1
|
Wang Y, Zhao Y, Gong W, Hou Y, Ren J, Duan C, Zhang H, Nie X, Li J. Aspirin exposure coupled with hypoxia interferes energy metabolism, antioxidant and autophagic processes and causes liver injury in estuarine goby Mugilogobius chulae. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135071. [PMID: 38996678 DOI: 10.1016/j.jhazmat.2024.135071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024]
Abstract
Toxicity assessments of pollutants often overlook the impact of environmental factors like hypoxia, which can alter chemical toxicity with unexpected consequences. In this study, Mugilogobius chulae, an estuarine fish, was used to investigate the effects of hypoxia (H), aspirin (ASA), and their combination (H_ASA) exposure over 24, 72, and 168 h. We employed RNA-seq analysis, expression of key gene expression profiling, enzymatic activity assays, and histopathological and ultrastructural examinations of liver tissue to explore the effects and mechanisms of ASA-coupled hypoxia exposure in fish. Results showed that glycolysis was inhibited, and lipolysis was enhanced in ASA/H_ASA groups. The PPAR signaling pathway was activated, increasing fatty acid β-oxidation and lipophagy to mitigate energy crisis. Both ASA and H_ASA exposures induced p53 expression and inhibited the TOR pathway to combat environmental stress. However, a greater energy demand and heightened sensitivity to ASA were observed in H_ASA compared to ASA exposure. Disruptions in energy and detoxification pathways led to increased stress responses, including enhanced antioxidant activities, autophagy, and apoptotic events, as observed in organelle structures. Overall, sub-chronic H_ASA exposure caused liver injury in M. chulae by affecting energy metabolism, antioxidant regulation, and autophagy processes. This study highlights the influence of hypoxia on ASA toxicity in fish, providing valuable insights for ecological risk assessment of NSAIDs.
Collapse
Affiliation(s)
- Yimeng Wang
- Department of Ecology, Jinan University, Guangzhou 510632, China; Guangdong Provincial Key Laboratory of Laboratory Animals, Guangdong Laboratory Animals Monitoring Institute, Guangzhou 510663, China
| | - Yufei Zhao
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Weibo Gong
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Yingshi Hou
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Jinzhi Ren
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Chunni Duan
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Huiyu Zhang
- Department of Ecology, Jinan University, Guangzhou 510632, China
| | - Xiangping Nie
- Department of Ecology, Jinan University, Guangzhou 510632, China; Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, Jinan University, Guangzhou 510632, China.
| | - Jianjun Li
- Guangdong Provincial Key Laboratory of Laboratory Animals, Guangdong Laboratory Animals Monitoring Institute, Guangzhou 510663, China
| |
Collapse
|
2
|
Paoletti N, Supuran CT. Benzothiazole derivatives in the design of antitumor agents. Arch Pharm (Weinheim) 2024; 357:e2400259. [PMID: 38873921 DOI: 10.1002/ardp.202400259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 06/15/2024]
Abstract
Benzothiazoles are a class of heterocycles with multiple applications as anticancer, antibiotic, antiviral, and anti-inflammatory agents. Benzothiazole is a privileged scaffold in drug discovery programs for modulating a variety of biological functions. This review focuses on the design and synthesis of new benzothiazole derivatives targeting hypoxic tumors. Cancer is a major health problem, being among the leading causes of death. Tumor-hypoxic areas promote proliferation, malignancy, and resistance to drug treatment, leading to the dysregulation of key signaling pathways that involve drug targets such as vascular endothelial growth factor, epidermal growth factor receptor, hepatocyte growth factor receptor, dual-specificity protein kinase, cyclin-dependent protein kinases, casein kinase 2, Rho-related coil formation protein kinase, tunica interna endothelial cell kinase, cyclooxygenase-2, adenosine kinase, lysophosphatidic acid acyltransferases, stearoyl-CoA desaturase, peroxisome proliferator-activated receptors, thioredoxin, heat shock proteins, and carbonic anhydrase IX/XII. In turn, they regulate angiogenesis, proliferation, differentiation, and cell survival, controlling the cell cycle, inflammation, the immune system, and metabolic alterations. A wide diversity of benzothiazoles were reported over the last years to interfere with various proteins involved in tumorigenesis and, more specifically, in hypoxic tumors. Many hypoxic targets are overexpressed as a result of the hypoxia-inducible factor activation cascade and may not be present in normal tissues, providing a potential strategy for selectively targeting hypoxic cancers.
Collapse
Affiliation(s)
- Niccolò Paoletti
- Department of Neurofarba, Section of Pharmaceutical & Nutraceutical Sciences, Polo Scientifico, University of Florence, Sesto Fiorentino (Firenze), Italy
| | - Claudiu T Supuran
- Department of Neurofarba, Section of Pharmaceutical & Nutraceutical Sciences, Polo Scientifico, University of Florence, Sesto Fiorentino (Firenze), Italy
| |
Collapse
|
3
|
Zhou L, Xiang H, Liu S, Chen H, Yang Y, Zhang J, Cai W. Folic Acid Functionalized AQ4N/Gd@PDA Nanoplatform with Real-Time Monitoring of Hypoxia Relief and Enhanced Synergistic Chemo/Photothermal Therapy in Glioma. Int J Nanomedicine 2024; 19:3367-3386. [PMID: 38617794 PMCID: PMC11012807 DOI: 10.2147/ijn.s451921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/27/2024] [Indexed: 04/16/2024] Open
Abstract
Purpose Hypoxia is often associated with glioma chemoresistance, and alleviating hypoxia is also crucial for improving treatment efficacy. However, although there are already some methods that can improve efficacy by alleviating hypoxia, real-time monitoring that can truly achieve hypoxia relief and efficacy feedback still needs to be explored. Methods AQ4N/Gd@PDA-FA nanoparticles (AGPF NPs) were synthesized using a one-pot method and were characterized. The effects of AGPF NPs on cell viability, cellular uptake, and apoptosis were investigated using the U87 cell line. Moreover, the effectiveness of AGPF NPs in alleviating hypoxia was explored in tumor-bearing mice through photoacoustic imaging. In addition, the diagnosis and treatment effect of AGPF NPs were evaluated by magnetic resonance imaging (MRI) and bioluminescent imaging (BLI) on orthotopic glioma mice respectively. Results In vitro experiments showed that AGPF NPs had good dispersion, stability, and controlled release. AGPF NPs were internalized by cells through endocytosis, and could significantly reduce the survival rate of U87 cells and increase apoptosis under irradiation. In addition, we monitored blood oxygen saturation at the tumor site in real-time through photoacoustic imaging (PAI), and the results showed that synergistic mild-photothermal therapy/chemotherapy effectively alleviated tumor hypoxia. Finally, in vivo anti-tumor experiments have shown that synergistic therapy can effectively alleviate hypoxia and inhibit the growth of orthotopic gliomas. Conclusion This work not only provides an effective means for real-time monitoring of the dynamic feedback between tumor hypoxia relief and therapeutic efficacy, but also offers a potential approach for the clinical treatment of gliomas.
Collapse
Affiliation(s)
- Longjiang Zhou
- Department of Neurology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, People’s Republic of China
| | - Haitao Xiang
- Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, 215028, People’s Republic of China
| | - Susu Liu
- School of Life Science and Technology, Xidian University and Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi’an, 710126, People’s Republic of China
| | - Honglin Chen
- Department of Neurosurgery, Suqian First Hospital, Suqian, 223800, People’s Republic of China
| | - Yuanwei Yang
- Department of Neurosurgery, Suqian First Hospital, Suqian, 223800, People’s Republic of China
| | - Jianyong Zhang
- Department of Neurosurgery, Suqian First Hospital, Suqian, 223800, People’s Republic of China
| | - Wei Cai
- Department of Neurosurgery, Suqian First Hospital, Suqian, 223800, People’s Republic of China
| |
Collapse
|
4
|
Pacheco-Torres J, Sharma RK, Mironchik Y, Wildes F, Brennen WN, Artemov D, Krishnamachary B, Bhujwalla ZM. Prostate fibroblasts and prostate cancer associated fibroblasts exhibit different metabolic, matrix degradation and PD-L1 expression responses to hypoxia. Front Mol Biosci 2024; 11:1354076. [PMID: 38584702 PMCID: PMC10995317 DOI: 10.3389/fmolb.2024.1354076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/06/2024] [Indexed: 04/09/2024] Open
Abstract
Fibroblasts are versatile cells that play a major role in wound healing by synthesizing and remodeling the extracellular matrix (ECM). In cancers, fibroblasts play an expanded role in tumor progression and dissemination, immunosuppression, and metabolic support of cancer cells. In prostate cancer (PCa), fibroblasts have been shown to induce growth and increase metastatic potential. To further understand differences in the functions of human PCa associated fibroblasts (PCAFs) compared to normal prostate fibroblasts (PFs), we investigated the metabolic profile and ECM degradation characteristics of PFs and PCAFs using a magnetic resonance imaging and spectroscopy compatible intact cell perfusion assay. To further understand how PFs and PCAFs respond to hypoxic tumor microenvironments that are often observed in PCa, we characterized the effects of hypoxia on PF and PCAF metabolism, invasion and PD-L1 expression. We found that under normoxia, PCAFs displayed decreased ECM degradation compared to PFs. Under hypoxia, ECM degradation by PFs increased, whereas PCAFs exhibited decreased ECM degradation. Under both normoxia and hypoxia, PCAFs and PFs showed significantly different metabolic profiles. PD-L1 expression was intrinsically higher in PCAFs compared to PFs. Under hypoxia, PD-L1 expression increased in PCAFs but not in PFs. Our data suggest that PCAFs may not directly induce ECM degradation to assist in tumor dissemination, but may instead create an immune suppressive tumor microenvironment that further increases under hypoxic conditions. Our data identify the intrinsic metabolic, ECM degradation and PD-L1 expression differences between PCAFs and PFs under normoxia and hypoxia that may provide novel targets in PCa treatment.
Collapse
Affiliation(s)
- Jesus Pacheco-Torres
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC, Madrid, Spain
| | - Raj Kumar Sharma
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | | | - Flonne Wildes
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - W. Nathaniel Brennen
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Dmitri Artemov
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Zaver M. Bhujwalla
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
5
|
Qin X, Wang B, Lu X, Song Y, Wang W. Identification and Validation of a PEX5-Dependent Signature for Prognostic Prediction in Glioma. Biomolecules 2024; 14:314. [PMID: 38540734 PMCID: PMC10967733 DOI: 10.3390/biom14030314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/22/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Gliomas, the most prevalent and lethal form of brain cancer, are known to exhibit metabolic alterations that facilitate tumor growth, invasion, and resistance to therapies. Peroxisomes, essential organelles responsible for fatty acid oxidation and reactive oxygen species (ROS) homeostasis, rely on the receptor PEX5 for the import of metabolic enzymes into their matrix. However, the prognostic significance of peroxisomal enzymes for glioma patients remains unclear. In this study, we elucidate that PEX5 is indispensable for the cell growth, migration, and invasion of glioma cells. We establish a robust prognosis model based on the expression of peroxisomal enzymes, whose localization relies on PEX5. This PEX5-dependent signature not only serves as a robust prognosis model capable of accurately predicting outcomes for glioma patients, but also effectively distinguishes several clinicopathological features, including the grade, isocitrate dehydrogenase (IDH) mutation, and 1p19q codeletion status. Furthermore, we developed a nomogram that integrates the prognostic model with other clinicopathological factors, demonstrating highly accurate performance in estimating patient survival. Patients classified into the high-risk group based on our prognostic model exhibited an immunosuppressive microenvironment. Finally, our validation reveals that the elevated expression of GSTK1, an antioxidant enzyme within the signature, promotes the cell growth and migration of glioma cells, with this effect dependent on the peroxisomal targeting signal recognized by PEX5. These findings identify the PEX5-dependent signature as a promising prognostic tool for gliomas.
Collapse
Affiliation(s)
| | | | | | | | - Wei Wang
- Department of Human Anatomy, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China
| |
Collapse
|
6
|
Rahane D, Dhingra T, Chalavady G, Datta A, Ghosh B, Rana N, Borah A, Saraf S, Bhattacharya P. Hypoxia and its effect on the cellular system. Cell Biochem Funct 2024; 42:e3940. [PMID: 38379257 DOI: 10.1002/cbf.3940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 02/22/2024]
Abstract
Eukaryotic cells utilize oxygen for different functions of cell organelles owing to cellular survival. A balanced oxygen homeostasis is an essential requirement to maintain the regulation of normal cellular systems. Any changes in the oxygen level are stressful and can alter the expression of different homeostasis regulatory genes and proteins. Lack of oxygen or hypoxia results in oxidative stress and formation of hypoxia inducible factors (HIF) and reactive oxygen species (ROS). Substantial cellular damages due to hypoxia have been reported to play a major role in various pathological conditions. There are different studies which demonstrated that the functions of cellular system are disrupted by hypoxia. Currently, study on cellular effects following hypoxia is an important field of research as it not only helps to decipher different signaling pathway modulation, but also helps to explore novel therapeutic strategies. On the basis of the beneficial effect of hypoxia preconditioning of cellular organelles, many therapeutic investigations are ongoing as a promising disease management strategy in near future. Hence, the present review discusses about the effects of hypoxia on different cellular organelles, mechanisms and their involvement in the progression of different diseases.
Collapse
Affiliation(s)
- Dipali Rahane
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Tannu Dhingra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Guruswami Chalavady
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Aishika Datta
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Bijoyani Ghosh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Nikita Rana
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| | - Shailendra Saraf
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| |
Collapse
|
7
|
Gao D, Zhou Q, Hou D, Zhang X, Ge Y, Zhu Q, Yin J, Qi X, Liu Y, Lou M, Zhou L, Bi Y. A novel peroxisome-related gene signature predicts clinical prognosis and is associated with immune microenvironment in low-grade glioma. PeerJ 2024; 12:e16874. [PMID: 38406287 PMCID: PMC10885797 DOI: 10.7717/peerj.16874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 01/11/2024] [Indexed: 02/27/2024] Open
Abstract
Low-grade glioma (LGG), a common primary tumor, mainly originates from astrocytes and oligodendrocytes. Increasing evidence has shown that peroxisomes function in the regulation of tumorigenesis and development of cancer. However, the prognostic value of peroxisome-related genes (PRGs) in LGG has not been reported. Therefore, it is necessary to construct a prognostic risk model for LGG patients based on the expression profiles of peroxisome-related genes. Our study mainly concentrated on developing a peroxisome-related gene signature for overall survival (OS) prediction in LGG patients. First, according to these peroxisome-related genes, all LGG patients from The Cancer Genome Atlas (TCGA) database could be divided into two subtypes. Univariate Cox regression analysis was used to find prognostic peroxisome-related genes in TCGA_LGG dataset, and least absolute shrinkage and selection operator Cox regression analysis was employed to establish a 14-gene signature. The risk score based on the signature was positively associated with unfavorable prognosis. Then, multivariate Cox regression incorporating additional clinical characteristics showed that the 14-gene signature was an independent predictor of LGG. Time-dependent ROC curves revealed good performance of this prognostic signature in LGG patients. The performance about predicting OS of LGG was validated using the GSE107850 dataset derived from the Gene Expression Omnibus (GEO) database. Furethermore, we constructed a nomogram model based on the gene signature and age, which showed a better prognostic power. Gene ontology (GO) and Kyoto Encylopedia of Genes and Genomes (KEGG) analyses showed that neuroactive ligand-receptor interaction and phagosome were enriched and that the immune status was decreased in the high-risk group. Finally, cell counting kit-8 (CCK8) were used to detect cell proliferation of U251 and A172 cells. Inhibition of ATAD1 (ATPase family AAA domain-containing 1) and ACBD5 (Acyl-CoA binding-domain-containing-5) expression led to significant inhibition of U251 and A172 cell proliferation. Flow cytometry detection showed that ATAD1 and ACBD5 could induce apoptosis of U251 and A172 cells. Therefore, through bioinformatics methods and cell experiments, our study developed a new peroxisome-related gene signature that migh t help improve personalized OS prediction in LGG patients.
Collapse
Affiliation(s)
- Dandan Gao
- Oncology and Hematology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Qiangyi Zhou
- Neurosurgery, Shanghai General Hospital, Shanghai, China
| | - Dianqi Hou
- Neurosurgery, Shanghai General Hospital, Shanghai, China
| | - Xiaoqing Zhang
- Oncology and Hematology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Yiqin Ge
- Department of Neurosurgery, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingwei Zhu
- Neurosurgery, Shanghai General Hospital, Shanghai, China
| | - Jian Yin
- Neurosurgery, Shanghai General Hospital, Shanghai, China
| | - Xiangqian Qi
- Neurosurgery, Shanghai General Hospital, Shanghai, China
| | - Yaohua Liu
- Neurosurgery, Shanghai General Hospital, Shanghai, China
| | - Meiqing Lou
- Neurosurgery, Shanghai General Hospital, Shanghai, China
| | - Li Zhou
- Department of Oncology, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Yunke Bi
- Neurosurgery, Shanghai General Hospital, Shanghai, China
| |
Collapse
|
8
|
Li Y, Pan Y, Zhao X, Wu S, Li F, Wang Y, Liu B, Zhang Y, Gao X, Wang Y, Zhou H. Peroxisome proliferator-activated receptors: A key link between lipid metabolism and cancer progression. Clin Nutr 2024; 43:332-345. [PMID: 38142478 DOI: 10.1016/j.clnu.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/26/2023]
Abstract
Lipids represent the essential components of membranes, serve as fuels for high-energy processes, and play crucial roles in signaling and cellular function. One of the key hallmarks of cancer is the reprogramming of metabolic pathways, especially abnormal lipid metabolism. Alterations in lipid uptake, lipid desaturation, de novo lipogenesis, lipid droplets, and fatty acid oxidation in cancer cells all contribute to cell survival in a changing microenvironment by regulating feedforward oncogenic signals, key oncogenic functions, oxidative and other stresses, immune responses, or intercellular communication. Peroxisome proliferator-activated receptors (PPARs) are transcription factors activated by fatty acids and act as core lipid sensors involved in the regulation of lipid homeostasis and cell fate. In addition to regulating whole-body energy homeostasis in physiological states, PPARs play a key role in lipid metabolism in cancer, which is receiving increasing research attention, especially the fundamental molecular mechanisms and cancer therapies targeting PPARs. In this review, we discuss how cancer cells alter metabolic patterns and regulate lipid metabolism to promote their own survival and progression through PPARs. Finally, we discuss potential therapeutic strategies for targeting PPARs in cancer based on recent studies from the last five years.
Collapse
Affiliation(s)
- Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yujie Pan
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Xiaodong Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Shouwang Wu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yuxiong Wang
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yanghe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Xin Gao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
9
|
Darwish A, Pammer M, Gallyas F, Vígh L, Balogi Z, Juhász K. Emerging Lipid Targets in Glioblastoma. Cancers (Basel) 2024; 16:397. [PMID: 38254886 PMCID: PMC10814456 DOI: 10.3390/cancers16020397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
GBM accounts for most of the fatal brain cancer cases, making it one of the deadliest tumor types. GBM is characterized by severe progression and poor prognosis with a short survival upon conventional chemo- and radiotherapy. In order to improve therapeutic efficiency, considerable efforts have been made to target various features of GBM. One of the targetable features of GBM is the rewired lipid metabolism that contributes to the tumor's aggressive growth and penetration into the surrounding brain tissue. Lipid reprogramming allows GBM to acquire survival, proliferation, and invasion benefits as well as supportive modulation of the tumor microenvironment. Several attempts have been made to find novel therapeutic approaches by exploiting the lipid metabolic reprogramming in GBM. In recent studies, various components of de novo lipogenesis, fatty acid oxidation, lipid uptake, and prostaglandin synthesis have been considered promising targets in GBM. Emerging data also suggest a significant role hence therapeutic potential of the endocannabinoid metabolic pathway in GBM. Here we review the lipid-related GBM characteristics in detail and highlight specific targets with their potential therapeutic use in novel antitumor approaches.
Collapse
Affiliation(s)
- Ammar Darwish
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Milán Pammer
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Ferenc Gallyas
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - László Vígh
- Institute of Biochemistry, HUN-REN Biological Research Center, 6726 Szeged, Hungary
| | - Zsolt Balogi
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Kata Juhász
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
10
|
Jin Y, Tan Y, Wu J, Ren Z. Lipid droplets: a cellular organelle vital in cancer cells. Cell Death Discov 2023; 9:254. [PMID: 37474495 PMCID: PMC10359296 DOI: 10.1038/s41420-023-01493-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/24/2023] [Accepted: 06/16/2023] [Indexed: 07/22/2023] Open
Abstract
Lipid droplets (LDs) are cellular organelles comprising a core of neutral lipids (glycerides, sterols) encased within a single phospholipid membrane, responsible for storing surplus lipids and furnishing cellular energy. LDs engage in lipid synthesis, catabolism, and transport processes by interacting with other organelles (e.g., endoplasmic reticulum, mitochondria), and they play critical roles in regulating cellular stress and immunity. Recent research has uncovered that an elevated number of LDs is a hallmark of cancer cells, attributable to their enhanced lipid uptake and synthesis capacity, with lipids stored as LDs. Depletion of LDs in cancer cells induces apoptosis, prompting the emergence of small molecule antitumor drugs targeting LDs or key factors (e.g., FASN, SCD1) within the lipid synthesis pathway. Advancements in LD isolation and artificial synthesis have demonstrated their potential applicability in antitumor research. LDs extracted from murine adipose tissue and incubated with lipophilic antitumor drugs yield drug-coated LDs, which promote apoptosis in cancer cells. Furthermore, LDs have been employed as biological lenses to augment the resolution of subcellular structures (microfilaments, microtubules), facilitating the observation of intricate structures within thicker cells, including cancer cells. This review delineates the functional and metabolic mechanisms of LDs in cancer cells and encapsulates recent progress in LD-centered antitumor research, offering novel insights for tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Yi Jin
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, Hubei, P. R. China
| | - Yanjie Tan
- Institute of Biomedical Sciences, Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, P. R. China
| | - Jian Wu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, Hubei, P. R. China
| | - Zhuqing Ren
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, Hubei, P. R. China.
- Hubei Hongshan Laboratory, Wuhan, P. R. China.
| |
Collapse
|
11
|
Hammoudeh N, Soukkarieh C, Murphy DJ, Hanano A. Mammalian lipid droplets: structural, pathological, immunological and anti-toxicological roles. Prog Lipid Res 2023; 91:101233. [PMID: 37156444 DOI: 10.1016/j.plipres.2023.101233] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
Mammalian lipid droplets (LDs) are specialized cytosolic organelles consisting of a neutral lipid core surrounded by a membrane made up of a phospholipid monolayer and a specific population of proteins that varies according to the location and function of each LD. Over the past decade, there have been significant advances in the understanding of LD biogenesis and functions. LDs are now recognized as dynamic organelles that participate in many aspects of cellular homeostasis plus other vital functions. LD biogenesis is a complex, highly-regulated process with assembly occurring on the endoplasmic reticulum although aspects of the underpinning molecular mechanisms remain elusive. For example, it is unclear how many enzymes participate in the biosynthesis of the neutral lipid components of LDs and how this process is coordinated in response to different metabolic cues to promote or suppress LD formation and turnover. In addition to enzymes involved in the biosynthesis of neutral lipids, various scaffolding proteins play roles in coordinating LD formation. Despite their lack of ultrastructural diversity, LDs in different mammalian cell types are involved in a wide range of biological functions. These include roles in membrane homeostasis, regulation of hypoxia, neoplastic inflammatory responses, cellular oxidative status, lipid peroxidation, and protection against potentially toxic intracellular fatty acids and lipophilic xenobiotics. Herein, the roles of mammalian LDs and their associated proteins are reviewed with a particular focus on their roles in pathological, immunological and anti-toxicological processes.
Collapse
Affiliation(s)
- Nour Hammoudeh
- Department of Animal Biology, Faculty of Sciences, University of Damascus, Damascus, Syria
| | - Chadi Soukkarieh
- Department of Animal Biology, Faculty of Sciences, University of Damascus, Damascus, Syria
| | - Denis J Murphy
- School of Applied Sciences, University of South Wales, Pontypridd, CF37 1DL, Wales, United Kingdom..
| | - Abdulsamie Hanano
- Department of Molecular Biology and Biotechnology, Atomic Energy Commission of Syria (AECS), P.O. Box 6091, Damascus, Syria..
| |
Collapse
|
12
|
Korbecki J, Bosiacki M, Gutowska I, Chlubek D, Baranowska-Bosiacka I. Biosynthesis and Significance of Fatty Acids, Glycerophospholipids, and Triacylglycerol in the Processes of Glioblastoma Tumorigenesis. Cancers (Basel) 2023; 15:cancers15072183. [PMID: 37046844 PMCID: PMC10093493 DOI: 10.3390/cancers15072183] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/01/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
One area of glioblastoma research is the metabolism of tumor cells and detecting differences between tumor and healthy brain tissue metabolism. Here, we review differences in fatty acid metabolism, with a particular focus on the biosynthesis of saturated fatty acids (SFA), monounsaturated fatty acids (MUFA), and polyunsaturated fatty acids (PUFA) by fatty acid synthase (FASN), elongases, and desaturases. We also describe the significance of individual fatty acids in glioblastoma tumorigenesis, as well as the importance of glycerophospholipid and triacylglycerol synthesis in this process. Specifically, we show the significance and function of various isoforms of glycerol-3-phosphate acyltransferases (GPAT), 1-acylglycerol-3-phosphate O-acyltransferases (AGPAT), lipins, as well as enzymes involved in the synthesis of phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylserine (PS), phosphatidylinositol (PI), and cardiolipin (CL). This review also highlights the involvement of diacylglycerol O-acyltransferase (DGAT) in triacylglycerol biosynthesis. Due to significant gaps in knowledge, the GEPIA database was utilized to demonstrate the significance of individual enzymes in glioblastoma tumorigenesis. Finally, we also describe the significance of lipid droplets in glioblastoma and the impact of fatty acid synthesis, particularly docosahexaenoic acid (DHA), on cell membrane fluidity and signal transduction from the epidermal growth factor receptor (EGFR).
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zyty 28 Str., 65-046 Zielona Góra, Poland
| | - Mateusz Bosiacki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
- Department of Functional Diagnostics and Physical Medicine, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, Żołnierska 54 Str., 71-210 Szczecin, Poland
| | - Izabela Gutowska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| |
Collapse
|
13
|
Aoki T, Kinoshita J, Munesue S, Hamabe-Horiike T, Yamaguchi T, Nakamura Y, Okamoto K, Moriyama H, Nakamura K, Harada S, Yamamoto Y, Inaki N, Fushida S. Hypoxia-Induced CD36 Expression in Gastric Cancer Cells Promotes Peritoneal Metastasis via Fatty Acid Uptake. Ann Surg Oncol 2022; 30:3125-3136. [PMID: 36042102 PMCID: PMC10085939 DOI: 10.1245/s10434-022-12465-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/08/2022] [Indexed: 12/21/2022]
Abstract
Abstract
Background
The lipid scavenger receptor cluster of differentiation 36 (CD36) has been shown to have a pro-metastatic function in several cancers. Adipose tissue, a favorable site for peritoneal metastasis (PM) from gastric cancer (GC), promotes this process by providing free fatty acids (FFAs); however, the role of CD36 in PM progression from GC remains to be elucidated.
Materials and Methods
We evaluated CD36 expression in the GC cells under various conditions. CD36 overexpressing (CD36OE) MKN45 cells were prepared and their migration and invasive properties were assessed. A PM mouse model was used to investigate the biological effects of palmitic acid (PA) and CD36. Furthermore, we examined the clinical role of CD36 expression in 82 human PM samples by immunohistochemical staining.
Results
Hypoxia markedly increased CD36 expression in GC cells. In normoxia, only CD36OE MKN45 cells treated with PA showed an increase in migration and invasion abilities. An increased expression of active Rac1 and Cdc42 was observed, which decreased following etomoxir treatment. Conversely, hypoxia increased those capacities of both vector and CD36OE MKN45 cells. In a mouse model transplanted with CD36OE MKN45 cells, more peritoneal tumors were observed in the high-fat diet group than those in the normal diet group. In clinical samples, 80% of PM lesions expressed CD36, consistent with hypoxic regions, indicating a significant association with prognosis.
Conclusion
Our findings indicate that a hypoxia in the peritoneal cavity induces CD36 expression in GC cells, which contributes to PM through the uptake of FFAs.
Collapse
Affiliation(s)
- Tatsuya Aoki
- Department of Gastrointestinal Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Jun Kinoshita
- Department of Gastrointestinal Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan.
| | - Seiichi Munesue
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Toshihide Hamabe-Horiike
- Center for Biomedical Research and Education, School of Medicine, Kanazawa University, Kanazawa, Japan
| | - Takahisa Yamaguchi
- Department of Gastrointestinal Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yusuke Nakamura
- Department of Gastrointestinal Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Koichi Okamoto
- Department of Gastrointestinal Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hideki Moriyama
- Department of Gastrointestinal Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Keishi Nakamura
- Department of Gastrointestinal Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Shinichi Harada
- Center for Biomedical Research and Education, School of Medicine, Kanazawa University, Kanazawa, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Noriyuki Inaki
- Department of Gastrointestinal Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Sachio Fushida
- Department of Gastrointestinal Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| |
Collapse
|
14
|
Smolič T, Zorec R, Vardjan N. Pathophysiology of Lipid Droplets in Neuroglia. Antioxidants (Basel) 2021; 11:22. [PMID: 35052526 PMCID: PMC8773017 DOI: 10.3390/antiox11010022] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, increasing evidence regarding the functional importance of lipid droplets (LDs), cytoplasmic storage organelles in the central nervous system (CNS), has emerged. Although not abundantly present in the CNS under normal conditions in adulthood, LDs accumulate in the CNS during development and aging, as well as in some neurologic disorders. LDs are actively involved in cellular lipid turnover and stress response. By regulating the storage of excess fatty acids, cholesterol, and ceramides in addition to their subsequent release in response to cell needs and/or environmental stressors, LDs are involved in energy production, in the synthesis of membranes and signaling molecules, and in the protection of cells against lipotoxicity and free radicals. Accumulation of LDs in the CNS appears predominantly in neuroglia (astrocytes, microglia, oligodendrocytes, ependymal cells), which provide trophic, metabolic, and immune support to neuronal networks. Here we review the most recent findings on the characteristics and functions of LDs in neuroglia, focusing on astrocytes, the key homeostasis-providing cells in the CNS. We discuss the molecular mechanisms affecting LD turnover in neuroglia under stress and how this may protect neural cell function. We also highlight the role (and potential contribution) of neuroglial LDs in aging and in neurologic disorders.
Collapse
Affiliation(s)
- Tina Smolič
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.S.); (R.Z.)
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.S.); (R.Z.)
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia
| | - Nina Vardjan
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; (T.S.); (R.Z.)
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia
| |
Collapse
|
15
|
Ryan F, Khoshnam SE, Khodagholi F, Ashabi G, Ahmadiani A. How cytosolic compartments play safeguard functions against neuroinflammation and cell death in cerebral ischemia. Metab Brain Dis 2021; 36:1445-1467. [PMID: 34173922 DOI: 10.1007/s11011-021-00770-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 06/06/2021] [Indexed: 11/26/2022]
Abstract
Ischemic stroke is the second leading cause of mortality and disability globally. Neuronal damage following ischemic stroke is rapid and irreversible, and eventually results in neuronal death. In addition to activation of cell death signaling, neuroinflammation is also considered as another pathogenesis that can occur within hours after cerebral ischemia. Under physiological conditions, subcellular organelles play a substantial role in neuronal functionality and viability. However, their functions can be remarkably perturbed under neurological disorders, particularly cerebral ischemia. Therefore, their biochemical and structural response has a determining role in the sequel of neuronal cells and the progression of disease. However, their effects on cell death and neuroinflammation, as major underlying mechanisms of ischemic stroke, are still not understood. This review aims to provide a comprehensive overview of the contribution of each organelle on these pathological processes after ischemic stroke.
Collapse
Affiliation(s)
- Fari Ryan
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Centre, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghorbangol Ashabi
- Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, PO Box: 1417613151, Tehran, Iran.
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Léger JAD, Athanasio CG, Zhera A, Chauhan MF, Simmons DBD. Hypoxic responses in Oncorhynchus mykiss involve angiogenesis, lipid, and lactate metabolism, which may be triggered by the cortisol stress response and epigenetic methylation. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 39:100860. [PMID: 34126312 DOI: 10.1016/j.cbd.2021.100860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 05/14/2021] [Accepted: 06/01/2021] [Indexed: 10/21/2022]
Abstract
The incidence of hypoxia in water bodies is increasing more rapidly than aquatic life can adapt. This study aimed to determine the effects of hypoxia on fish physiology, as well as protein expression through proteomics. To do this, 40 rainbow trout were divided into normoxic control (11.5 mg/L dissolved oxygen) and hypoxic treatment (5 mg/L dissolved oxygen) tanks for a period of 7 days. Fish were then anesthetized and blood was sampled. Fish were then euthanized and heart and liver samples were taken. Blood glucose, cortisol and lipid, body and liver mass, fork length, hematocrit and, blood cell counts and global heart methylation were measured. Red blood cell counts were significantly lower, while hematocrit and mean corpuscular volume were significantly higher in the hypoxic treatment. Global DNA methylation was significantly decreased in hypoxic heart tissue. Plasma cortisol and 18:1 monoacylglyerol increased, while 15:0-18:1 phosphatidylethanolamine, and 18:1 lysophosphatidylethanolamine decreased in plasma of rainbow trout under hypoxic conditions. Plasma proteomics revealed 70 significantly altered proteins (p < 0.05) in the hypoxia treatment (Data are available via ProteomeXchange with identifier PXD026589). Many of these molecular changes appear to be related to the observed increase in red blood cell volume and epigenetic modifications, as well as to angiogenesis, lipid, and glucose metabolism. This study highlights a range of cellular and molecular responses in the blood and plasma of freshwater fish that may be phenotypic adaptions to hypoxia, and that could aid in diagnosing the health status of wild fish populations using several, potential, discovered biomarkers.
Collapse
Affiliation(s)
- Jessica A D Léger
- University of Ontario Institute of Technology, 2000 Simcoe Street North, Oshawa, ON L1G 0C5, Canada.
| | - Camila G Athanasio
- University of Ontario Institute of Technology, 2000 Simcoe Street North, Oshawa, ON L1G 0C5, Canada
| | - Aaleen Zhera
- University of Ontario Institute of Technology, 2000 Simcoe Street North, Oshawa, ON L1G 0C5, Canada.
| | - Mohammed Faiz Chauhan
- University of Ontario Institute of Technology, 2000 Simcoe Street North, Oshawa, ON L1G 0C5, Canada.
| | - Denina B D Simmons
- University of Ontario Institute of Technology, 2000 Simcoe Street North, Oshawa, ON L1G 0C5, Canada.
| |
Collapse
|
17
|
Smolič T, Tavčar P, Horvat A, Černe U, Halužan Vasle A, Tratnjek L, Kreft ME, Scholz N, Matis M, Petan T, Zorec R, Vardjan N. Astrocytes in stress accumulate lipid droplets. Glia 2021; 69:1540-1562. [PMID: 33609060 PMCID: PMC8248329 DOI: 10.1002/glia.23978] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 01/14/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023]
Abstract
When the brain is in a pathological state, the content of lipid droplets (LDs), the lipid storage organelles, is increased, particularly in glial cells, but rarely in neurons. The biology and mechanisms leading to LD accumulation in astrocytes, glial cells with key homeostatic functions, are poorly understood. We imaged fluorescently labeled LDs by microscopy in isolated and brain tissue rat astrocytes and in glia-like cells in Drosophila brain to determine the (sub)cellular localization, mobility, and content of LDs under various stress conditions characteristic for brain pathologies. LDs exhibited confined mobility proximal to mitochondria and endoplasmic reticulum that was attenuated by metabolic stress and by increased intracellular Ca2+ , likely to enhance the LD-organelle interaction imaged by electron microscopy. When de novo biogenesis of LDs was attenuated by inhibition of DGAT1 and DGAT2 enzymes, the astrocyte cell number was reduced by ~40%, suggesting that in astrocytes LD turnover is important for cell survival and/or proliferative cycle. Exposure to noradrenaline, a brain stress response system neuromodulator, and metabolic and hypoxic stress strongly facilitated LD accumulation in astrocytes. The observed response of stressed astrocytes may be viewed as a support for energy provision, but also to be neuroprotective against the stress-induced lipotoxicity.
Collapse
Affiliation(s)
- Tina Smolič
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
| | - Petra Tavčar
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
| | - Anemari Horvat
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia.,Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| | - Urška Černe
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
| | - Ana Halužan Vasle
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
| | - Larisa Tratnjek
- Faculty of Medicine, Institute of Cell Biology, University of Ljubljana, Ljubljana, Slovenia
| | - Mateja Erdani Kreft
- Faculty of Medicine, Institute of Cell Biology, University of Ljubljana, Ljubljana, Slovenia
| | - Nicole Scholz
- Division of General Biochemistry, Medical Faculty, Rudolf Schönheimer Institute of Biochemistry, Leipzig University, Leipzig, Germany
| | - Maja Matis
- Medical Faculty, Institute of Cell Biology, University of Münster, Münster, Germany.,Cells in Motion Interfaculty Centre, University of Münster, Münster, Germany
| | - Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia.,Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| | - Nina Vardjan
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia.,Laboratory of Cell Engineering, Celica Biomedical, Ljubljana, Slovenia
| |
Collapse
|
18
|
Matsushita Y, Nakagawa H, Koike K. Lipid Metabolism in Oncology: Why It Matters, How to Research, and How to Treat. Cancers (Basel) 2021; 13:474. [PMID: 33530546 PMCID: PMC7865757 DOI: 10.3390/cancers13030474] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/11/2022] Open
Abstract
Lipids in our body, which are mainly composed of fatty acids, triacylglycerides, sphingolipids, phospholipids, and cholesterol, play important roles at the cellular level. In addition to being energy sources and structural components of biological membranes, several types of lipids serve as signaling molecules or secondary messengers. Metabolic reprogramming has been recognized as a hallmark of cancer, but changes in lipid metabolism in cancer have received less attention compared to glucose or glutamine metabolism. However, recent innovations in mass spectrometry- and chromatography-based lipidomics technologies have increased our understanding of the role of lipids in cancer. Changes in lipid metabolism, so-called "lipid metabolic reprogramming", can affect cellular functions including the cell cycle, proliferation, growth, and differentiation, leading to carcinogenesis. Moreover, interactions between cancer cells and adjacent immune cells through altered lipid metabolism are known to support tumor growth and progression. Characterization of cancer-specific lipid metabolism can be used to identify novel metabolic targets for cancer treatment, and indeed, several clinical trials are currently underway. Thus, we discuss the latest findings on the roles of lipid metabolism in cancer biology and introduce current advances in lipidomics technologies, focusing on their applications in cancer research.
Collapse
Affiliation(s)
| | - Hayato Nakagawa
- Department of Gastroenterology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; (Y.M.); (K.K.)
| | | |
Collapse
|
19
|
Povero D, Johnson SM, Liu J. Hypoxia, hypoxia-inducible gene 2 (HIG2)/HILPDA, and intracellular lipolysis in cancer. Cancer Lett 2020; 493:71-79. [PMID: 32818550 PMCID: PMC11218043 DOI: 10.1016/j.canlet.2020.06.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/27/2020] [Accepted: 06/13/2020] [Indexed: 12/16/2022]
Abstract
Tumor tissues are chronically exposed to hypoxia owing to aberrant vascularity. Hypoxia induces metabolic alterations in cancer, thereby promoting aggressive malignancy and metastasis. While previous efforts largely focused on adaptive responses in glucose and glutamine metabolism, recent studies have begun to yield important insight into the hypoxic regulation of lipid metabolic reprogramming in cancer. Emerging evidence points to lipid droplet (LD) accumulation as a hallmark of hypoxic cancer cells. One critical underlying mechanism involves the inhibition of adipose triglyceride lipase (ATGL)-mediated intracellular lipolysis by a small protein encoded by hypoxia-inducible gene 2 (HIG2), also known as hypoxia inducible lipid droplet associated (HILPDA). In this review we summarize and discuss recent key findings on hypoxia-dependent regulation of metabolic adaptations especially lipolysis in cancer. We also pose several questions and hypotheses pertaining to the metabolic impact of lipolytic regulation in cancer under hypoxia and during hypoxia-reoxygenation transition.
Collapse
Affiliation(s)
- Davide Povero
- From Department of Biochemistry and Molecular Biology, Rochester, MN, 55905, USA; Division of Endocrinology, Rochester, MN, 55905, USA
| | - Scott M Johnson
- From Department of Biochemistry and Molecular Biology, Rochester, MN, 55905, USA; Mayo Clinic College of Medicine & Science, Rochester, MN, 55905, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, 55905, USA
| | - Jun Liu
- From Department of Biochemistry and Molecular Biology, Rochester, MN, 55905, USA; Division of Endocrinology, Rochester, MN, 55905, USA.
| |
Collapse
|
20
|
WY-14643 Regulates CYP1B1 Expression through Peroxisome Proliferator-Activated Receptor α-Mediated Signaling in Human Breast Cancer Cells. Int J Mol Sci 2019; 20:ijms20235928. [PMID: 31775380 PMCID: PMC6928855 DOI: 10.3390/ijms20235928] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 12/14/2022] Open
Abstract
Human cytochrome P450 1B1 (CYP1B1)-mediated biotransformation of endobiotics and xenobiotics plays an important role in the progression of human breast cancer. In this study, we investigated the effects of WY-14643, a peroxisome proliferator-activated receptor α (PPARα) agonist, on CYP1B1 expression and the related mechanism in MCF7 breast cancer cells. We performed quantitative reverse transcription-polymerase chain reaction, transient transfection, and chromatin immunoprecipitation to evaluate the effects of PPARα on peroxisome proliferator response element (PPRE)-mediated transcription. WY-14643 increased the protein and mRNA levels of CYP1B1, as well as promoter activity, in MCF-7 cells. Moreover, WY-14643 plus GW6471, a PPARα antagonist, significantly inhibited the WY-14643-mediated increase in CYP1B1 expression. PPARα knockdown by a small interfering RNA markedly suppressed the induction of CYP1B1 expression by WY-14643, suggesting that WY-14643 induces CYP1B1 expression via a PPARα-dependent mechanism. Bioinformatics analysis identified putative PPREs (−833/−813) within the promoter region of the CYP1B1 gene. Inactivation of these putative PPREs by deletion mutagenesis suppressed the WY-14643-mediated induction of CYP1B1 promoter activation. Furthermore, WY-14643 induced PPARα to assume a form capable of binding specifically to the PPRE-binding site in the CYP1B1 promoter. Our findings suggest that WY-14643 induces the expression of CYP1B1 through activation of PPARα.
Collapse
|
21
|
Thomas LW, Ashcroft M. Exploring the molecular interface between hypoxia-inducible factor signalling and mitochondria. Cell Mol Life Sci 2019; 76:1759-1777. [PMID: 30767037 PMCID: PMC6453877 DOI: 10.1007/s00018-019-03039-y] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/09/2019] [Accepted: 02/01/2019] [Indexed: 12/19/2022]
Abstract
Oxygen is required for the survival of the majority of eukaryotic organisms, as it is important for many cellular processes. Eukaryotic cells utilize oxygen for the production of biochemical energy in the form of adenosine triphosphate (ATP) generated from the catabolism of carbon-rich fuels such as glucose, lipids and glutamine. The intracellular sites of oxygen consumption-coupled ATP production are the mitochondria, double-membraned organelles that provide a dynamic and multifaceted role in cell signalling and metabolism. Highly evolutionarily conserved molecular mechanisms exist to sense and respond to changes in cellular oxygen levels. The primary transcriptional regulators of the response to decreased oxygen levels (hypoxia) are the hypoxia-inducible factors (HIFs), which play important roles in both physiological and pathophysiological contexts. In this review we explore the relationship between HIF-regulated signalling pathways and the mitochondria, including the regulation of mitochondrial metabolism, biogenesis and distribution.
Collapse
Affiliation(s)
- Luke W Thomas
- University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH, UK
| | - Margaret Ashcroft
- University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH, UK.
| |
Collapse
|
22
|
Kagawa Y, Umaru BA, Ariful I, Shil SK, Miyazaki H, Yamamoto Y, Ogata M, Owada Y. Role of FABP7 in tumor cell signaling. Adv Biol Regul 2019; 71:206-218. [PMID: 30245263 DOI: 10.1016/j.jbior.2018.09.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 09/13/2018] [Accepted: 09/13/2018] [Indexed: 06/08/2023]
Abstract
Lipids are major molecules for the function of organisms and are involved in the pathophysiology of various diseases. Fatty acids (FAs) signaling and their metabolism are some of the most important pathways in tumor development, as lipids serve as energetic sources during carcinogenesis. Fatty acid binding proteins (FABPs) facilitate FAs transport to different cell organelles, modulating their metabolism along with mediating other physiological activities. FABP7, brain-typed FABP, is thought to be an important molecule for cell proliferation in healthy as well as diseased organisms. Several studies on human tumors and tumor-derived cell lines put FABP7 in the center of tumorigenesis, and its high expression level has been reported to correlate with poor prognosis in different tumor types. Several types of FABP7-expressing tumors have shown an up-regulation of cell signaling activity, but molecular mechanisms of FABP7 involvement in tumorigenesis still remain elusive. In this review, we focus on the expression and function of FABP7 in different tumors, and possible mechanisms of FABP7 in tumor proliferation and migration.
Collapse
Affiliation(s)
- Yoshiteru Kagawa
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Banlanjo A Umaru
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Islam Ariful
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Pharmacy, University of Rajshahi, Rajshahi, Bangladesh
| | - Subrata Kumar Shil
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hirofumi Miyazaki
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yui Yamamoto
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Anatomy, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Masaki Ogata
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Anatomy, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yuji Owada
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
23
|
Hernández-Corbacho MJ, Obeid LM. A novel role for DGATs in cancer. Adv Biol Regul 2018; 72:89-101. [PMID: 30579761 DOI: 10.1016/j.jbior.2018.12.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/09/2018] [Accepted: 12/10/2018] [Indexed: 02/06/2023]
Affiliation(s)
- María José Hernández-Corbacho
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY, 11794, USA
| | - Lina M Obeid
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY, 11794, USA; The Northport VA Medical Center, Northport, NY, 11768, USA.
| |
Collapse
|
24
|
Abstract
Peroxisomes are key metabolic organelles, which contribute to cellular lipid metabolism, e.g. the β-oxidation of fatty acids and the synthesis of myelin sheath lipids, as well as cellular redox balance. Peroxisomal dysfunction has been linked to severe metabolic disorders in man, but peroxisomes are now also recognized as protective organelles with a wider significance in human health and potential impact on a large number of globally important human diseases such as neurodegeneration, obesity, cancer, and age-related disorders. Therefore, the interest in peroxisomes and their physiological functions has significantly increased in recent years. In this review, we intend to highlight recent discoveries, advancements and trends in peroxisome research, and present an update as well as a continuation of two former review articles addressing the unsolved mysteries of this astonishing organelle. We summarize novel findings on the biological functions of peroxisomes, their biogenesis, formation, membrane dynamics and division, as well as on peroxisome-organelle contacts and cooperation. Furthermore, novel peroxisomal proteins and machineries at the peroxisomal membrane are discussed. Finally, we address recent findings on the role of peroxisomes in the brain, in neurological disorders, and in the development of cancer.
Collapse
Affiliation(s)
- Markus Islinger
- Institute of Neuroanatomy, Center for Biomedicine and Medical Technology Mannheim, Medical Faculty Manheim, University of Heidelberg, 68167, Mannheim, Germany
| | - Alfred Voelkl
- Institute for Anatomy and Cell Biology, University of Heidelberg, 69120, Heidelberg, Germany
| | - H Dariush Fahimi
- Institute for Anatomy and Cell Biology, University of Heidelberg, 69120, Heidelberg, Germany
| | | |
Collapse
|
25
|
Ma L, Zhang B, Zhou C, Li Y, Li B, Yu M, Luo Y, Gao L, Zhang D, Xue Q, Qiu Q, Lin B, Zou J, Yang H. The comparison genomics analysis with glioblastoma multiforme (GBM) cells under 3D and 2D cell culture conditions. Colloids Surf B Biointerfaces 2018; 172:665-673. [PMID: 30243220 DOI: 10.1016/j.colsurfb.2018.09.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/27/2018] [Accepted: 09/14/2018] [Indexed: 12/16/2022]
Abstract
GBM, the most common and aggressive malignant primary brain tumors which needs new research approach to reveal the underline molecular mechanism of tumor progression. The 3D in vitro tumor model can be a simple and effective way to study tumor characteristics with ability to replicate of the tumor milieu. In the current study, we adopted the DNA microarray to analyze the gene expression of GBM tumor cells cultured under 2D cell culture flasks and 3D PLA porous scaffolds for 4,7 and 14 days. For 14 day old cultures, 8117 and 3060 genes expression were upregulated and downregulated respectively. Further KEGG pathway analysis revealed, the upregulated genes were mainly enriched/involved in PPAR and PI3K-Akt signaling pathways whereas the downregulated genes were mainly contributed in metabolism, ECM related and TGF-beta pathways. Thus, our approach of establishing 3D in vitro tumor model provides realistic results and proves itself a powerful tool for understanding the inner nature of GBM and can be considered as potential platform for drug screening.
Collapse
Affiliation(s)
- Liang Ma
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China.
| | - Bin Zhang
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China.
| | - Changchun Zhou
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People's Republic of China
| | - Yuting Li
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Binjie Li
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Mengfei Yu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, People's Republic of China
| | - Yichen Luo
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Lei Gao
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Duo Zhang
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge, CB2 1PZ, United Kingdom
| | - Qian Xue
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Qingchong Qiu
- Zhejiang California International NanoSystems Institute, Zhejiang University, Hangzhou, 310029, People's Republic of China
| | - Biaoyang Lin
- Zhejiang California International NanoSystems Institute, Zhejiang University, Hangzhou, 310029, People's Republic of China
| | - Jun Zou
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Huayong Yang
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China
| |
Collapse
|
26
|
Petan T, Jarc E, Jusović M. Lipid Droplets in Cancer: Guardians of Fat in a Stressful World. Molecules 2018; 23:molecules23081941. [PMID: 30081476 PMCID: PMC6222695 DOI: 10.3390/molecules23081941] [Citation(s) in RCA: 230] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 12/12/2022] Open
Abstract
Cancer cells possess remarkable abilities to adapt to adverse environmental conditions. Their survival during severe nutrient and oxidative stress depends on their capacity to acquire extracellular lipids and the plasticity of their mechanisms for intracellular lipid synthesis, mobilisation, and recycling. Lipid droplets, cytosolic fat storage organelles present in most cells from yeast to men, are emerging as major regulators of lipid metabolism, trafficking, and signalling in various cells and tissues exposed to stress. Their biogenesis is induced by nutrient and oxidative stress and they accumulate in various cancers. Lipid droplets act as switches that coordinate lipid trafficking and consumption for different purposes in the cell, such as energy production, protection against oxidative stress or membrane biogenesis during rapid cell growth. They sequester toxic lipids, such as fatty acids, cholesterol and ceramides, thereby preventing lipotoxic cell damage and engage in a complex relationship with autophagy. Here, we focus on the emerging mechanisms of stress-induced lipid droplet biogenesis; their roles during nutrient, lipotoxic, and oxidative stress; and the relationship between lipid droplets and autophagy. The recently discovered principles of lipid droplet biology can improve our understanding of the mechanisms that govern cancer cell adaptability and resilience to stress.
Collapse
Affiliation(s)
- Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana SI-1000, Slovenia.
| | - Eva Jarc
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana SI-1000, Slovenia.
- Jožef Stefan International Postgraduate School, Ljubljana SI-1000, Slovenia.
| | - Maida Jusović
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana SI-1000, Slovenia.
- Jožef Stefan International Postgraduate School, Ljubljana SI-1000, Slovenia.
| |
Collapse
|
27
|
The Involvement of PPARs in the Peculiar Energetic Metabolism of Tumor Cells. Int J Mol Sci 2018; 19:ijms19071907. [PMID: 29966227 PMCID: PMC6073339 DOI: 10.3390/ijms19071907] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/10/2018] [Accepted: 06/24/2018] [Indexed: 12/13/2022] Open
Abstract
Energy homeostasis is crucial for cell fate, since all cellular activities are strongly dependent on the balance between catabolic and anabolic pathways. In particular, the modulation of metabolic and energetic pathways in cancer cells has been discussed in some reports, but subsequently has been neglected for a long time. Meanwhile, over the past 20 years, a recovery of the study regarding cancer metabolism has led to an increasing consideration of metabolic alterations in tumors. Cancer cells must adapt their metabolism to meet their energetic and biosynthetic demands, which are associated with the rapid growth of the primary tumor and colonization of distinct metastatic sites. Cancer cells are largely dependent on aerobic glycolysis for their energy production, but are also associated with increased fatty acid synthesis and increased rates of glutamine consumption. In fact, emerging evidence has shown that therapeutic resistance to cancer treatment may arise from the deregulation of glucose metabolism, fatty acid synthesis, and glutamine consumption. Cancer cells exhibit a series of metabolic alterations induced by mutations that lead to a gain-of-function of oncogenes, and a loss-of-function of tumor suppressor genes, including increased glucose consumption, reduced mitochondrial respiration, an increase of reactive oxygen species, and cell death resistance; all of these are responsible for cancer progression. Cholesterol metabolism is also altered in cancer cells and supports uncontrolled cell growth. In this context, we discuss the roles of peroxisome proliferator-activated receptors (PPARs), which are master regulators of cellular energetic metabolism in the deregulation of the energetic homeostasis, which is observed in cancer. We highlight the different roles of PPAR isotypes and the differential control of their transcription in various cancer cells.
Collapse
|
28
|
Hypoxia-derived exosomes induce putative altered pathways in biosynthesis and ion regulatory channels in glioblastoma cells. Biochem Biophys Rep 2018; 14:104-113. [PMID: 29872742 PMCID: PMC5986551 DOI: 10.1016/j.bbrep.2018.03.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/08/2018] [Accepted: 03/29/2018] [Indexed: 12/31/2022] Open
Abstract
Hypoxia, a hallmark characteristic of glioblastoma (GBM) induces changes in the transcriptome and the proteome of tumor cells. We discovered that hypoxic stress produces significant qualitative and quantitative changes in the protein content of secreted exosomes from GBM cells. Among the proteins found to be selectively elevated in hypoxic exosomes were protein-lysine 6-oxidase (LOX), thrombospondin-1 (TSP1), vascular derived endothelial factor (VEGF) and a disintegrin and metalloproteinase with thrombospondin motifs 1 (ADAMTS1), well studied contributors to tumor progression, metastasis and angiogenesis. Our findings demonstrate that hypoxic exosomes induce differential gene expression in recipient glioma cells. Glioma cells stimulated with hypoxic exosomes showed a marked upregulation of small nucleolar RNA, C/D box 116–21 (SNORD116-21) transcript among others while significantly downregulated the potassium voltage-gated channel subfamily J member 3 (KCNJ3) message. This differential expression of certain genes is governed by the protein cargo being transferred via exosomes. Additionally, compared to normoxic exosomes, hypoxic exosomes increased various angiogenic related parameters vis-à-vis, overall tube length, branching intervals and length of isolated branches studied in tube formation assay with endothelial progenitor cells (EPCs). Thus, the intercellular communication facilitated via exosomes secreted from hypoxic GBM cells induce marked changes in the expression of genes in neighboring normoxic tumor cells and possibly in surrounding stromal cells, many of which are involved in cancer progression and treatment resistance mechanisms. In GBM, hypoxic stress induces profound changes in the protein content of secreted exosomes. Hypoxic exosomal contents induce angiogenesis and significant changes in recipient GBM cell transcriptome. Hypoxic exosomes play a major role leading to tumor proliferation, tumor growth and cell survival.
Collapse
|
29
|
Gabano E, Ravera M, Trivero F, Tinello S, Gallina A, Zanellato I, Gariboldi MB, Monti E, Osella D. The cisplatin-based Pt(iv)-diclorofibrato multi-action anticancer prodrug exhibits excellent performances also under hypoxic conditions. Dalton Trans 2018; 47:8268-8282. [DOI: 10.1039/c7dt04614f] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The cisplatin/clofibrato combos are multi-action Pt(iv) complexes active on a panel of human tumor cell lines, also under hypoxic conditions.
Collapse
Affiliation(s)
- Elisabetta Gabano
- Dipartimento di Scienze e Innovazione Tecnologica
- Università del Piemonte Orientale
- 15121 Alessandria
- Italy
| | - Mauro Ravera
- Dipartimento di Scienze e Innovazione Tecnologica
- Università del Piemonte Orientale
- 15121 Alessandria
- Italy
| | - Francesca Trivero
- Dipartimento di Scienze e Innovazione Tecnologica
- Università del Piemonte Orientale
- 15121 Alessandria
- Italy
| | - Stefano Tinello
- Dipartimento di Scienze e Innovazione Tecnologica
- Università del Piemonte Orientale
- 15121 Alessandria
- Italy
| | - Andrea Gallina
- Dipartimento di Scienze e Innovazione Tecnologica
- Università del Piemonte Orientale
- 15121 Alessandria
- Italy
| | - Ilaria Zanellato
- Dipartimento di Scienze e Innovazione Tecnologica
- Università del Piemonte Orientale
- 15121 Alessandria
- Italy
| | - Marzia B. Gariboldi
- Dipartimento di Biotecnologie e Scienze della Vita
- Università dell'Insubria
- 21052 Busto Arsizio
- Italy
| | - Elena Monti
- Dipartimento di Biotecnologie e Scienze della Vita
- Università dell'Insubria
- 21052 Busto Arsizio
- Italy
| | - Domenico Osella
- Dipartimento di Scienze e Innovazione Tecnologica
- Università del Piemonte Orientale
- 15121 Alessandria
- Italy
| |
Collapse
|
30
|
Ammazzalorso A, De Lellis L, Florio R, Bruno I, De Filippis B, Fantacuzzi M, Giampietro L, Maccallini C, Perconti S, Verginelli F, Cama A, Amoroso R. Cytotoxic effect of a family of peroxisome proliferator-activated receptor antagonists in colorectal and pancreatic cancer cell lines. Chem Biol Drug Des 2017; 90:1029-1035. [PMID: 28544586 DOI: 10.1111/cbdd.13026] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/22/2022]
Abstract
Recent studies report an interesting role of peroxisome proliferator-activated receptor (PPAR) antagonists in different tumor models, being these compounds able to perturb metabolism and viability in cancer cells. In this work, the identification of a novel PPAR antagonist, showing inhibitory activity on PPARα and a weaker antagonism on PPARγ, is described. The activity of this compound and of a series of chemical analogues was investigated in selected tumor cell lines, expressing both PPARα and PPARγ. Data obtained show a dose-dependent cytotoxic effect of the novel PPAR antagonist in colorectal and pancreatic cancer models.
Collapse
Affiliation(s)
- Alessandra Ammazzalorso
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy
| | - Laura De Lellis
- Unità di Patologia Generale, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy.,Aging Research Center (Ce.S.I.), Università "G. d'Annunzio", Chieti, Italy
| | - Rosalba Florio
- Unità di Patologia Generale, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy.,Aging Research Center (Ce.S.I.), Università "G. d'Annunzio", Chieti, Italy
| | - Isabella Bruno
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy
| | - Barbara De Filippis
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy
| | - Marialuigia Fantacuzzi
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy
| | - Letizia Giampietro
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy
| | - Cristina Maccallini
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy
| | - Silvia Perconti
- Aging Research Center (Ce.S.I.), Università "G. d'Annunzio", Chieti, Italy
| | - Fabio Verginelli
- Unità di Patologia Generale, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy.,Aging Research Center (Ce.S.I.), Università "G. d'Annunzio", Chieti, Italy
| | - Alessandro Cama
- Unità di Patologia Generale, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy.,Aging Research Center (Ce.S.I.), Università "G. d'Annunzio", Chieti, Italy
| | - Rosa Amoroso
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy
| |
Collapse
|
31
|
Fidoamore A, Cristiano L, Laezza C, Galzio R, Benedetti E, Cinque B, Antonosante A, d'Angelo M, Castelli V, Cifone MG, Ippoliti R, Giordano A, Cimini A. Energy metabolism in glioblastoma stem cells: PPARα a metabolic adaptor to intratumoral microenvironment. Oncotarget 2017; 8:108430-108450. [PMID: 29312541 PMCID: PMC5752454 DOI: 10.18632/oncotarget.19086] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 06/10/2017] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma (GB), the most-common cancer in the adult brain, despite surgery and radio/ chemotherapy, is to date almost incurable. Many hypoxic tumors, including GB, show metabolic reprogramming to sustain uncontrolled proliferation, hypoxic conditions and angiogenesis. Peroxisome Proliferator-activated Receptors (PPAR), particularly the α isotype, have been involved in the control of energetic metabolism. Herein, we characterized patient-derived GB neurospheres focusing on their energetic metabolism and PPARα expression. Moreover, we used a specific PPARα antagonist and studied its effects on the energetic metabolism and cell proliferation/survival of GB stem cells. The results obtained demonstrate that tumor neurospheres are metabolically reprogrammed up-regulating glucose transporter, glucose uptake and glycogen and lipid storage, mainly under hypoxic culture conditions. Treatment with the PPARα antagonist GW6471 resulted in decreased cell proliferation and neurospheres formation. Therefore, PPARα antagonism arises as a potent new strategy as adjuvant to gold standard therapies for GB for counteracting recurrences and opening the way for pre-clinical trials for this class of compounds. When tumor neurospheres were grown in hypoxic conditions in the presence of different glucose concentrations, the most diluted one (0.25g/L) mimicking the real concentration present in the neurosphere core, PPARα increase/PPARγ decrease, increased proliferation and cholesterol content, decreased glycogen particles and LDs were observed. All these responses were reverted by the 72 h treatment with the PPARα antagonist.
Collapse
Affiliation(s)
- Alessia Fidoamore
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Loredana Cristiano
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Chiara Laezza
- Institute of Endocrinology and Experimental Oncology, IEOS, CNR, Naples, Italy
| | - Renato Galzio
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Benedetta Cinque
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Andrea Antonosante
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Maria Grazia Cifone
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Department of Biology, Temple University, Philadelphia, Pennsylvania, USA.,Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Department of Biology, Temple University, Philadelphia, Pennsylvania, USA.,National Institute for Nuclear Physics (INFN), Gran Sasso National Laboratory (LNGS), Assergi, Italy
| |
Collapse
|
32
|
Florio R, De Lellis L, di Giacomo V, Di Marcantonio MC, Cristiano L, Basile M, Verginelli F, Verzilli D, Ammazzalorso A, Prasad SC, Cataldi A, Sanna M, Cimini A, Mariani-Costantini R, Mincione G, Cama A. Effects of PPARα inhibition in head and neck paraganglioma cells. PLoS One 2017; 12:e0178995. [PMID: 28594934 PMCID: PMC5464765 DOI: 10.1371/journal.pone.0178995] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 05/22/2017] [Indexed: 01/21/2023] Open
Abstract
Head and neck paragangliomas (HNPGLs) are rare tumors that may cause important morbidity, because of their tendency to infiltrate the skull base. At present, surgery is the only therapeutic option, but radical removal may be difficult or impossible. Thus, effective targets and molecules for HNPGL treatment need to be identified. However, the lack of cellular models for this rare tumor hampers this task. PPARα receptor activation was reported in several tumors and this receptor appears to be a promising therapeutic target in different malignancies. Considering that the role of PPARα in HNPGLs was never studied before, we analyzed the potential of modulating PPARα in a unique model of HNPGL cells. We observed an intense immunoreactivity for PPARα in HNPGL tumors, suggesting that this receptor has an important role in HNPGL. A pronounced nuclear expression of PPARα was also confirmed in HNPGL-derived cells. The specific PPARα agonist WY14643 had no effect on HNPGL cell viability, whereas the specific PPARα antagonist GW6471 reduced HNPGL cell viability and growth by inducing cell cycle arrest and caspase-dependent apoptosis. GW6471 treatment was associated with a marked decrease of CDK4, cyclin D3 and cyclin B1 protein expression, along with an increased expression of p21 in HNPGL cells. Moreover, GW6471 drastically impaired clonogenic activity of HNPGL cells, with a less marked effect on cell migration. Notably, the effects of GW6471 on HNPGL cells were associated with the inhibition of the PI3K/GSK3β/β-catenin signaling pathway. In conclusion, the PPARα antagonist GW6471 reduces HNPGL cell viability, interfering with cell cycle and inducing apoptosis. The mechanisms affecting HNPGL cell viability involve repression of the PI3K/GSK3β/β-catenin pathway. Therefore, PPARα could represent a novel therapeutic target for HNPGL.
Collapse
Affiliation(s)
- Rosalba Florio
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Unit of General Pathology, CeSI-MeT, “G. d’Annunzio” University, Chieti, Italy
| | - Laura De Lellis
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Unit of General Pathology, CeSI-MeT, “G. d’Annunzio” University, Chieti, Italy
- * E-mail: (LDL); (AC)
| | - Viviana di Giacomo
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Maria Carmela Di Marcantonio
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Loredana Cristiano
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Mariangela Basile
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Fabio Verginelli
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Unit of General Pathology, CeSI-MeT, “G. d’Annunzio” University, Chieti, Italy
| | - Delfina Verzilli
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | | | | | - Amelia Cataldi
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Mario Sanna
- Department of Otology and Skull Base Surgery, Gruppo Otologico, Piacenza, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, United States of America
- Gran Sasso National Laboratory (LNGS), National Institute for Nuclear Physics (INFN), Assergi, Italy
| | - Renato Mariani-Costantini
- Unit of General Pathology, CeSI-MeT, “G. d’Annunzio” University, Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Gabriella Mincione
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Alessandro Cama
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Unit of General Pathology, CeSI-MeT, “G. d’Annunzio” University, Chieti, Italy
- * E-mail: (LDL); (AC)
| |
Collapse
|
33
|
Raysi Dehcordi S, Ricci A, Di Vitantonio H, De Paulis D, Luzzi S, Palumbo P, Cinque B, Tempesta D, Coletti G, Cipolloni G, Cifone MG, Galzio R. Stemness Marker Detection in the Periphery of Glioblastoma and Ability of Glioblastoma to Generate Glioma Stem Cells: Clinical Correlations. World Neurosurg 2017; 105:895-905. [PMID: 28559081 DOI: 10.1016/j.wneu.2017.05.099] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/16/2017] [Accepted: 05/18/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Recent studies suggested glioma stem cells (GSCs) are key contributors to therapeutic resistance of glioblastoma multiforme (GBM) and are responsible for GBM recurrence. METHODS We characterized the phenotype of cancer cells in the core and periphery of 20 GBM tumors, correlating clinical outcome to the ability to form GSCs and distinguishing survival based on Ki-67 staining. RESULTS Similar levels of methylguanine-deoxyribonucleic acid methyltransferase were found in the core and periphery of GBM tumors, whereas Ki-67 was reduced in the periphery. Similar levels of stemness markers in the periphery and in the core of all GBM cultures were found. Only cells expressing >30% SOX2 levels were able to produce neurospheres. Immunophenotypic analysis showed higher levels of stemness markers in GSC cultures than in all GBM primary cultures. GSC in vitro production and coexpression of Ki-67 >5% negatively correlated with outcome. CONCLUSIONS Not all GBM cultures can generate GSCs, and this capacity is linked to >30% SOX2 levels. The ability to form spheres negatively correlated to survival, and the detection of >5% Ki-67 levels may be useful to identify patients at risk of disease progression. The presence of GSC-/SOX-2-/Ki-67- cells may be regarded as a new prognostic factor. The presence of stemness markers and methylguanine-deoxyribonucleic acid methyltransferase in the periphery of GBM tumors may be the reason for treatment failure and recurrence. Development of stem cell-targeted therapies and elaboration of more aggressive treatments represent an opportunity to eliminate the GBM source and the nidus of recurrence.
Collapse
Affiliation(s)
- Soheila Raysi Dehcordi
- Operative Unit of Neurosurgery, San Salvatore Hospital, L'Aquila, Italy; Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Alessandro Ricci
- Operative Unit of Neurosurgery, San Salvatore Hospital, L'Aquila, Italy
| | | | - Danilo De Paulis
- Operative Unit of Neurosurgery, San Salvatore Hospital, L'Aquila, Italy
| | - Sabino Luzzi
- Operative Unit of Neurosurgery, San Salvatore Hospital, L'Aquila, Italy
| | - Paola Palumbo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Benedetta Cinque
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Daniela Tempesta
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Gino Coletti
- Operative Unit of Pathology, San Salvatore Hospital, L'Aquila, Italy
| | | | - Maria Grazia Cifone
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Renato Galzio
- Operative Unit of Neurosurgery, San Salvatore Hospital, L'Aquila, Italy; Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
34
|
Haynes HR, White P, Hares KM, Redondo J, Kemp KC, Singleton WGB, Killick-Cole CL, Stevens JR, Garadi K, Guglani S, Wilkins A, Kurian KM. The transcription factor PPARα is overexpressed and is associated with a favourable prognosis in IDH-wildtype primary glioblastoma. Histopathology 2017; 70:1030-1043. [PMID: 27926792 DOI: 10.1111/his.13142] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 11/29/2016] [Indexed: 12/26/2022]
Abstract
AIMS PPARα agonists are in current clinical use as hypolipidaemic agents and show significant antineoplastic effects in human glioblastoma models. To date however, the expression of PPARα in large-scale glioblastoma datasets has not been examined. We aimed to investigate the expression of the transcription factor PPARα in primary glioblastoma, the relationship between PPARα expression and patients' clinicopathological features and other molecular markers associated with gliomagenesis. METHODS AND RESULTS With protein immunoblotting techniques and reverse transcription quantitative real-time PCR, PPARα was found to be significantly overexpressed in glioblastoma compared with control brain tissue (P = 0.032 and P = 0.005). PPARA gene expression was found to be enriched in the classical glioblastoma subtype within The Cancer Genome Atlas (TCGA) dataset. Although not associated with overall survival when assessed by immunohistochemistry, cross-validation with the TCGA dataset and multivariate analyses identified PPARA gene expression as an independent prognostic marker for overall survival (P = 0.042). Finally, hierarchical clustering revealed novel, significant associations between high PPARA expression and a putative set of glioblastoma molecular mediators including EMX2, AQP4, and NTRK2. CONCLUSIONS PPARα is overexpressed in primary glioblastoma and high PPARA expression functions as an independent prognostic marker in the glioblastoma TCGA dataset. Further studies are required to explore genetic associations with high PPARA expression and to analyse the predictive role of PPARα expression in glioblastoma models in response to PPARα agonists.
Collapse
Affiliation(s)
- Harry R Haynes
- Brain Tumour Research Group, Institute of Clinical Neurosciences, University of Bristol, Bristol, UK
| | - Paul White
- Applied Statistics Group, University of the West of England, Bristol, UK
| | - Kelly M Hares
- MS and Stem Cell Research Group, Institute of Clinical Neurosciences, University of Bristol, Bristol, UK
| | - Juliana Redondo
- MS and Stem Cell Research Group, Institute of Clinical Neurosciences, University of Bristol, Bristol, UK
| | - Kevin C Kemp
- MS and Stem Cell Research Group, Institute of Clinical Neurosciences, University of Bristol, Bristol, UK
| | - William G B Singleton
- Functional Neurosurgery Research Group, Institute of Clinical Neurosciences, University of Bristol, Bristol, UK
| | - Clare L Killick-Cole
- Functional Neurosurgery Research Group, Institute of Clinical Neurosciences, University of Bristol, Bristol, UK
| | | | - Krishnakumar Garadi
- Bristol Haematology and Oncology Centre, University Hospital Bristol Trust, Bristol, UK
| | - Sam Guglani
- Gloucestershire Oncology Centre, Gloucestershire Hospitals NHS Foundation Trust, Cheltenham, UK
| | - Alastair Wilkins
- MS and Stem Cell Research Group, Institute of Clinical Neurosciences, University of Bristol, Bristol, UK
| | - Kathreena M Kurian
- Brain Tumour Research Group, Institute of Clinical Neurosciences, University of Bristol, Bristol, UK
| |
Collapse
|
35
|
Benedetti E, d'Angelo M, Ammazzalorso A, Gravina GL, Laezza C, Antonosante A, Panella G, Cinque B, Cristiano L, Dhez AC, Astarita C, Galzio R, Cifone MG, Ippoliti R, Amoroso R, Di Cesare E, Giordano A, Cimini A. PPARα Antagonist AA452 Triggers Metabolic Reprogramming and Increases Sensitivity to Radiation Therapy in Human Glioblastoma Primary Cells. J Cell Physiol 2016; 232:1458-1466. [PMID: 27736000 DOI: 10.1002/jcp.25648] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/11/2016] [Indexed: 02/01/2023]
Abstract
Glioblastoma (GB) is the most common cancer in the brain and with an increasing incidence. Despite major advances in the field, there is no curative therapy for GB to date. Many solid tumors, including GB, experienced metabolic reprogramming in order to sustain uncontrolled proliferation, hypoxic conditions, and angiogenesis. PPARs, member of the steroid hormone receptor superfamily, are particularly involved in the control of energetic metabolism, particularly lipid metabolism, which has been reported deregulated in gliomas. PPARα was previously indicated by us as a potential therapeutic target for this neoplasm, due to the malignancy grade dependency of its expression, being particularly abundant in GB. In this work, we used a new PPARα antagonist on patient-derived GB primary cells, with particular focus on the effects on lipid metabolism and response to radiotherapy. The results obtained demonstrated that blocking PPARα results in cell death induction, increase of radiosensitivity, and decrease of migration. Therefore, AA452 is proposed as a new adjuvant for the gold standard therapies for GB, opening the possibility for preclinical and clinical trials for this class of compounds. J. Cell. Physiol. 232: 1458-1466, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Elisabetta Benedetti
- Depatment of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Michele d'Angelo
- Depatment of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | | | - Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Chiara Laezza
- Institute of Endocrinology and Experimental Oncology (IEOS), CNR, Naples, Italy
| | - Andrea Antonosante
- Depatment of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Gloria Panella
- Depatment of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Benedetta Cinque
- Depatment of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Loredana Cristiano
- Depatment of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Anne Chloè Dhez
- Depatment of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Carlo Astarita
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, Pennsylvania.,Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Renato Galzio
- Depatment of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Maria Grazia Cifone
- Depatment of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Rodolfo Ippoliti
- Depatment of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Rosa Amoroso
- Department of Pharmacy, G. d'Annunzio University, Chieti, Italy
| | - Ernesto Di Cesare
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, Pennsylvania.,Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Annamaria Cimini
- Depatment of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, Pennsylvania.,Gran Sasso National Laboratory (LNGS), National Institute for Nuclear Physics (INFN), Assergi, Italy
| |
Collapse
|
36
|
Koizume S, Miyagi Y. Lipid Droplets: A Key Cellular Organelle Associated with Cancer Cell Survival under Normoxia and Hypoxia. Int J Mol Sci 2016; 17:ijms17091430. [PMID: 27589734 PMCID: PMC5037709 DOI: 10.3390/ijms17091430] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/15/2016] [Accepted: 08/24/2016] [Indexed: 12/20/2022] Open
Abstract
The Warburg effect describes the phenomenon by which cancer cells obtain energy from glycolysis even under normoxic (O₂-sufficient) conditions. Tumor tissues are generally exposed to hypoxia owing to inefficient and aberrant vasculature. Cancer cells have multiple molecular mechanisms to adapt to such stress conditions by reprogramming the cellular metabolism. Hypoxia-inducible factors are major transcription factors induced in cancer cells in response to hypoxia that contribute to the metabolic changes. In addition, cancer cells within hypoxic tumor areas have reduced access to serum components such as nutrients and lipids. However, the effect of such serum factor deprivation on cancer cell biology in the context of tumor hypoxia is not fully understood. Cancer cells are lipid-rich under normoxia and hypoxia, leading to the increased generation of a cellular organelle, the lipid droplet (LD). In recent years, the LD-mediated stress response mechanisms of cancer cells have been revealed. This review focuses on the production and functions of LDs in various types of cancer cells in relation to the associated cellular environment factors including tissue oxygenation status and metabolic mechanisms. This information will contribute to the current understanding of how cancer cells adapt to diverse tumor environments to promote their survival.
Collapse
Affiliation(s)
- Shiro Koizume
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama 241-8515, Japan.
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama 241-8515, Japan.
| |
Collapse
|
37
|
Shah T, Wildes F, Kakkad S, Artemov D, Bhujwalla ZM. Lymphatic endothelial cells actively regulate prostate cancer cell invasion. NMR IN BIOMEDICINE 2016; 29:904-911. [PMID: 27149683 DOI: 10.1002/nbm.3543] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/23/2016] [Accepted: 03/29/2016] [Indexed: 06/05/2023]
Abstract
Lymphatic vessels serve as the primary route for metastatic spread to lymph nodes. However, it is not clear how interactions between cancer cells and lymphatic endothelial cells (LECs), especially within hypoxic microenvironments, affect the invasion of cancer cells. Here, using an MR compatible cell perfusion assay, we investigated the role of LEC-prostate cancer (PCa) cell interaction in the invasion and degradation of the extracellular matrix (ECM) by two human PCa cell lines, PC-3 and DU-145, under normoxia and hypoxia, and determined the metabolic changes that occurred under these conditions. We observed a significant increase in the invasion of ECM by invasive PC-3 cells, but not poorly invasive DU-145 cells when human dermal lymphatic microvascular endothelial cells (HMVEC-dlys) were present. Enhanced degradation of ECM by PC-3 cells in the presence of HMVEC-dlys identified interactions between HMVEC-dlys and PCa cells influencing cancer cell invasion. The enhanced ECM degradation was partly attributed to increased MMP-9 enzymatic activity in PC-3 cells when HMVEC-dlys were in close proximity. Significantly higher uPAR and MMP-9 expression levels observed in PC-3 cells compared to DU-145 cells may be one mechanism for increased invasion and degradation of matrigel by these cells irrespective of the presence of HMVEC-dlys. Hypoxia significantly decreased invasion by PC-3 cells, but this decrease was significantly attenuated when HMVEC-dlys were present. Significantly higher phosphocholine was observed in invasive PC-3 cells, while higher glycerophosphocholine was observed in DU-145 cells. These metabolites were not altered in the presence of HMVEC-dlys. Significantly increased lipid levels and lipid droplets were observed in PC-3 and DU-145 cells under hypoxia reflecting an adaptive survival response to oxidative stress. These results suggest that in vivo, invasive cells in or near lymphatic endothelial cells are likely to be more invasive and degrade the ECM to influence the metastatic cascade. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Tariq Shah
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Flonne Wildes
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samata Kakkad
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dmitri Artemov
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zaver M Bhujwalla
- JHU ICMIC Program, Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
38
|
Ammazzalorso A, Carrieri A, Verginelli F, Bruno I, Carbonara G, D'Angelo A, De Filippis B, Fantacuzzi M, Florio R, Fracchiolla G, Giampietro L, Giancristofaro A, Maccallini C, Cama A, Amoroso R. Synthesis, in vitro evaluation, and molecular modeling investigation of benzenesulfonimide peroxisome proliferator-activated receptors α antagonists. Eur J Med Chem 2016; 114:191-200. [PMID: 26974385 DOI: 10.1016/j.ejmech.2016.02.064] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/24/2016] [Accepted: 02/25/2016] [Indexed: 02/02/2023]
Abstract
Recent evidences suggest a moderate activation of Peroxisome Proliferator-Activated Receptors (PPARs) could be favorable in metabolic diseases, reducing side effects given from full agonists. PPAR partial agonists and antagonists represent, to date, interesting tools to better elucidate biological processes modulated by these receptors. In this work are reported new benzenesulfonimide compounds able to block PPARα, synthesized and tested by transactivation assays and gene expression analysis. Some of these compounds showed a dose-dependent antagonistic behavior on PPARα, submicromolar potency, different profiles of selectivity versus PPARγ, and a repressive effect on CPT1A expression. Dockings and molecular dynamics on properly selected benzenesulfonimide derivatives furnished fresh insights into the molecular determinant most likely responsible for PPARα antagonism.
Collapse
Affiliation(s)
- Alessandra Ammazzalorso
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy.
| | - Antonio Carrieri
- Dipartimento di Farmacia-Scienze del Farmaco, Università di Bari, Italy
| | - Fabio Verginelli
- Unità di Patologia Generale, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy; Aging Research Center (Ce.S.I.), Università "G. d'Annunzio", Chieti, Italy
| | - Isabella Bruno
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy
| | | | - Alessandra D'Angelo
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy
| | - Barbara De Filippis
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy
| | - Marialuigia Fantacuzzi
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy
| | - Rosalba Florio
- Unità di Patologia Generale, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy; Aging Research Center (Ce.S.I.), Università "G. d'Annunzio", Chieti, Italy
| | | | - Letizia Giampietro
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy
| | - Antonella Giancristofaro
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy
| | - Cristina Maccallini
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy
| | - Alessandro Cama
- Unità di Patologia Generale, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy; Aging Research Center (Ce.S.I.), Università "G. d'Annunzio", Chieti, Italy
| | - Rosa Amoroso
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy.
| |
Collapse
|
39
|
Sandt C, Nadaradjane C, Richards R, Dumas P, Sée V. Use of infrared microspectroscopy to elucidate a specific chemical signature associated with hypoxia levels found in glioblastoma. Analyst 2016; 141:870-83. [DOI: 10.1039/c5an02112j] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Detection of the chemical signature associated with hypoxia in single glioblastoma cells by synchrotron infrared microspectroscopy.
Collapse
Affiliation(s)
- Christophe Sandt
- Synchrotron SOLEIL
- L'Orme des Merisiers
- 91192 Gif sur Yvette
- France
| | - Céline Nadaradjane
- Synchrotron SOLEIL
- L'Orme des Merisiers
- 91192 Gif sur Yvette
- France
- Department of Biochemistry
| | - Rosalie Richards
- Department of Biochemistry
- Institute of Integrative Biology
- University of Liverpool
- Liverpool
- UK
| | - Paul Dumas
- Synchrotron SOLEIL
- L'Orme des Merisiers
- 91192 Gif sur Yvette
- France
| | - Violaine Sée
- Department of Biochemistry
- Institute of Integrative Biology
- University of Liverpool
- Liverpool
- UK
| |
Collapse
|
40
|
Karsy M, Guan J, Jensen R, Huang LE, Colman H. The Impact of Hypoxia and Mesenchymal Transition on Glioblastoma Pathogenesis and Cancer Stem Cells Regulation. World Neurosurg 2015; 88:222-236. [PMID: 26724617 DOI: 10.1016/j.wneu.2015.12.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/01/2015] [Accepted: 12/01/2015] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is an aggressive primary brain tumor with potential for wide dissemination and resistance to standard treatments. Although GBM represents a single histopathologic diagnosis under current World Health Organization criteria, data from multiplatform molecular profiling efforts, including The Cancer Genome Atlas, indicate that multiple subgroups with distinct markers and biology exist. It remains unclear whether treatment resistance differs based on subgroup. Recent evidence suggests that hypoxia, or absence of normal tissue oxygenation, is important in generating tumor resistance through a signaling cascade driven by hypoxia-inducible factors and vascular endothelial growth factor. Hypoxia can result in isolation of tumor cells from therapeutic agents and activation of downstream tumor protective mechanisms. In addition, there are links between hypoxia and the phenomenon of mesenchymal transition in gliomas. Mesenchymal transformation in gliomas resembles at many levels the epithelial-mesenchymal transition that has been described in other solid tumors in which epithelial cells lose their epithelial characteristics and take on a more mesenchymal phenotype, but the mesenchymal transition in brain tumors is also distinct, perhaps related to the unique cell types and cellular organization in the brain and brain tumors. Cancer stem cells, which are specific cell populations involved in self-renewal, differentiation, and GBM pathophysiology, are also importantly regulated by hypoxia signaling pathways. In this review, we discuss the interplay of hypoxia and mesenchymal signaling in GBM including the key pathway regulators and downstream genes, the effect of these processes in regulation of the tumor microenvironment and cancer stem cells, and their role in treatment resistance.
Collapse
Affiliation(s)
- Michael Karsy
- Department of Neurosurgery, Clinical Neurosciences Center, Salt Lake City, Utah, USA
| | - Jian Guan
- Department of Neurosurgery, Clinical Neurosciences Center, Salt Lake City, Utah, USA
| | - Randy Jensen
- Department of Neurosurgery, Clinical Neurosciences Center, Salt Lake City, Utah, USA; Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - L Eric Huang
- Department of Neurosurgery, Clinical Neurosciences Center, Salt Lake City, Utah, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, Utah, USA
| | - Howard Colman
- Department of Neurosurgery, Clinical Neurosciences Center, Salt Lake City, Utah, USA; Huntsman Cancer Institute, Salt Lake City, Utah, USA.
| |
Collapse
|
41
|
Lewis CA, Brault C, Peck B, Bensaad K, Griffiths B, Mitter R, Chakravarty P, East P, Dankworth B, Alibhai D, Harris AL, Schulze A. SREBP maintains lipid biosynthesis and viability of cancer cells under lipid- and oxygen-deprived conditions and defines a gene signature associated with poor survival in glioblastoma multiforme. Oncogene 2015; 34:5128-40. [PMID: 25619842 DOI: 10.1038/onc.2014.439] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 11/04/2014] [Accepted: 12/05/2014] [Indexed: 12/16/2022]
Abstract
Oxygen and nutrient limitation are common features of the tumor microenvironment and are associated with cancer progression and induction of metastasis. The inefficient vascularization of tumor tissue also limits the penetration of other serum-derived factors, such as lipids and lipoproteins, which can be rate limiting for cell proliferation and survival. Here we have investigated the effect of hypoxia and serum deprivation on sterol regulatory element-binding protein (SREBP) activity and the expression of lipid metabolism genes in human glioblastoma multiforme (GBM) cancer cells. We found that SREBP transcriptional activity was induced by serum depletion both in normoxic and hypoxic cells and that activation of SREBP was required to maintain the expression of fatty acid and cholesterol metabolism genes under hypoxic conditions. Moreover, expression of stearoyl-CoA desaturase, the enzyme required for the generation of mono-unsaturated fatty acids, and fatty acid-binding protein 7, a regulator of glioma stem cell function, was strongly dependent on SREBP function. Inhibition of SREBP function blocked lipid biosynthesis in hypoxic cancer cells and impaired cell survival under hypoxia and in a three-dimensional spheroid model. Finally, gene expression analysis revealed that SREBP defines a gene signature that is associated with poor survival in glioblastoma.
Collapse
Affiliation(s)
- C A Lewis
- Gene Expression Analysis Laboratory, Cancer Research UK London Research Institute, London, UK
| | - C Brault
- Theodor-Boveri-Institute, Biocenter, Am Hubland, Würzburg, Germany
| | - B Peck
- Gene Expression Analysis Laboratory, Cancer Research UK London Research Institute, London, UK
| | - K Bensaad
- CRUK Growth Factor Group, The Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, UK
| | - B Griffiths
- Gene Expression Analysis Laboratory, Cancer Research UK London Research Institute, London, UK
| | - R Mitter
- Bioinformatics and Biostatistics Service, Cancer Research UK London Research Institute, London, UK
| | - P Chakravarty
- Bioinformatics and Biostatistics Service, Cancer Research UK London Research Institute, London, UK
| | - P East
- Bioinformatics and Biostatistics Service, Cancer Research UK London Research Institute, London, UK
| | - B Dankworth
- Theodor-Boveri-Institute, Biocenter, Am Hubland, Würzburg, Germany
| | - D Alibhai
- Light Microscopy Core, Cancer Research UK London Research Institute, London, UK
| | - A L Harris
- CRUK Growth Factor Group, The Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, UK
| | - A Schulze
- Gene Expression Analysis Laboratory, Cancer Research UK London Research Institute, London, UK
- Theodor-Boveri-Institute, Biocenter, Am Hubland, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken Würzburg, Germany
| |
Collapse
|
42
|
Schrader M, Costello JL, Godinho LF, Azadi AS, Islinger M. Proliferation and fission of peroxisomes - An update. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:971-83. [PMID: 26409486 DOI: 10.1016/j.bbamcr.2015.09.024] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/16/2015] [Accepted: 09/21/2015] [Indexed: 12/23/2022]
Abstract
In mammals, peroxisomes perform crucial functions in cellular metabolism, signalling and viral defense which are essential to the health and viability of the organism. In order to achieve this functional versatility peroxisomes dynamically respond to molecular cues triggered by changes in the cellular environment. Such changes elicit a corresponding response in peroxisomes, which manifests itself as a change in peroxisome number, altered enzyme levels and adaptations to the peroxisomal structure. In mammals the generation of new peroxisomes is a complex process which has clear analogies to mitochondria, with both sharing the same division machinery and undergoing a similar division process. How the regulation of this division process is integrated into the cell's response to different stimuli, the signalling pathways and factors involved, remains somewhat unclear. Here, we discuss the mechanism of peroxisomal fission, the contributions of the various division factors and examine the potential impact of post-translational modifications, such as phosphorylation, on the proliferation process. We also summarize the signalling process and highlight the most recent data linking signalling pathways with peroxisome proliferation.
Collapse
Affiliation(s)
- Michael Schrader
- College of Life and Environmental Sciences, Biosciences, University of Exeter, EX4 4QJ, Exeter Devon, UK; Centre for Cell Biology, Department of Biology, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - Joseph L Costello
- College of Life and Environmental Sciences, Biosciences, University of Exeter, EX4 4QJ, Exeter Devon, UK
| | - Luis F Godinho
- Centre for Cell Biology, Department of Biology, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Afsoon S Azadi
- College of Life and Environmental Sciences, Biosciences, University of Exeter, EX4 4QJ, Exeter Devon, UK
| | - Markus Islinger
- Neuroanatomy, Center for Biomedicine and Medical Technology Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| |
Collapse
|
43
|
Ammazzalorso A, Tricca ML, Bruno I, De Filippis B, Di Matteo M, Fantacuzzi M, Giampietro L, Maccallini C, Mollica A, Amoroso R. Titanium-Promoted Acylation of Sulfonamides toN-Acylsulfonamide PPARαAntagonists. SYNTHETIC COMMUN 2015. [DOI: 10.1080/00397911.2015.1092552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
44
|
Farr RL, Lismont C, Terlecky SR, Fransen M. Peroxisome biogenesis in mammalian cells: The impact of genes and environment. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1049-60. [PMID: 26305119 DOI: 10.1016/j.bbamcr.2015.08.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 08/13/2015] [Accepted: 08/18/2015] [Indexed: 01/16/2023]
Abstract
The initiation and progression of many human diseases are mediated by a complex interplay of genetic, epigenetic, and environmental factors. As all diseases begin with an imbalance at the cellular level, it is essential to understand how various types of molecular aberrations, metabolic changes, and environmental stressors function as switching points in essential communication networks. In recent years, peroxisomes have emerged as important intracellular hubs for redox-, lipid-, inflammatory-, and nucleic acid-mediated signaling pathways. In this review, we focus on how nature and nurture modulate peroxisome biogenesis and function in mammalian cells. First, we review emerging evidence that changes in peroxisome activity can be linked to the epigenetic regulation of cell function. Next, we outline how defects in peroxisome biogenesis may directly impact cellular pathways involved in the development of disease. In addition, we discuss how changes in the cellular microenvironment can modulate peroxisome biogenesis and function. Finally, given the importance of peroxisome function in multiple aspects of health, disease, and aging, we highlight the need for more research in this still understudied field.
Collapse
Affiliation(s)
- Rebecca L Farr
- Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven-University of Leuven, Herestraat 49 box 601, B-3000 Leuven, Belgium; Department of Pharmacology, Wayne State University School of Medicine, 540 E. Canfield Ave., Detroit, MI 48201, USA
| | - Celien Lismont
- Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven-University of Leuven, Herestraat 49 box 601, B-3000 Leuven, Belgium
| | - Stanley R Terlecky
- Department of Pharmacology, Wayne State University School of Medicine, 540 E. Canfield Ave., Detroit, MI 48201, USA
| | - Marc Fransen
- Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven-University of Leuven, Herestraat 49 box 601, B-3000 Leuven, Belgium.
| |
Collapse
|
45
|
Cavazos DA, Brenner AJ. Hypoxia in astrocytic tumors and implications for therapy. Neurobiol Dis 2015; 85:227-233. [PMID: 26094595 DOI: 10.1016/j.nbd.2015.06.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 06/01/2015] [Accepted: 06/12/2015] [Indexed: 01/10/2023] Open
Abstract
Glioblastoma (GBM, Grade IV astrocytoma) is the most common and most aggressive of the primary malignant brain tumors in adults. Hypoxia is a distinct feature in GBM and plays a significant role in tumor progression, resistance to treatment and poor outcomes. This review considers the effects of hypoxia on astrocytic tumors and the mechanisms that contribute to tumor progression and therapeutic resistance, with a focus on the vascular changes, chemotaxic signaling pathways and metabolic alterations involved.
Collapse
Affiliation(s)
- David A Cavazos
- Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA
| | - Andrew J Brenner
- Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA.
| |
Collapse
|
46
|
Bensaad K, Favaro E, Lewis CA, Peck B, Lord S, Collins JM, Pinnick KE, Wigfield S, Buffa FM, Li JL, Zhang Q, Wakelam MJO, Karpe F, Schulze A, Harris AL. Fatty acid uptake and lipid storage induced by HIF-1α contribute to cell growth and survival after hypoxia-reoxygenation. Cell Rep 2014; 9:349-365. [PMID: 25263561 DOI: 10.1016/j.celrep.2014.08.056] [Citation(s) in RCA: 456] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 07/16/2014] [Accepted: 08/22/2014] [Indexed: 01/22/2023] Open
Abstract
An in vivo model of antiangiogenic therapy allowed us to identify genes upregulated by bevacizumab treatment, including Fatty Acid Binding Protein 3 (FABP3) and FABP7, both of which are involved in fatty acid uptake. In vitro, both were induced by hypoxia in a hypoxia-inducible factor-1α (HIF-1α)-dependent manner. There was a significant lipid droplet (LD) accumulation in hypoxia that was time and O2 concentration dependent. Knockdown of endogenous expression of FABP3, FABP7, or Adipophilin (an essential LD structural component) significantly impaired LD formation under hypoxia. We showed that LD accumulation is due to FABP3/7-dependent fatty acid uptake while de novo fatty acid synthesis is repressed in hypoxia. We also showed that ATP production occurs via β-oxidation or glycogen degradation in a cell-type-dependent manner in hypoxia-reoxygenation. Finally, inhibition of lipid storage reduced protection against reactive oxygen species toxicity, decreased the survival of cells subjected to hypoxia-reoxygenation in vitro, and strongly impaired tumorigenesis in vivo.
Collapse
Affiliation(s)
- Karim Bensaad
- CRUK Hypoxia and Angiogenesis Group, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK.
| | - Elena Favaro
- CRUK Hypoxia and Angiogenesis Group, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Caroline A Lewis
- Gene Expression Analysis Laboratory, Cancer Research UK, London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Barrie Peck
- Gene Expression Analysis Laboratory, Cancer Research UK, London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Simon Lord
- CRUK Hypoxia and Angiogenesis Group, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Jennifer M Collins
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Katherine E Pinnick
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Simon Wigfield
- CRUK Hypoxia and Angiogenesis Group, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Francesca M Buffa
- CRUK Hypoxia and Angiogenesis Group, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Ji-Liang Li
- CRUK Hypoxia and Angiogenesis Group, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Qifeng Zhang
- The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | | | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK; NIHR Oxford Biomedical Research Centre, OUH Trust, Churchill Hospital, Oxford OX3 7LF, UK
| | - Almut Schulze
- Gene Expression Analysis Laboratory, Cancer Research UK, London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK; Department of Biochemistry and Molecular Biology, Theodor Boveri Institute, Biocenter, Am Hubland, 97074 Würzburg, Germany
| | - Adrian L Harris
- CRUK Hypoxia and Angiogenesis Group, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| |
Collapse
|
47
|
Binello E, Mormone E, Emdad L, Kothari H, Germano IM. Characterization of fenofibrate-mediated anti-proliferative pro-apoptotic effects on high-grade gliomas and anti-invasive effects on glioma stem cells. J Neurooncol 2014; 117:225-34. [PMID: 24493576 DOI: 10.1007/s11060-014-1385-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 01/20/2014] [Indexed: 11/24/2022]
Abstract
Glioblastoma is the most common, and at the same time, most aggressive type of high-grade glioma (HGG). The prognosis of glioblastoma patients treated with standard therapy including surgery, temozolomide and radiation therapy remains poor. Peroxisome proliferator-activated receptor-α (PPARα) agonists are in widespread clinical use for the treatment of hyperlipidemia. Recent evidence has suggested a potential role in various cancers including glioblastoma. In this study, we characterized the effects of PPARα agonist, fenofibrate, directly on HGG cells and glioma stem cells (GSC). Fenofibrate exhibited dose-dependent p53-independent anti-proliferative effects on HGG starting at 25 μM and pro-apoptotic effects starting at 50 μM, suggesting that the anti-proliferative actions are present only at 25 μM. PPARα was expressed in all HGG cell lines. Inhibition of PPARα with specific inhibitor GW6471 did not affect either proliferation or apoptosis suggesting that these are PPARα-independent effects. Fenofibrate treatment of HGG cells robustly diminished the expression of key signaling pathways, including NF-κB and cyclin D1. Phosphorylation of Akt was also diminished, with no change in total Akt. Effects on apoptotic signaling molecules, Bax and Bcl-xL, had a trend towards pro-apoptotic effects. With respect to GSC, fenofibrate treatment at 25 μM significantly decreased invasion in association with a decrease in CD133 and Oct4 expression. Overall, results support consideration of fenofibrate as an anti-glioma agent and establish its potential as an adjunct treatment strategy for HGG. Translation to the clinical setting could be rapid given its current use as a clinical agent and its low toxicity profile.
Collapse
Affiliation(s)
- Emanuela Binello
- Department of Neurosurgery, Mount Sinai School of Medicine, 1 Gustave L. Levy Place, Box 1136, New York, NY, 10029, USA
| | | | | | | | | |
Collapse
|
48
|
Bensaad K, Harris AL. Hypoxia and metabolism in cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 772:1-39. [PMID: 24272352 DOI: 10.1007/978-1-4614-5915-6_1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Interest in targeting metabolism has been renewed in recent years as research increases understanding of the altered metabolic profile of tumor cells compared with that of normal cells. Metabolic reprogramming allows cancer cells to survive and proliferate in the hostile tumor microenvironment. These metabolic changes support energy generation, anabolic processes, and the maintenance of redox potential, mechanisms that are all essential for the proliferation and survival of tumor cells. The metabolic switch in a number of key metabolic pathways is mainly regulated by genetic events, rendering cancer cells addicted to certain nutrients, such as glutamine. In addition, hypoxia is induced when highly proliferative tumor cells distance themselves from an oxygen supply. Hypoxia-inducible factor 1α is largely responsible for alterations in metabolism that support the survival of hypoxic tumor cells. Metabolic alterations and dependencies of cancer cells may be exploited to improve anticancer therapy. This chapter reviews the main aspects of altered metabolism in cancer cells, emphasizing recent advances in glucose, glutamine, and lipid metabolism.
Collapse
Affiliation(s)
- Karim Bensaad
- CRUK Hypoxia and Angiogenesis Group, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK,
| | | |
Collapse
|
49
|
García-Álvarez I, Garrido L, Romero-Ramírez L, Nieto-Sampedro M, Fernández-Mayoralas A, Campos-Olivas R. The effect of antitumor glycosides on glioma cells and tissues as studied by proton HR-MAS NMR spectroscopy. PLoS One 2013; 8:e78391. [PMID: 24194925 PMCID: PMC3806797 DOI: 10.1371/journal.pone.0078391] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 09/20/2013] [Indexed: 12/21/2022] Open
Abstract
The effect of the treatment with glycolipid derivatives on the metabolic profile of intact glioma cells and tumor tissues, investigated using proton high resolution magic angle spinning (1H HR-MAS) nuclear magnetic resonance (NMR) spectroscopy, is reported here. Two compounds were used, a glycoside and its thioglycoside analogue, both showing anti-proliferative activity on glioma C6 cell cultures; however, only the thioglycoside exhibited antitumor activity in vivo. At the drug concentrations showing anti-proliferative activity in cell culture (20 and 40 µM), significant increases in choline containing metabolites were observed in the 1H NMR spectra of the same intact cells. In vivo experiments in nude mice bearing tumors derived from implanted C6 glioma cells, showed that reduction of tumor volume was associated with significant changes in the metabolic profile of the same intact tumor tissues; and were similar to those observed in cell culture. Specifically, the activity of the compounds is mainly associated with an increase in choline and phosphocholine, in both the cell cultures and tumoral tissues. Taurine, a metabolite that has been considered a biomarker of apoptosis, correlated with the reduction of tumor volume. Thus, the results indicate that the mode of action of the glycoside involves, at least in part, alteration of phospholipid metabolism, resulting in cell death.
Collapse
Affiliation(s)
- Isabel García-Álvarez
- Unidad de Neurología Experimental, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla-La Mancha (SESCAM), Toledo, Spain
- Instituto de Química Orgánica General, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- * E-mail: (IG-A); (RC-O)
| | - Leoncio Garrido
- Instituto de Ciencia y Tecnología de Polímeros, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Lorenzo Romero-Ramírez
- Unidad de Neurología Experimental, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla-La Mancha (SESCAM), Toledo, Spain
- Instituto Cajal de Neurobiología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Manuel Nieto-Sampedro
- Unidad de Neurología Experimental, Hospital Nacional de Parapléjicos, Servicio de Salud de Castilla-La Mancha (SESCAM), Toledo, Spain
- Instituto Cajal de Neurobiología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Alfonso Fernández-Mayoralas
- Instituto de Química Orgánica General, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Ramón Campos-Olivas
- Spectroscopy and NMR Unit, Structural Biology and Biocomputing Programme, Spanish National Cancer Center (CNIO), Madrid, Spain
- * E-mail: (IG-A); (RC-O)
| |
Collapse
|
50
|
Ammazzalorso A, De Filippis B, Giampietro L, Amoroso R. Blocking the peroxisome proliferator-activated receptor (PPAR): an overview. ChemMedChem 2013; 8:1609-16. [PMID: 23939910 DOI: 10.1002/cmdc.201300250] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Indexed: 12/19/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) have been studied extensively over the last few decades and have been assessed as molecular targets for the development of drugs against metabolic disorders. A rapid increase in understanding of the physiology and pharmacology of these receptors has occurred, together with the identification of novel chemical structures that are able to activate the various PPAR subtypes. More recent evidence suggests that moderate activation of these receptors could be favorable in pathological situations due to a decrease in the side effects brought about by PPAR agonists. PPAR partial agonists and antagonists are interesting tools that are currently used to better elucidate the biological processes modulated by this family of nuclear receptors. Herein we present an overview of the various molecular structures that are able to block each of the PPAR subtypes, with a focus on promising therapeutic applications.
Collapse
Affiliation(s)
- Alessandra Ammazzalorso
- Dipartimento di Farmacia, Università "G. d'Annunzio" via dei Vestini 31, 66100 Chieti (Italy)
| | | | | | | |
Collapse
|