1
|
Li X, Mao J. Research progress on the role of lipoxygenase and its inhibitors in prostate cancer. Future Oncol 2024:1-20. [PMID: 39535136 DOI: 10.1080/14796694.2024.2419356] [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: 05/23/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Prostate cancer (PCa) has become a common disease among middle-aged and elderly men. The lipoxygenase (LOX) pathway plays a crucial role in the occurrence, development, invasion and metastasis of PCa and is therefore considered a new target for the prevention and treatment of PCa. 5-LOX and 12-LOX have a promoting effect on the occurrence, development, invasion and metastasis of PCa. 15-LOX-2 has an inhibitory effect on PCa. LOX inhibitors can effectively inhibit the metabolic activity of LOX. The research aims to review the mechanism of action and inhibitors of LOX in PCa, in order to provide relevant references for the prevention and treatment of PCa.
Collapse
Affiliation(s)
- Xiaobing Li
- Chongqing Medical & Pharmaceutical College, Chongqing, 400030, China
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Jingxin Mao
- Chongqing Medical & Pharmaceutical College, Chongqing, 400030, China
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
2
|
Zhuravlev A, Gavrilyuk V, Chen X, Aksenov V, Kuhn H, Ivanov I. Structural and Functional Biology of Mammalian ALOX Isoforms with Particular Emphasis on Enzyme Dimerization and Their Allosteric Properties. Int J Mol Sci 2024; 25:12058. [PMID: 39596127 PMCID: PMC11593649 DOI: 10.3390/ijms252212058] [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: 10/02/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
The human genome involves six functional arachidonic acid (AA) lipoxygenase (ALOX) genes, and the corresponding enzymes (ALOX15, ALOX15B, ALOX12, ALOX12B, ALOXE3, ALOX5) have been implicated in cell differentiations and in the pathogenesis of inflammatory, hyperproliferative, metabolic, and neurological disorders. Humans express two different AA 15-lipoxygenating ALOX isoforms, and these enzymes are called ALOX15 (15-LOX1) and ALOX15B (15-LOX2). Chromosomal localization, sequence alignments, and comparison of the enzyme properties suggest that pig and mouse ALOX15 orthologs (leukocyte-type 12-LOX) on the one hand and rabbit and human ALOX15 orthologs on the other (reticulocyte-type 15-LOX1) belong to the same enzyme family despite their different reaction specificities with AA as a substrate. In contrast, human ALOX12 (platelet-type 12-LOX), as well as pig and mouse ALOX15 (leukocyte-type 12-LOX), belong to different enzyme families, although they exhibit a similar reaction specificity with AA as a substrate. The complex multiplicity of mammalian ALOX isoforms and the controversial enzyme nomenclatures are highly confusing and prompted us to summarize the current knowledge on the biological functions, enzymatic properties, and allosteric regulation mechanisms of mammalian ALOX15, ALOX15B, and ALOX12 orthologs that belong to three different enzyme sub-families.
Collapse
Affiliation(s)
- Alexander Zhuravlev
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Vernadskogo pr. 86, Moscow 119571, Russia; (A.Z.); (V.A.); (I.I.)
| | - Viktor Gavrilyuk
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Vernadskogo pr. 86, Moscow 119571, Russia; (A.Z.); (V.A.); (I.I.)
| | - Xin Chen
- Department of Biochemistry, Charite, University Medicine Berlin, Corporate Member of Free University Berlin, Humboldt University Berlin and Berlin Institute of Health, Charitéplatz 1, D-10117 Berlin, Germany;
| | - Vladislav Aksenov
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Vernadskogo pr. 86, Moscow 119571, Russia; (A.Z.); (V.A.); (I.I.)
| | - Hartmut Kuhn
- Department of Biochemistry, Charite, University Medicine Berlin, Corporate Member of Free University Berlin, Humboldt University Berlin and Berlin Institute of Health, Charitéplatz 1, D-10117 Berlin, Germany;
| | - Igor Ivanov
- Lomonosov Institute of Fine Chemical Technologies, MIREA—Russian Technological University, Vernadskogo pr. 86, Moscow 119571, Russia; (A.Z.); (V.A.); (I.I.)
| |
Collapse
|
3
|
Chen T, Liu J, Wang C, Wang Z, Zhou J, Lin J, Mao J, Pan T, Wang J, Xu H, He X, Wu D, Liu Z. ALOX5 contributes to glioma progression by promoting 5-HETE-mediated immunosuppressive M2 polarization and PD-L1 expression of glioma-associated microglia/macrophages. J Immunother Cancer 2024; 12:e009492. [PMID: 39142719 PMCID: PMC11332009 DOI: 10.1136/jitc-2024-009492] [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] [Accepted: 07/30/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Oxylipin metabolism plays an essential role in glioma progression and immune modulation in the tumor microenvironment. Lipid metabolic reprogramming has been linked to macrophage remodeling, while the understanding of oxylipins and their catalyzed enzymes lipoxygenases in the regulation of glioma-associated microglia/macrophages (GAMs) remains largely unexplored. METHODS To explore the pathophysiological relevance of oxylipin in human glioma, we performed Ultra-high performance liquid chromatography-MS/MS (UHPLC-MS/MS) analysis in human glioma and non-tumor brain tissues. To comprehensively investigate the role of arachidonate lipoxygenase 5 (ALOX5) in glioma, we performed in vivo bioluminescent imaging, immunofluorescence staining and flow cytometry analysis on tumors from orthotopic glioma-bearing mice. We developed an ALOX5-targeted nanobody, and tested its anti-glioma efficacy of combination therapy with α-programmed cell death protein-1 (PD-1). RESULTS In this study, we found that ALOX5 and its oxylipin 5-hydroxyeicosatetraenoic acid (5-HETE) are upregulated in glioma, accumulating programmed death-ligand 1 (PD-L1)+ M2-GAMs and orchestrating an immunosuppressive tumor microenvironment. Mechanistically, 5-HETE derived from ALOX5-overexpressing glioma cells, promotes GAMs migration, PD-L1 expression, and M2 polarization by facilitating nuclear translocation of nuclear factor erythroid 2-related factor 2. Additionally, a nanobody targeting ALOX5 is developed that markedly suppresses 5-HETE efflux from glioma cells, attenuates M2 polarization of GAMs, and consequently ameliorates glioma progression. Furthermore, the combination therapy of the ALOX5-targeted nanobody plus α-PD-1 exhibits superior anti-glioma efficacy. CONCLUSIONS Our findings reveal a pivotal role of the ALOX5/5-HETE axis in regulating GAMs and highlight the ALOX5-targeted nanobody as a potential therapeutic agent, which could potentiate immune checkpoint therapy for glioma.
Collapse
Affiliation(s)
- Tao Chen
- Department of Neurosurgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Centre, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Jiangang Liu
- Department of Neurosurgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Chenci Wang
- Department of Oncology, Funan County People's Hospital, Fuyang, Anhui, China
| | - Zhengwei Wang
- Department of Neurosurgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Jiayi Zhou
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jiani Lin
- Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Centre, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Jie Mao
- Department of Neurosurgery, Longgang Central Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Tingzheng Pan
- Department of Neurosurgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Jianwei Wang
- Department of Neurosurgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Hongchao Xu
- Department of Neurosurgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Xiaosheng He
- Department of Neurosurgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Dinglan Wu
- Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Centre, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Zhuohao Liu
- Department of Neurosurgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Centre, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, People's Republic of China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| |
Collapse
|
4
|
Thanh HD, Lee S, Nguyen TT, Huu TN, Ahn EJ, Cho SH, Kim MS, Moon KS, Jung C. Temozolomide promotes matrix metalloproteinase 9 expression through p38 MAPK and JNK pathways in glioblastoma cells. Sci Rep 2024; 14:14341. [PMID: 38906916 PMCID: PMC11192740 DOI: 10.1038/s41598-024-65398-2] [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: 12/29/2023] [Accepted: 06/19/2024] [Indexed: 06/23/2024] Open
Abstract
Glioblastoma (GBM) is a highly aggressive and deadly brain cancer. Temozolomide (TMZ) is the standard chemotherapeutic agent for GBM, but the majority of patients experience recurrence and invasion of tumor cells. We investigated whether TMZ treatment of GBM cells regulates matrix metalloproteinases (MMPs), which have the main function to promote tumor cell invasion. TMZ effectively killed GL261, U343, and U87MG cells at a concentration of 500 µM, and surviving cells upregulated MMP9 expression and its activity but not those of MMP2. TMZ also elevated levels of MMP9 mRNA and MMP9 promoter activity. Subcutaneous graft tumors survived from TMZ treatment also exhibited increased expression of MMP9 and enhanced gelatinolytic activity. TMZ-mediated MMP9 upregulation was specifically mediated through the phosphorylation of p38 and JNK. This then stimulates AP-1 activity through the upregulation of c-Fos and c-Jun. Inhibition of the p38, JNK, or both pathways counteracted the TMZ-induced upregulation of MMP9 and AP-1. This study proposes a potential adverse effect of TMZ treatment for GBM: upregulation of MMP9 expression potentially associated with increased invasion and poor prognosis. This study also provides valuable insights into the molecular mechanisms by which TMZ treatment leads to increased MMP9 expression in GBM cells.
Collapse
Affiliation(s)
- Hien Duong Thanh
- Department of Anatomy, Chonnam National University Medical School, Gwangju, 61469, Korea
| | - Sueun Lee
- Department of Anatomy, Chonnam National University Medical School, Gwangju, 61469, Korea
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju-Si, 58245, Jeollanam-Do, Korea
| | - Thuy Thi Nguyen
- Department of Anatomy, Chonnam National University Medical School, Gwangju, 61469, Korea
| | - Thang Nguyen Huu
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, 61469, Korea
| | - Eun-Jung Ahn
- Department of Neurosurgery, Chonnam National University Hwasun Hospital and Medical School, Hwasun, 58128, Jeollanam-Do, Korea
| | - Sang-Hee Cho
- Department of Hemato-Oncology, Chonnam National University Medical School, Gwangju, 61469, Korea
| | - Min Soo Kim
- Department of Statistics, College of Natural Sciences, Chonnam National University, Gwangju, 61186, Korea
| | - Kyung-Sub Moon
- Department of Neurosurgery, Chonnam National University Hwasun Hospital and Medical School, Hwasun, 58128, Jeollanam-Do, Korea
| | - Chaeyong Jung
- Department of Anatomy, Chonnam National University Medical School, Gwangju, 61469, Korea.
| |
Collapse
|
5
|
Amoah AS, Pestov NB, Korneenko TV, Prokhorenko IA, Kurakin GF, Barlev NA. Lipoxygenases at the Intersection of Infection and Carcinogenesis. Int J Mol Sci 2024; 25:3961. [PMID: 38612771 PMCID: PMC11011848 DOI: 10.3390/ijms25073961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/08/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
The persisting presence of opportunistic pathogens like Pseudomonas aeruginosa poses a significant threat to many immunocompromised cancer patients with pulmonary infections. This review highlights the complexity of interactions in the host's defensive eicosanoid signaling network and its hijacking by pathogenic bacteria to their own advantage. Human lipoxygenases (ALOXs) and their mouse counterparts are integral elements of the innate immune system, mostly operating in the pro-inflammatory mode. Taking into account the indispensable role of inflammation in carcinogenesis, lipoxygenases have counteracting roles in this process. In addition to describing the structure-function of lipoxygenases in this review, we discuss their roles in such critical processes as cancer cell signaling, metastases, death of cancer and immune cells through ferroptosis, as well as the roles of ALOXs in carcinogenesis promoted by pathogenic infections. Finally, we discuss perspectives of novel oncotherapeutic approaches to harness lipoxygenase signaling in tumors.
Collapse
Affiliation(s)
- Abdul-Saleem Amoah
- Institute of Biomedical Chemistry, Moscow 119121, Russia; (A.-S.A.); (N.A.B.)
- Laboratory of Molecular Oncology, Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Nikolay B. Pestov
- Institute of Biomedical Chemistry, Moscow 119121, Russia; (A.-S.A.); (N.A.B.)
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (T.V.K.); (I.A.P.)
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Moscow 108819, Russia
- Vavilov Institute of General Genetics, Moscow 119991, Russia
| | - Tatyana V. Korneenko
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (T.V.K.); (I.A.P.)
| | - Igor A. Prokhorenko
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (T.V.K.); (I.A.P.)
| | - Georgy F. Kurakin
- Department of Biochemistry, Pirogov Russian National Research Medical University, Moscow 117513, Russia;
| | - Nickolai A. Barlev
- Institute of Biomedical Chemistry, Moscow 119121, Russia; (A.-S.A.); (N.A.B.)
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Moscow 108819, Russia
| |
Collapse
|
6
|
Chen HC, Chang WC, Chuang JY, Chang KY, Liou JP, Hsu TI. The complex role of eicosanoids in the brain: Implications for brain tumor development and therapeutic opportunities. Biochim Biophys Acta Rev Cancer 2023; 1878:188957. [PMID: 37488051 DOI: 10.1016/j.bbcan.2023.188957] [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: 05/31/2023] [Revised: 07/17/2023] [Accepted: 07/17/2023] [Indexed: 07/26/2023]
Abstract
Eicosanoids are a family of bioactive lipids that play diverse roles in the normal physiology of the brain, including neuronal signaling, synaptic plasticity, and regulation of cerebral blood flow. In the brain, eicosanoids are primarily derived from arachidonic acid, which is released from membrane phospholipids in response to various stimuli. Prostaglandins (PGs) and leukotrienes (LTs) are the major classes of eicosanoids produced in the brain, and they act through specific receptors to modulate various physiological and pathological processes. Dysregulation of eicosanoids has been implicated in the development and progression of brain tumors, including glioblastoma (GBM), meningioma, and medulloblastoma. Eicosanoids have been shown to promote tumor cell proliferation, migration, invasion, angiogenesis, and resistance to therapy. Particularly, PGE2 promotes GBM cell survival and resistance to chemotherapy. Understanding the role of eicosanoids in brain tumors can inform the development of diagnostic and prognostic biomarkers, as well as therapeutic strategies that target eicosanoid pathways. Cyclooxygenase (COX)-2 and 5-lipoxygenase (LOX) inhibitors have been shown to reduce the growth and invasiveness of GBM cells. Moreover, eicosanoids have immunomodulatory effects that can impact the immune response to brain tumors. Understanding the role of eicosanoids in the immune response to brain tumors can inform the development of immunotherapy approaches for these tumors. Overall, the complex role of eicosanoids in the brain underscores the importance of further research to elucidate their functions in normal physiology and disease, and highlights the potential for developing novel therapeutic approaches that target eicosanoid pathways in brain tumors.
Collapse
Affiliation(s)
- Hsien-Chung Chen
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; Department of Neurosurgery, Shuang Ho Hospital, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan
| | - Wen-Chang Chang
- TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Jian-Ying Chuang
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan; International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei 110, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan
| | - Kwang-Yu Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
| | - Jing-Ping Liou
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan; School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Tsung-I Hsu
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan; International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei 110, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan.
| |
Collapse
|
7
|
Kao TJ, Lin CL, Yang WB, Li HY, Hsu TI. Dysregulated lipid metabolism in TMZ-resistant glioblastoma: pathways, proteins, metabolites and therapeutic opportunities. Lipids Health Dis 2023; 22:114. [PMID: 37537607 PMCID: PMC10398973 DOI: 10.1186/s12944-023-01881-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/26/2023] [Indexed: 08/05/2023] Open
Abstract
Glioblastoma (GBM) is a highly aggressive and lethal brain tumor with limited treatment options, such as the chemotherapeutic agent, temozolomide (TMZ). However, many GBM tumors develop resistance to TMZ, which is a major obstacle to effective therapy. Recently, dysregulated lipid metabolism has emerged as an important factor contributing to TMZ resistance in GBM. The dysregulation of lipid metabolism is a hallmark of cancer and alterations in lipid metabolism have been linked to multiple aspects of tumor biology, including proliferation, migration, and resistance to therapy. In this review, we aimed to summarize current knowledge on lipid metabolism in TMZ-resistant GBM, including key metabolites and proteins involved in lipid synthesis, uptake, and utilization, and recent advances in the application of metabolomics to study lipid metabolism in GBM. We also discussed the potential of lipid metabolism as a target for novel therapeutic interventions. Finally, we highlighted the challenges and opportunities associated with developing these interventions for clinical use, and the need for further research to fully understand the role of lipid metabolism in TMZ resistance in GBM. Our review suggests that targeting dysregulated lipid metabolism may be a promising approach to overcome TMZ resistance and improve outcomes in patients with GBM.
Collapse
Affiliation(s)
- Tzu-Jen Kao
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, 110, Taiwan
- International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, 110, Taiwan
- TMU Research Center of Neuroscience, Taipei Medical University, Taipei, 110, Taiwan
| | | | - Wen-Bin Yang
- TMU Research Center of Neuroscience, Taipei Medical University, Taipei, 110, Taiwan
| | - Hao-Yi Li
- Department of Biochemistry, Ludwig-Maximilians-University, Munich, 81377, Germany
- Gene Center, Ludwig-Maximilians-University, Munich, 81377, Germany
| | - Tsung-I Hsu
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, 110, Taiwan.
- International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, 110, Taiwan.
- TMU Research Center of Neuroscience, Taipei Medical University, Taipei, 110, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei, 110, Taiwan.
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, 110, Taiwan.
| |
Collapse
|
8
|
Zhuravlev A, Cruz A, Aksenov V, Golovanov A, Lluch JM, Kuhn H, González-Lafont À, Ivanov I. Different Structures-Similar Effect: Do Substituted 5-(4-Methoxyphenyl)-1 H-indoles and 5-(4-Methoxyphenyl)-1 H-imidazoles Represent a Common Pharmacophore for Substrate Selective Inhibition of Linoleate Oxygenase Activity of ALOX15? Molecules 2023; 28:5418. [PMID: 37513289 PMCID: PMC10383952 DOI: 10.3390/molecules28145418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/04/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Mammalian 15-lipoxygenases (ALOX15) are lipid peroxidizing enzymes that exhibit variable functionality in different cancer and inflammation models. The pathophysiological role of linoleic acid- and arachidonic acid-derived ALOX15 metabolites rendered this enzyme a target for pharmacological research. Several indole and imidazole derivatives inhibit the catalytic activity of rabbit ALOX15 in a substrate-specific manner, but the molecular basis for this allosteric inhibition remains unclear. Here, we attempt to define a common pharmacophore, which is critical for this allosteric inhibition. We found that substituted imidazoles induce weaker inhibitory effects when compared with the indole derivatives. In silico docking studies and molecular dynamics simulations using a dimeric allosteric enzyme model, in which the inhibitor occupies the substrate-binding pocket of one monomer, whereas the substrate fatty acid is bound at the catalytic center of another monomer within the ALOX15 dimer, indicated that chemical modification of the core pharmacophore alters the enzyme-inhibitor interactions, inducing a reduced inhibitory potency. In our dimeric ALOX15 model, the structural differences induced by inhibitor binding are translated to the hydrophobic dimerization cluster and affect the structures of enzyme-substrate complexes. These data are of particular importance since substrate-specific inhibition may contribute to elucidation of the putative roles of ALOX15 metabolites derived from different polyunsaturated fatty acids in mammalian pathophysiology.
Collapse
Affiliation(s)
- Alexander Zhuravlev
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, Vernadskogo pr. 86, 119571 Moscow, Russia
| | - Alejandro Cruz
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Vladislav Aksenov
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, Vernadskogo pr. 86, 119571 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklihio-Maklaja Str., 16/10c4, 117997 Moscow, Russia
| | - Alexey Golovanov
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, Vernadskogo pr. 86, 119571 Moscow, Russia
| | - José M Lluch
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Hartmut Kuhn
- Department of Biochemistry, Charite-University Medicine Berlin, Corporate Member of Free University Berlin and Humboldt University Berlin, Charitéplatz 1, D-10117 Berlin, Germany
| | - Àngels González-Lafont
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Igor Ivanov
- Lomonosov Institute of Fine Chemical Technologies, MIREA-Russian Technological University, Vernadskogo pr. 86, 119571 Moscow, Russia
| |
Collapse
|
9
|
Silva JA, Colquhoun A. Effect of Polyunsaturated Fatty Acids on Temozolomide Drug-Sensitive and Drug-Resistant Glioblastoma Cells. Biomedicines 2023; 11:biomedicines11030779. [PMID: 36979758 PMCID: PMC10045395 DOI: 10.3390/biomedicines11030779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Glioblastomas (GBMs) are notoriously difficult to treat, and the development of multiple drug resistance (MDR) is common during the course of the disease. The polyunsaturated fatty acids (PUFAs) gamma-linolenic acid (GLA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA) have been reported to improve MDR in several tumors including breast, bladder, and leukaemia. However, the effects of PUFAs on GBM cell MDR are poorly understood. The present study investigated the effects of PUFAs on cellular responses to temozolomide (TMZ) in U87MG cells and the TMZ-resistant (TMZR) cells derived from U87MG. Cells were treated with PUFAs in the absence or presence of TMZ and dose–response, viable cell counting, gene expression, Western blotting, flow cytometry, gas chromatography-mass spectrometry (GCMS), and drug efflux studies were performed. The development of TMZ resistance caused an increase in ABC transporter ABCB1 and ABCC1 expression. GLA-, EPA-, and DHA-treated cells had altered fatty acid composition and accumulated lipid droplets in the cytoplasm. The most significant reduction in cell growth was seen for the U87MG and TMZR cells in the presence of EPA. GLA and EPA caused more significant effects on ABC transporter expression than DHA. GLA and EPA in combination with TMZ caused significant reductions in rhodamine 123 efflux from U87MG cells but not from TMZR cells. Overall, these findings support the notion that PUFAs can modulate ABC transporters in GBM cells.
Collapse
|
10
|
Korbecki J, Rębacz-Maron E, Kupnicka P, Chlubek D, Baranowska-Bosiacka I. Synthesis and Significance of Arachidonic Acid, a Substrate for Cyclooxygenases, Lipoxygenases, and Cytochrome P450 Pathways in the Tumorigenesis of Glioblastoma Multiforme, Including a Pan-Cancer Comparative Analysis. Cancers (Basel) 2023; 15:cancers15030946. [PMID: 36765904 PMCID: PMC9913267 DOI: 10.3390/cancers15030946] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive gliomas. New and more effective therapeutic approaches are being sought based on studies of the various mechanisms of GBM tumorigenesis, including the synthesis and metabolism of arachidonic acid (ARA), an omega-6 polyunsaturated fatty acid (PUFA). PubMed, GEPIA, and the transcriptomics analysis carried out by Seifert et al. were used in writing this paper. In this paper, we discuss in detail the biosynthesis of this acid in GBM tumors, with a special focus on certain enzymes: fatty acid desaturase (FADS)1, FADS2, and elongation of long-chain fatty acids family member 5 (ELOVL5). We also discuss ARA metabolism, particularly its release from cell membrane phospholipids by phospholipase A2 (cPLA2, iPLA2, and sPLA2) and its processing by cyclooxygenases (COX-1 and COX-2), lipoxygenases (5-LOX, 12-LOX, 15-LOX-1, and 15-LOX-2), and cytochrome P450. Next, we discuss the significance of lipid mediators synthesized from ARA in GBM cancer processes, including prostaglandins (PGE2, PGD2, and 15-deoxy-Δ12,14-PGJ2 (15d-PGJ2)), thromboxane A2 (TxA2), oxo-eicosatetraenoic acids, leukotrienes (LTB4, LTC4, LTD4, and LTE4), lipoxins, and many others. These lipid mediators can increase the proliferation of GBM cancer cells, cause angiogenesis, inhibit the anti-tumor response of the immune system, and be responsible for resistance to treatment.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Ewa Rębacz-Maron
- Department of Ecology and Anthropology, Institute of Biology, University of Szczecin, Wąska 13, 71-415 Szczecin, Poland
| | - Patrycja Kupnicka
- 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
- Correspondence: ; Tel.: +48-914-661-515
| |
Collapse
|
11
|
Zhou Y, Fang C, Xu H, Yuan L, Liu Y, Wang X, Zhang A, Shao A, Zhou D. Ferroptosis in glioma treatment: Current situation, prospects and drug applications. Front Oncol 2022; 12:989896. [PMID: 36249003 PMCID: PMC9557197 DOI: 10.3389/fonc.2022.989896] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Ferroptosis is a regulatory form of iron-dependent cell death caused by the accumulation of lipid-based reactive oxygen species (ROS) and differs from apoptosis, pyroptosis, and necrosis. Especially in neoplastic diseases, the susceptibility of tumor cells to ferroptosis affects prognosis and is associated with complex effects. Gliomas are the most common primary intracranial tumors, accounting for disease in 81% of patients with malignant brain tumors. An increasing number of studies have revealed the particular characteristics of iron metabolism in glioma cells. Therefore, agents that target a wide range of molecules involved in ferroptosis may regulate this process and enhance glioma treatment. Here, we review the underlying mechanisms of ferroptosis and summarize the potential therapeutic options for targeting ferroptosis in glioma.
Collapse
Affiliation(s)
- Yuhang Zhou
- Health Management Center, Tongde Hospital of Zhejiang Province, Hangzhou, China
- The First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chaoyou Fang
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Houshi Xu
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ling Yuan
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yibo Liu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anke Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Anke Zhang, ; Anwen Shao, ; Danyang Zhou,
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Anke Zhang, ; Anwen Shao, ; Danyang Zhou,
| | - Danyang Zhou
- Health Management Center, Tongde Hospital of Zhejiang Province, Hangzhou, China
- *Correspondence: Anke Zhang, ; Anwen Shao, ; Danyang Zhou,
| |
Collapse
|
12
|
El Khayari A, Bouchmaa N, Taib B, Wei Z, Zeng A, El Fatimy R. Metabolic Rewiring in Glioblastoma Cancer: EGFR, IDH and Beyond. Front Oncol 2022; 12:901951. [PMID: 35912242 PMCID: PMC9329787 DOI: 10.3389/fonc.2022.901951] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/21/2022] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM), a highly invasive and incurable tumor, is the humans’ foremost, commonest, and deadliest brain cancer. As in other cancers, distinct combinations of genetic alterations (GA) in GBM induce a diversity of metabolic phenotypes resulting in enhanced malignancy and altered sensitivity to current therapies. Furthermore, GA as a hallmark of cancer, dysregulated cell metabolism in GBM has been recently linked to the acquired GA. Indeed, Numerous point mutations and copy number variations have been shown to drive glioma cells’ metabolic state, affecting tumor growth and patient outcomes. Among the most common, IDH mutations, EGFR amplification, mutation, PTEN loss, and MGMT promoter mutation have emerged as key patterns associated with upregulated glycolysis and OXPHOS glutamine addiction and altered lipid metabolism in GBM. Therefore, current Advances in cancer genetic and metabolic profiling have yielded mechanistic insights into the metabolism rewiring of GBM and provided potential avenues for improved therapeutic modalities. Accordingly, actionable metabolic dependencies are currently used to design new treatments for patients with glioblastoma. Herein, we capture the current knowledge of genetic alterations in GBM, provide a detailed understanding of the alterations in metabolic pathways, and discuss their relevance in GBM therapy.
Collapse
Affiliation(s)
- Abdellatif El Khayari
- Institute of Biological Sciences (ISSB-P), Mohammed VI Polytechnic University (UM6P), Ben-Guerir, Morocco
| | - Najat Bouchmaa
- Institute of Biological Sciences (ISSB-P), Mohammed VI Polytechnic University (UM6P), Ben-Guerir, Morocco
| | - Bouchra Taib
- Institute of Sport Professions (IMS), Ibn Tofail University, Avenida de l’Université, Kenitra, Morocco
- Research Unit on Metabolism, Physiology and Nutrition, Department of Biology, Faculty of Science, Ibn Tofail University, Kenitra, Morocco
| | - Zhiyun Wei
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ailiang Zeng
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rachid El Fatimy
- Institute of Biological Sciences (ISSB-P), Mohammed VI Polytechnic University (UM6P), Ben-Guerir, Morocco
- *Correspondence: Rachid El Fatimy,
| |
Collapse
|
13
|
Rao W, Yin K. Bone Marrow Mesenchymal Stem Cells (BMSC)-Derived MicroRNA-189 Inhibits Glioma Tumorigenesis Through Suppressing Tumor Necrosis Factor- α (TNF- α)-Mediated Nuclear Factor Kappa Light Chain Enhancer of Activated B Cells (NF- κB) Signaling Pathway. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study aims at investigating the mechanism underlying bone marrow mesenchymal stem cells (BMSC) function in glioma. Glioma cells were administered with plasmids loading NF-κB siRNA, microRNA (miRNA)-189 inhibitor, or miR-189 mimics for transfection followed by analysis
of miR-189 expression by RT-qPCR, cell apoptosis by flow cytometry, cell proliferation by MTT assay,invasion and migration by Transwell assay, inflammatory factors secretion by ELISA as well as proteins expression by western blot. A mouse model of glioma was established to detect the in
vivo effect of BMSCs. miR-189 was lowly expressed in glioma cell lines but enriched in BMSCs. When miR-189 was silenced, cell proliferation, invasion and migration were potentiated and apoptosis was decreased, along with enhancement of N-cadherin, Vimentin, MMP-2 and and MMP-9, and decline
in Bax, cleaved casepase-3 and cleaved PARP. Silencing of NF-κB reversed the effect of miR-189 inhibitor on cell progression, accompanied with reduction of inflammatory factors. BMSCs treatment effectively promoted miR-189 expression in glioma and inactivated TNF-α/NF-κB
signaling, thereby suppressing tumor growth. In conclusion, miR-189 derived from BMSC inhibits glioma progression through regulation of TNF-α/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Wenxu Rao
- Department of Neurosurgery, Fuyang District Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang 310000, China
| | - Kang Yin
- Department of Neurosurgery, Fuyang District Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
14
|
Oxidized linoleic acid metabolites maintain mechanical and thermal hypersensitivity during sub-chronic inflammatory pain. Biochem Pharmacol 2022; 198:114953. [PMID: 35149052 DOI: 10.1016/j.bcp.2022.114953] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/04/2022] [Accepted: 02/04/2022] [Indexed: 12/13/2022]
Abstract
Inflammatory pain serves as a protective defense mechanism which becomes pathological when it turns into chronic inflammatory pain. This transition is mediated by a variety of peripheral mediators that sensitize nociceptors and increase pain perception in sensory neurons. Besides cytokines, chemokines and growth factors, accumulating evidence shows that oxidized lipids, such as eicosanoids and oxidized linoleic acid metabolites, contribute to this sensitization process. Most notably, the oxidized linoleic acid metabolite and partial TRPV1 agonist 9-HODE (hydroxyoctadecadienoic acid) was shown to be involved in this sensitization process. However, it is still unknown how some of the oxidized linoleic acid metabolites are synthesized in the inflammatory environment and in which phase of inflammation they become relevant. Here we show that the concentrations of oxidized linoleic acid metabolites, especially 9-HODE and 13-HODE, are significantly increased in inflamed paw tissue and the corresponding dorsal root ganglia in the sub-chronic phase of inflammation. Surprisingly, classical inflammatory lipid markers, such as prostaglandins were at basal levels in this phase of inflammation. Moreover, we revealed the cell type specific synthesis pathways of oxidized linoleic acid metabolites in primary macrophages, primary neutrophils and dorsal root ganglia. Finally, we show that blocking the most elevated metabolites 9-HODE and 13-HODE at the site of inflammation in the sub-chronic phase of inflammation, leads to a significant relief of mechanical and thermal hypersensitivity in vivo. In summary, these data offer an approach to specifically target oxidized linoleic acid metabolites in the transition of acute inflammatory pain to chronic inflammatory pain.
Collapse
|
15
|
Harnessing oxidative stress for anti-glioma therapy. Neurochem Int 2022; 154:105281. [PMID: 35038460 DOI: 10.1016/j.neuint.2022.105281] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 12/22/2021] [Accepted: 01/10/2022] [Indexed: 02/06/2023]
Abstract
Glioma cells use intermediate levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) for growth and invasion, and suppressing these reactive molecules thus may compromise processes that are vital for glioma survival. Increased oxidative stress has been identified in glioma cells, in particular in glioma stem-like cells. Studies have shown that these cells harbor potent antioxidant defenses, although endogenous protection against nitrosative stress remains understudied. The enhancement of oxidative or nitrosative stress offers a potential target for triggering glioma cell death, but whether oxidative and nitrosative stresses can be combined for therapeutic effects requires further research. The optimal approach of harnessing oxidative stress for anti-glioma therapy should include the induction of free radical-induced oxidative damage and the suppression of antioxidant defense mechanisms selectively in glioma cells. However, selective induction of oxidative/nitrosative stress in glioma cells remains a therapeutic challenge, and research into selective drug delivery systems is ongoing. Because of multifactorial mechanisms of glioma growth, progression, and invasion, prospective oncological therapies may include not only therapeutic oxidative/nitrosative stress but also inhibition of oncogenic kinases, antioxidant molecules, and programmed cell death mediators.
Collapse
|
16
|
Salemizadeh Parizi M, Salemizadeh Parizi F, Abdolhosseini S, Vanaei S, Manzouri A, Ebrahimzadeh F. Myeloid-derived suppressor cells (MDSCs) in brain cancer: challenges and therapeutic strategies. Inflammopharmacology 2021; 29:1613-1624. [PMID: 34613567 DOI: 10.1007/s10787-021-00878-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022]
Abstract
The most fatal malignancy of the central nervous system (CNS) is glioblastoma. Brain cancer is a 'cold' tumor because of fewer immunoregulatory cells and more immunosuppressive cells. Due to the cold nature of brain cancers, conventional treatments which are used to manage glioma patients show little effectiveness. Glioma patients even showed resistance to immune checkpoint blockade (ICB) and no significant efficacy. It has been shown that myeloid-derived suppressor cells (MDSCs) account for approximately 30-50% of the tumor mass in glioma. This study aimed to review MDSC function in brain cancer, as well as possible treatments and related challenges. In brain cancer and glioma, several differences in the context of MDSCs have been reported, including disagreements about the MDSC subtype that has the most inhibitory function in the brain, or inhibitory function of regulatory B cells (Bregs). There are also serious challenges in treating glioma patients. In addition to the cold nature of glioma, there are reports of an increase in MDSCs following conventional chemotherapy treatments. As a result, targeting MDSCs in combination with other therapies, such as ICB, is essential, and recent studies with the combination therapy approach have shown promising therapeutic effects in brain cancer.
Collapse
Affiliation(s)
| | | | | | - Shohreh Vanaei
- Department of Biomedical Engineering, Northeastern University, Boston, MA, USA
| | - Ali Manzouri
- School of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Farnoosh Ebrahimzadeh
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
17
|
Cyclooxygenase Inhibition Alters Proliferative, Migratory, and Invasive Properties of Human Glioblastoma Cells In Vitro. Int J Mol Sci 2021; 22:ijms22094297. [PMID: 33919029 PMCID: PMC8122446 DOI: 10.3390/ijms22094297] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/02/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023] Open
Abstract
Prostaglandin E2 (PGE2) is known to increase glioblastoma (GBM) cell proliferation and migration while cyclooxygenase (COX) inhibition decreases proliferation and migration. The present study investigated the effects of COX inhibitors and PGE2 receptor antagonists on GBM cell biology. Cells were grown with inhibitors and dose response, viable cell counting, flow cytometry, cell migration, gene expression, Western blotting, and gelatin zymography studies were performed. The stimulatory effects of PGE2 and the inhibitory effects of ibuprofen (IBP) were confirmed in GBM cells. The EP2 and EP4 receptors were identified as important mediators of the actions of PGE2 in GBM cells. The concomitant inhibition of EP2 and EP4 caused a significant decrease in cell migration which was not reverted by exogenous PGE2. In T98G cells exogenous PGE2 increased latent MMP2 gelatinolytic activity. The inhibition of COX1 or COX2 caused significant alterations in MMP2 expression and gelatinolytic activity in GBM cells. These findings provide further evidence for the importance of PGE2 signalling through the EP2 and the EP4 receptor in the control of GBM cell biology. They also support the hypothesis that a relationship exists between COX1 and MMP2 in GBM cells which merits further investigation as a novel therapeutic target for drug development.
Collapse
|