibit some RTKs. In this study, we found that the LXR agonist enhanced the anti-tumor activity of sorafenib in a subset of HCC cells with high LXR-β/α gene expression ratio. Mechanically, the activation of LXR suppressed sorafenib dependent recruitment of MET and epidermal growth factor receptor (EGFR) in lipid rafts through cholesterol efflux. Our findings imply that LXR agonist can serve as a potential sensitizer to enhance the anti-tumor effect of sorafenib.
Collapse
Affiliation(s)
- Weiqing Shao
- Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road (M), Shanghai 200040, China
| | - Wenwei Zhu
- Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road (M), Shanghai 200040, China
| | - Jing Lin
- Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road (M), Shanghai 200040, China
| | - Mengjun Luo
- Key Laboratory of Medical Molecular Virology (MOE & MOH), Institutes of Biomedical Sciences, Fudan University, 138 Yi Xue Yuan Road, Shanghai 200032, China
| | - Zhifei Lin
- Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road (M), Shanghai 200040, China
| | - Lu Lu
- Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road (M), Shanghai 200040, China
| | - Huliang Jia
- Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road (M), Shanghai 200040, China
| | - Lunxiu Qin
- Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road (M), Shanghai 200040, China; Institutes of Biomedical Sciences, Fudan University, 138 Yi Xue Yuan Road, Shanghai 200032, China.
| | - Ming Lu
- Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road (M), Shanghai 200040, China.
| | - Jinhong Chen
- Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road (M), Shanghai 200040, China.
| |
Collapse
|
175
|
Riscal R, Skuli N, Simon MC. Even Cancer Cells Watch Their Cholesterol! Mol Cell 2019; 76:220-231. [PMID: 31586545 DOI: 10.1016/j.molcel.2019.09.008] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/30/2019] [Accepted: 09/05/2019] [Indexed: 02/07/2023]
Abstract
Deregulated cell proliferation is an established feature of cancer, and altered tumor metabolism has witnessed renewed interest over the past decade, including the study of how cancer cells rewire metabolic pathways to renew energy sources and "building blocks" that sustain cell division. Microenvironmental oxygen, glucose, and glutamine are regarded as principal nutrients fueling tumor growth. However, hostile tumor microenvironments render O2/nutrient supplies chronically insufficient for increased proliferation rates, forcing cancer cells to develop strategies for opportunistic modes of nutrient acquisition. Recent work shows that cancer cells overcome this nutrient scarcity by scavenging other substrates, such as proteins and lipids, or utilizing adaptive metabolic pathways. As such, reprogramming lipid metabolism plays important roles in providing energy, macromolecules for membrane synthesis, and lipid-mediated signaling during cancer progression. In this review, we highlight more recently appreciated roles for lipids, particularly cholesterol and its derivatives, in cancer cell metabolism within intrinsically harsh tumor microenvironments.
Collapse
Affiliation(s)
- Romain Riscal
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicolas Skuli
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
176
|
Bi J, Ichu TA, Zanca C, Yang H, Zhang W, Gu Y, Chowdhry S, Reed A, Ikegami S, Turner KM, Zhang W, Villa GR, Wu S, Quehenberger O, Yong WH, Kornblum HI, Rich JN, Cloughesy TF, Cavenee WK, Furnari FB, Cravatt BF, Mischel PS. Oncogene Amplification in Growth Factor Signaling Pathways Renders Cancers Dependent on Membrane Lipid Remodeling. Cell Metab 2019; 30:525-538.e8. [PMID: 31303424 PMCID: PMC6742496 DOI: 10.1016/j.cmet.2019.06.014] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 04/28/2019] [Accepted: 06/12/2019] [Indexed: 01/11/2023]
Abstract
Advances in DNA sequencing technologies have reshaped our understanding of the molecular basis of cancer, providing a precise genomic view of tumors. Complementary biochemical and biophysical perspectives of cancer point toward profound shifts in nutrient uptake and utilization that propel tumor growth and major changes in the structure of the plasma membrane of tumor cells. The molecular mechanisms that bridge these fundamental aspects of tumor biology remain poorly understood. Here, we show that the lysophosphatidylcholine acyltransferase LPCAT1 functionally links specific genetic alterations in cancer with aberrant metabolism and plasma membrane remodeling to drive tumor growth. Growth factor receptor-driven cancers are found to depend on LPCAT1 to shape plasma membrane composition through enhanced saturated phosphatidylcholine content that is, in turn, required for the transduction of oncogenic signals. These results point to a genotype-informed strategy that prioritizes lipid remodeling pathways as therapeutic targets for diverse cancers.
Collapse
Affiliation(s)
- Junfeng Bi
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Taka-Aki Ichu
- Department of Chemistry, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ciro Zanca
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Huijun Yang
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wei Zhang
- Department of Medicine, UCSD School of Medicine, La Jolla, CA 92093, USA
| | - Yuchao Gu
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA; Department of Molecular and Medical Pharmacology David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA
| | - Sudhir Chowdhry
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Alex Reed
- Department of Chemistry, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Shiro Ikegami
- Department of Neurological Surgery, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Kristen M Turner
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wenjing Zhang
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Genaro R Villa
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA; Department of Molecular and Medical Pharmacology David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA
| | - Sihan Wu
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA
| | - Oswald Quehenberger
- Department of Medicine, UCSD School of Medicine, La Jolla, CA 92093, USA; Department of Pharmacology, UCSD School of Medicine, La Jolla, CA 92093, USA
| | - William H Yong
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Harley I Kornblum
- Department of Molecular and Medical Pharmacology David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen UCLA School of Medicine, Los Angeles, CA, USA
| | - Jeremy N Rich
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Timothy F Cloughesy
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Webster K Cavenee
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA; Department of Medicine, UCSD School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, UCSD School of Medicine, La Jolla, CA 92093, USA
| | - Frank B Furnari
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pathology, UCSD School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, UCSD School of Medicine, La Jolla, CA 92093, USA
| | - Benjamin F Cravatt
- Department of Chemistry, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Paul S Mischel
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pathology, UCSD School of Medicine, La Jolla, CA 92093, USA; Moores Cancer Center, UCSD School of Medicine, La Jolla, CA 92093, USA.
| |
Collapse
|
177
|
Zhou W, Wahl DR. Metabolic Abnormalities in Glioblastoma and Metabolic Strategies to Overcome Treatment Resistance. Cancers (Basel) 2019; 11:cancers11091231. [PMID: 31450721 PMCID: PMC6770393 DOI: 10.3390/cancers11091231] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/07/2019] [Accepted: 08/16/2019] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumor and is nearly universally fatal. Targeted therapy and immunotherapy have had limited success in GBM, leaving surgery, alkylating chemotherapy and ionizing radiation as the standards of care. Like most cancers, GBMs rewire metabolism to fuel survival, proliferation, and invasion. Emerging evidence suggests that this metabolic reprogramming also mediates resistance to the standard-of-care therapies used to treat GBM. In this review, we discuss the noteworthy metabolic features of GBM, the key pathways that reshape tumor metabolism, and how inhibiting abnormal metabolism may be able to overcome the inherent resistance of GBM to radiation and chemotherapy.
Collapse
Affiliation(s)
- Weihua Zhou
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel R Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
178
|
Dang Q, Chen YA, Hsieh JT. The dysfunctional lipids in prostate cancer. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2019; 7:273-280. [PMID: 31511833 PMCID: PMC6734041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 06/10/2023]
Abstract
Prostate cancer (PCa) is well-recognized as a lipid-enriched tumor. Lipids represent a diverse array of molecules essential to the cellular structure, defense, energy, and communication. Lipid metabolism can often become dysregulated during tumor development. The increasing body of knowledge on the biological actions of steroid hormone-androgens in PCa has led to the development of several targeted therapies that still represent the standard of care for cancer patients to this day. Sequencing technologies for functional analyses of androgen receptors (ARs) have revealed that AR is also a master regulator of cellular energy metabolism such as fatty acid ß-oxidation, and de novo lipid synthesis. In addition, bioactive lipids are also used as physiological signaling molecules, which have been shown to be involved in PCa progression. This review discusses the potent player(s) in altered lipid metabolism of PCa and describes how lipids and their interactions with proteins can be used for therapeutic advantage. We also discuss the possibility that the altered bioactive lipid mediators affect intracellular signaling pathway and the related transcriptional regulation be of therapeutic interest.
Collapse
Affiliation(s)
- Qiang Dang
- Department of Urology, University of Texas Southwestern Medical CenterDallas, TX 75390, USA
- Department of Urology, Nanfang Hospital, Southern Medical UniversityGuangzhou 510515, China
| | - Yu-An Chen
- Department of Urology, University of Texas Southwestern Medical CenterDallas, TX 75390, USA
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical CenterDallas, TX 75390, USA
| |
Collapse
|
179
|
McKinney A, Lindberg OR, Engler JR, Chen KY, Kumar A, Gong H, Lu KV, Simonds EF, Cloughesy TF, Liau LM, Prados M, Bollen AW, Berger MS, Shieh JTC, James CD, Nicolaides TP, Yong WH, Lai A, Hegi ME, Weiss WA, Phillips JJ. Mechanisms of Resistance to EGFR Inhibition Reveal Metabolic Vulnerabilities in Human GBM. Mol Cancer Ther 2019; 18:1565-1576. [PMID: 31270152 DOI: 10.1158/1535-7163.mct-18-1330] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 05/10/2019] [Accepted: 06/28/2019] [Indexed: 12/23/2022]
Abstract
Amplification of the epidermal growth factor receptor gene (EGFR) represents one of the most commonly observed genetic lesions in glioblastoma (GBM); however, therapies targeting this signaling pathway have failed clinically. Here, using human tumors, primary patient-derived xenografts (PDX), and a murine model for GBM, we demonstrate that EGFR inhibition leads to increased invasion of tumor cells. Further, EGFR inhibitor-treated GBM demonstrates altered oxidative stress, with increased lipid peroxidation, and generation of toxic lipid peroxidation products. A tumor cell subpopulation with elevated aldehyde dehydrogenase (ALDH) levels was determined to comprise a significant proportion of the invasive cells observed in EGFR inhibitor-treated GBM. Our analysis of the ALDH1A1 protein in newly diagnosed GBM revealed detectable ALDH1A1 expression in 69% (35/51) of the cases, but in relatively low percentages of tumor cells. Analysis of paired human GBM before and after EGFR inhibitor therapy showed an increase in ALDH1A1 expression in EGFR-amplified tumors (P < 0.05, n = 13 tumor pairs), and in murine GBM ALDH1A1-high clones were more resistant to EGFR inhibition than ALDH1A1-low clones. Our data identify ALDH levels as a biomarker of GBM cells with high invasive potential, altered oxidative stress, and resistance to EGFR inhibition, and reveal a therapeutic target whose inhibition should limit GBM invasion.
Collapse
Affiliation(s)
- Andrew McKinney
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Olle R Lindberg
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Jane R Engler
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Katharine Y Chen
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Anupam Kumar
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Henry Gong
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Kan V Lu
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Erin F Simonds
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California.,Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Timothy F Cloughesy
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.,Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Linda M Liau
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.,Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Michael Prados
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Andrew W Bollen
- Department of Pathology, Division of Neuropathology, University of California, San Francisco, San Francisco, California
| | - Mitchel S Berger
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California
| | - Joseph T C Shieh
- Division of Medical Genetics, Department of Pediatrics, UCSF Benioff Children's Hospital, University of California, San Francisco, San Francisco, California.,Institute for Human Genetics, University of California, San Francisco, San Francisco, California
| | - C David James
- Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Theodore P Nicolaides
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California.,Department of Pediatrics, UCSF Benioff Children's Hospital, University of California, San Francisco, San Francisco, California
| | - William H Yong
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Albert Lai
- UCLA Neuro-Oncology Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.,Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Monika E Hegi
- Neuroscience Research Center and Service of Neurosurgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - William A Weiss
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California.,Department of Neurology, University of California, San Francisco, San Francisco, California.,Department of Pediatrics, UCSF Benioff Children's Hospital, University of California, San Francisco, San Francisco, California
| | - Joanna J Phillips
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, San Francisco, California. .,Department of Pathology, Division of Neuropathology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
180
|
Gimple RC, Kidwell RL, Kim LJY, Sun T, Gromovsky AD, Wu Q, Wolf M, Lv D, Bhargava S, Jiang L, Prager BC, Wang X, Ye Q, Zhu Z, Zhang G, Dong Z, Zhao L, Lee D, Bi J, Sloan AE, Mischel PS, Brown JM, Cang H, Huan T, Mack SC, Xie Q, Rich JN. Glioma Stem Cell-Specific Superenhancer Promotes Polyunsaturated Fatty-Acid Synthesis to Support EGFR Signaling. Cancer Discov 2019; 9:1248-1267. [PMID: 31201181 DOI: 10.1158/2159-8290.cd-19-0061] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 05/03/2019] [Accepted: 06/11/2019] [Indexed: 01/02/2023]
Abstract
Glioblastoma ranks among the most aggressive and lethal of all human cancers. Functionally defined glioma stem cells (GSC) contribute to this poor prognosis by driving therapeutic resistance and maintaining cellular heterogeneity. To understand the molecular processes essential for GSC maintenance and tumorigenicity, we interrogated the superenhancer landscapes of primary glioblastoma specimens and in vitro GSCs. GSCs epigenetically upregulated ELOVL2, a key polyunsaturated fatty-acid synthesis enzyme. Targeting ELOVL2 inhibited glioblastoma cell growth and tumor initiation. ELOVL2 depletion altered cellular membrane phospholipid composition, disrupted membrane structural properties, and diminished EGFR signaling through control of fatty-acid elongation. In support of the translational potential of these findings, dual targeting of polyunsaturated fatty-acid synthesis and EGFR signaling had a combinatorial cytotoxic effect on GSCs. SIGNIFICANCE: Glioblastoma remains a devastating disease despite extensive characterization. We profiled epigenomic landscapes of glioblastoma to pinpoint cell state-specific dependencies and therapeutic vulnerabilities. GSCs utilize polyunsaturated fatty-acid synthesis to support membrane architecture, inhibition of which impairs EGFR signaling and GSC proliferation. Combinatorial targeting of these networks represents a promising therapeutic strategy.See related commentary by Affronti and Wellen, p. 1161.This article is highlighted in the In This Issue feature, p. 1143.
Collapse
Affiliation(s)
- Ryan C Gimple
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California.,Department of Pathology, Case Western University, Cleveland, Ohio
| | - Reilly L Kidwell
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California
| | - Leo J Y Kim
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California.,Department of Pathology, Case Western University, Cleveland, Ohio
| | - Tengqian Sun
- Salk Institute for Biological Studies, La Jolla, California
| | - Anthony D Gromovsky
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Qiulian Wu
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California
| | - Megan Wolf
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Deguan Lv
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California
| | - Shruti Bhargava
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California
| | - Li Jiang
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California
| | - Briana C Prager
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California.,Department of Pathology, Case Western University, Cleveland, Ohio.,Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio
| | - Xiuxing Wang
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California
| | - Qing Ye
- Salk Institute for Biological Studies, La Jolla, California
| | - Zhe Zhu
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California
| | - Guoxin Zhang
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California
| | - Zhen Dong
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California
| | - Linjie Zhao
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California
| | - Derrick Lee
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California
| | - Junfeng Bi
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, California
| | - Andrew E Sloan
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland Ohio.,Department of Neurological Surgery, University Hospitals-Cleveland Medical Center, Cleveland, Ohio
| | - Paul S Mischel
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, California.,Department of Pathology, UCSD School of Medicine, La Jolla, California.,Moores Cancer Center, UCSD School of Medicine, La Jolla, California
| | - J Mark Brown
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Hu Cang
- Salk Institute for Biological Studies, La Jolla, California
| | - Tao Huan
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephen C Mack
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas.,Dan L. Duncan Cancer Center, Houston, Texas
| | - Qi Xie
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California. .,Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Westlake University, Hangzhou, China
| | - Jeremy N Rich
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, San Diego, California. .,Moores Cancer Center, UCSD School of Medicine, La Jolla, California.,Department of Neurosciences, UCSD School of Medicine, La Jolla, California
| |
Collapse
|
181
|
Nguyen TTT, Ishida CT, Shang E, Shu C, Bianchetti E, Karpel-Massler G, Siegelin MD. Activation of LXR Receptors and Inhibition of TRAP1 Causes Synthetic Lethality in Solid Tumors. Cancers (Basel) 2019; 11:cancers11060788. [PMID: 31181660 PMCID: PMC6627953 DOI: 10.3390/cancers11060788] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 02/06/2023] Open
Abstract
Cholesterol is a pivotal factor for cancer cells to entertain their relentless growth. In this case, we provide a novel strategy to inhibit tumor growth by simultaneous activation of liver-X-receptors and interference with Tumor Necrosis Factor Receptor-associated Protein 1 (TRAP1). Informed by a transcriptomic and subsequent gene set enrichment analysis, we demonstrate that inhibition of TRAP1 results in suppression of the cholesterol synthesis pathway in stem-like and established glioblastoma (GBM) cells by destabilizing the transcription factor SREBP2. Notably, TRAP1 inhibition induced cell death, which was rescued by cholesterol and mevalonate. Activation of liver X receptor (LXR) by a clinically validated LXR agonist, LXR623, along with the TRAP1 inhibitor, gamitrinib (GTPP), results in synergistic reduction of tumor growth and cell death induction in a broad range of solid tumors, which is rescued by exogenous cholesterol. The LXR agonist and TRAP1 inhibitor mediated cell death is regulated at the level of Bcl-2 family proteins with an elevation of pro-apoptotic Noxa. Silencing of Noxa and its effector BAK attenuates cell death mediated by the combination treatment of LXR agonists and TRAP1 inhibition. Combined inhibition of TRAP1 and LXR agonists elicits a synergistic activation of the integrated stress response with an increase in activating transcription factor 4 (ATF4) driven by protein kinase RNA-like endoplasmic reticulum kinase (PERK). Silencing of ATF4 attenuates the increase of Noxa by using the combination treatment. Lastly, we demonstrate in patient-derived xenografts that the combination treatment of LXR623 and gamitrinib reduces tumor growth more potent than each compound. Taken together, these results suggest that TRAP1 inhibition and simultaneous activation of LXR might be a potent novel treatment strategy for solid malignancies.
Collapse
Affiliation(s)
- Trang Thi Thu Nguyen
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY 10032, USA.
| | - Chiaki Tsuge Ishida
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY 10032, USA.
| | - Enyuan Shang
- Department of Biological Sciences, Bronx Community College, City University of New York, Bronx, NY 10453, USA.
| | - Chang Shu
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY 10032, USA.
| | - Elena Bianchetti
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY 10032, USA.
| | | | - Markus D Siegelin
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
182
|
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: 1.6] [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
|
183
|
van Loon NM, Lindholm D, Zelcer N. The E3 ubiquitin ligase inducible degrader of the LDL receptor/myosin light chain interacting protein in health and disease. Curr Opin Lipidol 2019; 30:192-197. [PMID: 30896554 DOI: 10.1097/mol.0000000000000593] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW The RING E3 ubiquitin ligase inducible degrader of the LDL receptor (IDOL, also known as MYLIP) promotes ubiquitylation and subsequent lysosomal degradation of the LDL receptor (LDLR), thus acting to limit uptake of lipoprotein-derived cholesterol into cells. Next to the LDLR, IDOL also promotes degradation of two related receptors, the very LDL receptor (VLDLR) and apolipoprotein E receptor 2 (APOER2), which have important signaling functions in the brain. We review here the emerging role of IDOL in lipoprotein and energy metabolism, neurodegenerative diseases, and the potential for therapeutic targeting of IDOL. RECENT FINDINGS Genetic studies suggest an association between IDOL and lipoprotein metabolism in humans. Studies in rodents and nonhuman primates support an in-vivo role for IDOL in lipoprotein metabolism, and also uncovered an unexpected role in whole-body energy metabolism. Recent evaluation of IDOL function in the brain revealed a role in memory formation and progression of Alzheimer's disease. The report of the first IDOL inhibitor may facilitate further investigations on therapeutic strategies to target IDOL. SUMMARY IDOL is emerging as an important determinant of lipid and energy metabolism in metabolic disease as well as in Alzheimer's disease. IDOL targeting may be beneficial in treating these conditions.
Collapse
Affiliation(s)
- Nienke M van Loon
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Dan Lindholm
- Medicum, Department of Biochemistry and Developmental Biology, Medical Faculty, University of Helsinki
- Minerva Foundation Institute for Medical Research, Biomedicum-2, Helsinki, Finland
| | - Noam Zelcer
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| |
Collapse
|
184
|
Bilotta MT, Abruzzese MP, Molfetta R, Scarno G, Fionda C, Zingoni A, Soriani A, Garofalo T, Petrucci MT, Ricciardi MR, Paolini R, Santoni A, Cippitelli M. Activation of liver X receptor up-regulates the expression of the NKG2D ligands MICA and MICB in multiple myeloma through different molecular mechanisms. FASEB J 2019; 33:9489-9504. [PMID: 31125275 DOI: 10.1096/fj.201900319r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
NK cells have an important role in immunosurveillance of multiple myeloma (MM) progression, and their activity is enhanced by combination therapies able to regulate the expression of specific activating ligands. Liver X receptors (LXRs) are nuclear receptors and important regulators of intracellular cholesterol and lipid homeostasis. Moreover, they have regulatory roles in both cancer and immune response. Indeed, they can regulate inflammation and innate and acquired immunity. Furthermore, LXR activation directly acts in cancer cells (e.g., prostate, breast, melanoma, colon cancer, hepatocarcinoma, glioblastoma, and MM) that show an accumulation of cholesterol and alteration of LXR-mediated metabolic pathways. Here, we investigated the role of LXR and cholesterol on the expression of the NK cell-activating ligands major histocompatibility complex class I chain-related molecule A and B (MICA and MICB) in MM cells. The results shown in this work indicate that MM cells are responsive to LXR activation, which induces changes in the intracellular cholesterol content. These changes correlate with an enhanced expression of MICA and MICB in human MM cell lines and in primary malignant plasma cells, 2 ligands of the NK group 2D receptor (NKG2D)/CD314 activating receptor expressed in cytotoxic lymphocytes, rendering MM cells more sensitive to recognition, degranulation, and killing by NK cells. Mechanistically, we observed that LXR activation regulates MICA and MICB expression at different levels: MICA at the transcriptional level, enhancing mica promoter activity, and MICB by inhibiting its degradation in lysosomes. The present study provides evidence that activation of LXR, by enhancing NKG2D ligand expression, can promote NK cell-mediated cytotoxicity and suggests a novel immune-mediated mechanism involving modulation of intracellular cholesterol levels in cancer cells.-Bilotta, M. T., Abruzzese, M. P., Molfetta, R., Scarno, G., Fionda, C., Zingoni, A., Soriani, A., Garofalo, T., Petrucci, M. T., Ricciardi, M. R., Paolini, R., Santoni, A., Cippitelli, M. Activation of liver X receptor up-regulates the expression of the NKG2D ligands MICA and MICB in multiple myeloma through different molecular mechanisms.
Collapse
Affiliation(s)
| | | | - Rosa Molfetta
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Gianluca Scarno
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Cinzia Fionda
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessandra Zingoni
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessandra Soriani
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Tina Garofalo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Teresa Petrucci
- Division of Hematology, Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Maria Rosaria Ricciardi
- Department of Clinical and Molecular Medicine, Hematology, Sapienza University of Rome, Rome, Italy
| | - Rossella Paolini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.,Istituto Pasteur-Fondazione Cenci Bolognetti, Rome, Italy.,Istituto Mediterraneo di Neuroscienze Neuromed, Pozzilli, Italy
| | - Marco Cippitelli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
185
|
Liang X, Cao Y, Xiang S, Xiang Z. LXRα-mediated downregulation of EGFR suppress colorectal cancer cell proliferation. J Cell Biochem 2019; 120:17391-17404. [PMID: 31104333 DOI: 10.1002/jcb.29003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 12/20/2022]
Abstract
Liver X receptors (LXRs) are members of the nuclear receptor family, including the LXRα (NR1H3) and LXRβ (NR1H2) subtypes, which are related to the metabolism of glucose and cholesterol and possess anti-inflammatory functions. Mounting evidence has linked LXRs to the inhibition of cell proliferation in a variety of cancers. We revealed a differential distribution for NR1H3, but not for NR1H2, in colorectal cancer and adjacent normal tissues. We found that NR1H3 enhanced the inhibitory action of GW3965, an agonist of LXRs, on the proliferation of colorectal cancer cells. Upregulation of NR1H3 enhanced the inhibition of cell proliferation by GW3965 while silencing of NR1H3 attenuated the inhibitory effect of GW3965 on cell proliferation. Bioinformatic prediction and luciferase assays showed that NR1H3 was able to inhibit the activity of the epidermal growth factor receptor (EGFR) promoter. Moreover, we demonstrated that activation of NR1H3 inhibited the growth of transplanted tumors in an animal experiment, with the inhibition accompanied by downregulation of EGFR. Our findings suggest that NR1H3 controls cell proliferation by affecting EGFR promoter activity. The high expression of EGFR was due to the downregulation of NR1H3 which is a novel molecular mechanism in the development of colorectal cancer.
Collapse
Affiliation(s)
- Xiaolong Liang
- Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Cao
- Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Song Xiang
- Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zheng Xiang
- Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Department of General Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
186
|
Nalbuphine suppresses breast cancer stem-like properties and epithelial-mesenchymal transition via the AKT-NFκB signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:197. [PMID: 31092275 PMCID: PMC6521451 DOI: 10.1186/s13046-019-1184-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/17/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cancer pain is a debilitating disorder of human breast cancer and a primary determinant of the poor quality of life, and relieving pain is fundamental strategy in the cancer treatment. However, opioid analgesics, like morphine and fentanyl, which are widely used in cancer pain treatment have been reported to enhance stem-like traits and epithelial-mesenchymal transition (EMT) of breast cancer cells. As such, it is vital to make the best choice of analgesic for breast cancer management. METHODS MTT assays and colony formation assays were performed to examine tumor cell proliferation upon nalbuphine treatment. RT-PCR, western blot, flow cytometry, sphere formation, immunohistochemistry, transwell assays, wound healing assays and mouse xenograft were used to assess the biological effects of nalbuphine treatment. RESULTS Nalbuphine inhibited breast cancer cell growth and tumorigenesis, with little effect on noncancerous breast cell lines. Nalbuphine suppressed cancer stem-like traits and EMT in both breast cancer cells and mouse xenograft tumor tissues. Additionally, activation of AKT reversed the nalbuphine-induced inhibition of cancer stem-like properties, tumorigenesis and EMT. CONCLUSIONS Our results demonstrate a new role of nalbuphine in inhibiting cancer stem-like properties and EMT in addition to relieving pain, which suggests that nalbuphine may be effective in breast cancer treatment.
Collapse
|
187
|
Target identification reveals lanosterol synthase as a vulnerability in glioma. Proc Natl Acad Sci U S A 2019; 116:7957-7962. [PMID: 30923116 DOI: 10.1073/pnas.1820989116] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) remains an incurable childhood brain tumor for which novel therapeutic approaches are desperately needed. Previous studies have shown that the menin inhibitor MI-2 exhibits promising activity in preclinical DIPG and adult glioma models, although the mechanism underlying this activity is unknown. Here, using an integrated approach, we show that MI-2 exerts its antitumor activity in glioma largely independent of its ability to target menin. Instead, we demonstrate that MI-2 activity in glioma is mediated by disruption of cholesterol homeostasis, with suppression of cholesterol synthesis and generation of the endogenous liver X receptor ligand, 24,25-epoxycholesterol, resulting in cholesterol depletion and cell death. Notably, this mechanism is responsible for MI-2 activity in both DIPG and adult glioma cells. Metabolomic and biochemical analyses identify lanosterol synthase as the direct molecular target of MI-2, revealing this metabolic enzyme as a vulnerability in glioma and further implicating cholesterol homeostasis as an attractive pathway to target in this malignancy.
Collapse
|
188
|
The Roles of Low-Density Lipoprotein Receptor-Related Proteins 5, 6, and 8 in Cancer: A Review. JOURNAL OF ONCOLOGY 2019; 2019:4536302. [PMID: 31031810 PMCID: PMC6457291 DOI: 10.1155/2019/4536302] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 02/05/2019] [Accepted: 02/26/2019] [Indexed: 01/14/2023]
Abstract
Low-density lipoprotein receptor (LDLR) has been an object of research since the 1970s because of its role in various cell functions. The LDLR family members include LRP5, LRP6, and LRP8. Even though LRP5, 6, and 8 are in the same family, intriguingly, these three proteins have various roles in physiological events, as well as in regulating different mechanisms in various kinds of cancers. LRP5, LRP6, and LRP8 have been shown to play important roles in a broad panel of cancers. LRP5 is highly expressed in many tissues and is involved in the modulation of glucose-induced insulin secretion, bone development, and cholesterol metabolism, as well as cancer progression. Recently, LRP5 has also been shown to play a role in chondroblastic subtype of osteosarcoma (OS) and prostate cancer and also in noncancer case such as osteoporosis. LRP6, which has been previously discovered to share the same structures as LRP5, has also been associated with many cancer progressions such as human triple negative breast cancer (TNBC), primary chronic lymphocytic leukemia (CLL), nonsmall cell lung cancer (NSCL), lung squamous cell carcinoma (LSCC), and hepatocellular carcinoma (HCC). In addition to its role in cancer progression, LRP8 (apolipoprotein E receptor 2 [APOER2]) has also been demonstrated to regulate canonical Wnt/β-catenin signaling pathway whereby this pathway plays a role in cell migration and development. Therefore, this review aimed to elucidate the role of LRP 5, 6, and 8 in regulating the cancer progression.
Collapse
|
189
|
Xu F, Xiao H, Liu R, Yang Y, Zhang M, Chen L, Chen Z, Liu P, Huang H. Paeonol Ameliorates Glucose and Lipid Metabolism in Experimental Diabetes by Activating Akt. Front Pharmacol 2019; 10:261. [PMID: 30941042 PMCID: PMC6433795 DOI: 10.3389/fphar.2019.00261] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/01/2019] [Indexed: 01/09/2023] Open
Abstract
Our previous study proved that paeonol (Pae) could lower blood glucose levels of diabetic mice. There are also a few reports of its potential use for diabetes treatment. However, the role of Pae in regulating glucose and lipid metabolism in diabetes remains largely unknown. Considering the critical role of serine/threonine kinase B (Akt) in glucose and lipid metabolism, we explored whether Pae could improve glucose and lipid metabolism disorders via Akt. Here, we found that Pae attenuated fasting blood glucose, glycosylated serum protein, serum cholesterol and triglyceride (TG), hepatic glycogen, cholesterol and TG in diabetic mice. Moreover, Pae enhanced glucokinase (GCK) and low-density lipoprotein receptor (LDLR) protein expressions, and increased the phosphorylation of Akt. In insulin-resistant HepG2 cells, Pae increased glucose uptake and decreased lipid accumulation. What’s more, Pae elevated LDLR and GCK expressions as well as Akt phosphorylation, which was consistent with the in vivo results. Knockdown and inhibition experiments of Akt revealed that Pae regulated LDLR and GCK expressions through activation of Akt. Finally, molecular docking assay indicated the steady hydrogen bond was formed between Pae and Akt2. Experiments above suggested that Pae ameliorated glucose and lipid metabolism disorders and the underlying mechanism was closely related to the activation of Akt.
Collapse
Affiliation(s)
- Futian Xu
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, China.,Department of Endocrinology, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Haiming Xiao
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, China.,Department of Endocrinology, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Renbin Liu
- Department of Traditional Chinese Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Yan Yang
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, China
| | - Meng Zhang
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, China
| | - Lihao Chen
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, China
| | - Zhiquan Chen
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, China
| | - Peiqing Liu
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, China
| | - Heqing Huang
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, China.,Department of Endocrinology, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| |
Collapse
|
190
|
Wang Q, Shen B, Qin X, Liu S, Feng J. Akt/mTOR and AMPK signaling pathways are responsible for liver X receptor agonist GW3965-enhanced gefitinib sensitivity in non-small cell lung cancer cell lines. Transl Cancer Res 2019; 8:66-76. [PMID: 35116735 PMCID: PMC8797756 DOI: 10.21037/tcr.2018.12.34] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/14/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND This study was to systemically analyze the mechanism of LXR ligand GW3965-induced sensitivity to EGFR-TKI in EGFR-TKI-resistant non-small cell lung cancer (NSCLC) cell lines. METHODS Gefitinib-resistant PC9 cell line (EGFR exon 19 deletion) was treated with single and combined treatment with GW3965 and gefitinib. Cell viability, apoptosis and autophagy were detected using MTT, flow cytometric analysis and immunofluorescent analysis, respectively. Autophagy-related signaling pathways were detected using Western blot analysis. RESULTS Inhibited cell viability by single and combined treatment with gefitinib and GW3965 were observed. Combined treatment with gefitinib and GW3965 increased LC3 II/I ratio and Beclin 1 expression. Synergistic effect of gefitinib and GW3965 on apoptosis and autophagosome accumulation as well as on the inhibition of Akt/mTOR signaling and activation of AMP-activated protein kinase (AMPK) was observed in gefitinib-resistant PC9 cells. AMPK expression showed similar profile with apoptosis and autophagy of PC9 cells. CONCLUSIONS We confirmed that GW3965 and gefitinib showed synergistic effect on Akt/mTOR inhibition, apoptosis and autophagy of lung cancer cells. Gefitinib sensitivity in PC9 cell line might be mediated by Akt/mTOR, AMPK and JNK signaling pathways.
Collapse
Affiliation(s)
- Qingbo Wang
- The Fourth Clinical School of Nanjing Medical University, Nanjing 210029, China
| | - Bo Shen
- Department of Chemotherapy, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Nanjing 210009, China
| | - Xiaobing Qin
- The Fourth Clinical School of Nanjing Medical University, Nanjing 210029, China
| | - Siwen Liu
- Department of Chemotherapy, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Nanjing 210009, China
| | - Jifeng Feng
- Department of Chemotherapy, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Nanjing 210009, China
| |
Collapse
|
191
|
Masui K, Onizuka H, Cavenee WK, Mischel PS, Shibata N. Metabolic reprogramming in the pathogenesis of glioma: Update. Neuropathology 2019; 39:3-13. [PMID: 30609184 DOI: 10.1111/neup.12535] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 11/26/2018] [Accepted: 11/28/2018] [Indexed: 12/20/2022]
Abstract
Cancer is a genetic disease that is currently classified not only by its tissue and cell type of origin but increasingly by its molecular composition. Increasingly, tumor classification and subtyping is being performed based upon the oncogene gains, tumor suppressor losses, and associated epigenetic and transcriptional features. However, cancers, including brain tumors, are also characterized by profound alterations in cellular metabolism. At present, even though signature mutations in known metabolic enzymes are recognized as being important, the metabolic landscape of tumors is not currently incorporated into tumor diagnostic categories. Here we describe a set of recent discoveries on metabolic reprogramming driven by mutations in the genes for the isocitrate dehydrogenase (IDH) and receptor tyrosine kinase (RTK) pathways, which are the most commonly observed aberrations in diffuse gliomas. We highlight the importance of oncometabolites to dynamically shift the epigenetic landscape in IDH-mutant gliomas, and c-Myc and mechanistic target of rapamycin (mTOR) complexes in RTK-mutated gliomas to adapt to the microenvironment through metabolic reprogramming. These signify the integration of the genetic mutations with metabolic reprogramming and epigenetic shifts in diffuse gliomas, shedding new light onto potential patient subsets, coupled with information to guide the development of new therapeutic opportunities against the deadly types of brain tumors.
Collapse
Affiliation(s)
- Kenta Masui
- Department of Pathology, Division of Pathological Neuroscience, Tokyo Women's Medical University, Tokyo, Japan
| | - Hiromi Onizuka
- Department of Pathology, Division of Pathological Neuroscience, Tokyo Women's Medical University, Tokyo, Japan
| | - Webster K Cavenee
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, California, USA
| | - Paul S Mischel
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, California, USA
| | - Noriyuki Shibata
- Department of Pathology, Division of Pathological Neuroscience, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
192
|
Ganjali S, Ricciuti B, Pirro M, Butler AE, Atkin SL, Banach M, Sahebkar A. High-Density Lipoprotein Components and Functionality in Cancer: State-of-the-Art. Trends Endocrinol Metab 2019; 30:12-24. [PMID: 30473465 DOI: 10.1016/j.tem.2018.10.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 01/05/2023]
Abstract
Cancer is the second leading cause of death in western countries, and thus represents a major global public health issue. Whilst it is well-recognized that diet, obesity, and smoking are risk factors for cancer, the role of low levels of high-density lipoprotein cholesterol (HDL-C) in cancer is less well appreciated. Conflicting evidence suggests that serum HDL-C levels may be either positively or negatively associated with cancer incidence and mortality. Such disparate associations are supported in part by the multitude of high-density lipoprotein (HDL) functions that can all have an impact on cancer cell biology. The aim of this review is to provide a comprehensive overview of the crosstalk between HDLs and cancer, focusing on the molecular mechanisms underlying this association.
Collapse
Affiliation(s)
- Shiva Ganjali
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Biagio Ricciuti
- Department of Medical Oncology, S. Maria della Misericordia Hospital, Perugia, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy
| | - Alexandra E Butler
- Diabetes Research Center, Qatar Biomedical Research Institute, Doha, Qatar
| | | | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
193
|
Interventional Radiologic Therapies for Hepatocellular Carcinoma: From Where We Began to Where We Are Going. MOLECULAR AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/978-3-030-21540-8_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
194
|
Pattanayak SP, Bose P, Sunita P, Siddique MUM, Lapenna A. Bergapten inhibits liver carcinogenesis by modulating LXR/PI3K/Akt and IDOL/LDLR pathways. Biomed Pharmacother 2018; 108:297-308. [DOI: 10.1016/j.biopha.2018.08.145] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 08/27/2018] [Accepted: 08/28/2018] [Indexed: 11/30/2022] Open
|
195
|
Sigma-2 Receptor/TMEM97 and PGRMC-1 Increase the Rate of Internalization of LDL by LDL Receptor through the Formation of a Ternary Complex. Sci Rep 2018; 8:16845. [PMID: 30443021 PMCID: PMC6238005 DOI: 10.1038/s41598-018-35430-3] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/06/2018] [Indexed: 12/19/2022] Open
Abstract
CRISPR/Cas gene studies were conducted in HeLa cells where either PGRMC1, TMEM97 or both proteins were removed via gene editing. A series of radioligand binding studies, confocal microscopy studies, and internalization of radiolabeled or fluorescently tagged LDL particles were then conducted in these cells. The results indicate that PGRMC1 knockout (KO) did not reduce the density of binding sites for the sigma-2 receptor (σ2R) radioligands, [125I]RHM-4 or [3H]DTG, but a reduction in the receptor affinity of both radioligands was observed. TMEM97 KO resulted in a complete loss of binding of [125I]RHM-4 and a significant reduction in binding of [3H]DTG. TMEM97 KO and PGRMC1 KO resulted in an equal reduction in the rate of uptake of fluorescently-tagged or 3H-labeled LDL, and knocking out both proteins did not result in a further rate of reduction of LDL uptake. Confocal microscopy and Proximity Ligation Assay studies indicated a clear co-localization of LDLR, PGRMC1 and TMEM97. These data indicate that the formation of a ternary complex of LDLR-PGRMC1-TMEM97 is necessary for the rapid internalization of LDL by LDLR.
Collapse
|
196
|
Harachi M, Masui K, Okamura Y, Tsukui R, Mischel PS, Shibata N. mTOR Complexes as a Nutrient Sensor for Driving Cancer Progression. Int J Mol Sci 2018; 19:ijms19103267. [PMID: 30347859 PMCID: PMC6214109 DOI: 10.3390/ijms19103267] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 10/14/2018] [Accepted: 10/14/2018] [Indexed: 02/06/2023] Open
Abstract
Recent advancement in the field of molecular cancer research has clearly revealed that abnormality of oncogenes or tumor suppressor genes causes tumor progression thorough the promotion of intracellular metabolism. Metabolic reprogramming is one of the strategies for cancer cells to ensure their survival by enabling cancer cells to obtain the macromolecular precursors and energy needed for the rapid growth. However, an orchestration of appropriate metabolic reactions for the cancer cell survival requires the precise mechanism to sense and harness the nutrient in the microenvironment. Mammalian/mechanistic target of rapamycin (mTOR) complexes are known downstream effectors of many cancer-causing mutations, which are thought to regulate cancer cell survival and growth. Recent studies demonstrate the intriguing role of mTOR to achieve the feat through metabolic reprogramming in cancer. Importantly, not only mTORC1, a well-known regulator of metabolism both in normal and cancer cell, but mTORC2, an essential partner of mTORC1 downstream of growth factor receptor signaling, controls cooperatively specific metabolism, which nominates them as an essential regulator of cancer metabolism as well as a promising candidate to garner and convey the nutrient information from the surrounding environment. In this article, we depict the recent findings on the role of mTOR complexes in cancer as a master regulator of cancer metabolism and a potential sensor of nutrients, especially focusing on glucose and amino acid sensing in cancer. Novel and detailed molecular mechanisms that amino acids activate mTOR complexes signaling have been identified. We would also like to mention the intricate crosstalk between glucose and amino acid metabolism that ensures the survival of cancer cells, but at the same time it could be exploitable for the novel intervention to target the metabolic vulnerabilities of cancer cells.
Collapse
Affiliation(s)
- Mio Harachi
- Department of Pathology, Division of Pathological Neuroscience, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| | - Kenta Masui
- Department of Pathology, Division of Pathological Neuroscience, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| | - Yukinori Okamura
- Department of Pathology, Division of Pathological Neuroscience, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| | - Ryota Tsukui
- Department of Pathology, Division of Pathological Neuroscience, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| | - Paul S Mischel
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA.
| | - Noriyuki Shibata
- Department of Pathology, Division of Pathological Neuroscience, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| |
Collapse
|
197
|
Noch EK, Ramakrishna R, Magge R. Challenges in the Treatment of Glioblastoma: Multisystem Mechanisms of Therapeutic Resistance. World Neurosurg 2018; 116:505-517. [PMID: 30049045 DOI: 10.1016/j.wneu.2018.04.022] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 02/13/2018] [Indexed: 01/14/2023]
Abstract
Glioblastoma is one of the most lethal human cancers, with poor survival despite surgery, radiation treatment, and chemotherapy. Advances in the treatment of this type of brain tumor are limited because of several resistance mechanisms. Such mechanisms involve limited drug entry into the central nervous system compartment by the blood-brain barrier and by actions of the normal brain to counteract tumor-targeting medications. In addition, the vast heterogeneity in glioblastoma contributes to significant therapeutic resistance by preventing adequate control of the entire tumor mass by a single drug and by facilitating escape mechanisms from targeted agents. The stem cell-like characteristics of glioblastoma promote resistance to chemotherapy, radiation, and immunotherapy through upregulation of efflux transporters, promotion of glioblastoma stem cell proliferation in neurogenic zones, and immune suppression, respectively. Metabolic cascades in glioblastoma prevent effective treatments through the optimization of glucose use, the use of alternative nutrient precursors for energy production, and the induction of hypoxia to enhance tumor growth. In the era of precision medicine, an assortment of molecular techniques is being developed to target an individual's unique tumor, with the hope that this personalized strategy will bypass therapeutic resistance. Although each resistance mechanism presents an array of challenges to effective treatment of glioblastoma, as the field recognizes and addresses these difficulties, future treatments may have more efficacy and promise for patients with glioblastoma.
Collapse
Affiliation(s)
- Evan K Noch
- Department of Neurology, Weill Cornell Medical College, New York, New York, USA
| | - Rohan Ramakrishna
- Department of Neurological Surgery, Weill Cornell Medical College, New York, New York, USA.
| | - Rajiv Magge
- Department of Neurology, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
198
|
Jang HJ, Kim HS, Kim JH, Lee J. The Effect of Statin Added to Systemic Anticancer Therapy: A Meta-Analysis of Randomized, Controlled Trials. J Clin Med 2018; 7:jcm7100325. [PMID: 30287761 PMCID: PMC6210992 DOI: 10.3390/jcm7100325] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/25/2018] [Accepted: 10/02/2018] [Indexed: 12/19/2022] Open
Abstract
Preclinical studies have demonstrated that statins have anticancer properties and act in an additive or synergistic way when combined with anticancer therapy. We conducted this meta-analysis of randomized, controlled phase II or III trials to evaluate the effect of statins added to systemic anticancer therapy in patients with solid cancer. A systematic literature search was performed to identify all randomized trials that were designed to investigate the effect of statins in patients with cancer using PubMed, EMBASE, Google Scholar, and Web of Science (up to August 2018). From eight randomized controlled trials, 1760 patients were included in the pooled analyses of odds ratios (ORs) with 95% confidence intervals (CIs) for grade 3–5 adverse events (AEs) and overall response rate (ORR) and hazard ratios (HRs) with 95% CIs for progression-free survival (PFS) and overall survival (OS). The addition of statin to anticancer agents did not significantly increase the incidence of grade 3–5 AEs (OR = 1.03, 95% CI: 0.81–1.29, p = 0.78). However, the combination of statin and anticancer agents did not improve ORR (OR = 0.96, 95% CI: 0.77–1.20, p = 0.72) compared with that of anticancer therapy alone. In addition, statins added to systemic anticancer therapy failed to prolong PFS (HR = 0.99, 95% CI: 0.90–1.10, p = 0.92) and OS (HR = 0.91, 95% CI: 0.76–1.11, p = 0.52). In conclusion, this meta-analysis of randomized controlled trials does not support clinical benefits of statins added to systemic anticancer therapy in patients with solid cancer.
Collapse
Affiliation(s)
- Hyun Joo Jang
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Hwasung 18450, Gyeonggi-Do, Korea.
| | - Hyeong Su Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Korea.
| | - Jung Han Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07441, Korea.
| | - Jin Lee
- Division of Gastroenterology, Department of Internal Medicine, Dongtan Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Hwasung 18450, Gyeonggi-Do, Korea.
| |
Collapse
|
199
|
Huang J, Diaz-Meco MT, Moscat J. The macroenviromental control of cancer metabolism by p62. Cell Cycle 2018; 17:2110-2121. [PMID: 30198373 DOI: 10.1080/15384101.2018.1520566] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Metabolic reprogramming is a hallmark of cancer, but most studies focus on the molecular alterations in cancer cells and much less is known on the role of cancer metabolism, from a holistic perspective, for tumor initiation and progression. Increasing epidemiological evidence highlights the tremendous impact that cancer progression has on the host metabolism, especially in cachexia. However, how this benefits the tumor still is not completely understood. Here we review current studies on fatty acid oxidation in tumor cells as a potential therapeutic target in cancer, and how the redistribution of lipids from fat reservoirs to the cancer cell in the micro- and macro-environment impacts tumorigenesis by helping the tumor fulfill its energetic demands at the expense of fat. In this context, we also discuss the critical role of the signaling adaptor p62/Sequestosome 1(SQSTM1) in adipocytes in mediating tumor-induced fat reprograming and the feedback of adipose tissue on tumor aggressiveness via osteopontin and its potential implications in obesity-promoted cancer and fat cachexia. Collectively these studies highlight the importance of the symbiotic collaboration between adipose tissue and tumor to modulate the cancer metabolic fitness.
Collapse
Affiliation(s)
- Jianfeng Huang
- a Cancer Metabolism and Signaling Networks Program , Sanford Burnham Prebys Medical Discovery Institute , La Jolla , CA , USA
| | - Maria T Diaz-Meco
- a Cancer Metabolism and Signaling Networks Program , Sanford Burnham Prebys Medical Discovery Institute , La Jolla , CA , USA
| | - Jorge Moscat
- a Cancer Metabolism and Signaling Networks Program , Sanford Burnham Prebys Medical Discovery Institute , La Jolla , CA , USA
| |
Collapse
|
200
|
Vastrad C, Vastrad B. Bioinformatics analysis of gene expression profiles to diagnose crucial and novel genes in glioblastoma multiform. Pathol Res Pract 2018; 214:1395-1461. [PMID: 30097214 DOI: 10.1016/j.prp.2018.07.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 06/27/2018] [Accepted: 07/22/2018] [Indexed: 02/07/2023]
Abstract
Therefore, the current study aimed to diagnose the genes associated in the pathogenesis of GBM. The differentially expressed genes (DEGs) were diagnosed using the limma software package. The ToppFun was used to perform pathway and Gene Ontology (GO) enrichment analysis of the DEGs. Protein-protein interaction (PPI) networks, extracted modules, miRNA-target genes regulatory network and miRNA-target genes regulatory network were used to obtain insight into the actions of DEGs. Survival analysis for DEGs carried out. A total of 701 DEGs, including 413 upregulated and 288 downregulated genes, were diagnosed between U1118MG cell line (PK 11195 treated with 1 h exposure) and U1118MG cell line (PK 11195 treated with 24 h exposure). The up-regulated genes were enriched in superpathway of pyrimidine deoxyribonucleotides de novo biosynthesis, cell cycle, cell cycle process and chromosome. The down-regulated genes were enriched in folate transformations I, biosynthesis of amino acids, cellular amino acid metabolic process and vacuolar membrane. The current study screened the genes in PPI network, extracted modules, miRNA-target genes regulatory network and miRNA-target genes regulatory network with higher degrees as hub genes, which included MYC, TERF2IP, CDK1, EEF1G, TXNIP, SLC1A5, RGS4 and IER5L Survival suggested that low expressed NR4A2, SLC7 A5, CYR61 and ID1 in patients with GBM was linked with a positive prognosis for overall survival. In conclusion, the current study could improve our understanding of the molecular mechanisms in the progression of GBM, and these crucial as well as new molecular markers might be used as therapeutic targets for GBM.
Collapse
Affiliation(s)
- Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, 580001, Karanataka, India.
| | - Basavaraj Vastrad
- Department of Pharmaceutics, SET`S College of Pharmacy, Dharwad, Karnataka, 580002, India
| |
Collapse
|