1
|
Weng X, Gonzalez M, Angelia J, Piroozmand S, Jamehdor S, Behrooz AB, Latifi-Navid H, Ahmadi M, Pecic S. Lipidomics-driven drug discovery and delivery strategies in glioblastoma. Biochim Biophys Acta Mol Basis Dis 2024; 1871:167637. [PMID: 39722408 DOI: 10.1016/j.bbadis.2024.167637] [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: 09/28/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
With few viable treatment options, glioblastoma (GBM) is still one of the most aggressive and deadly types of brain cancer. Recent developments in lipidomics have demonstrated the potential of lipid metabolism as a therapeutic target in GBM. The thorough examination of lipids in biological systems, or lipidomics, is essential to comprehending the changed lipid profiles found in GBM, which are linked to the tumor's ability to grow, survive, and resist treatment. The use of lipidomics in drug delivery and discovery is examined in this study, focusing on how it may be used to find new biomarkers, create multi-target directed ligands, and improve drug delivery systems. We also cover the use of FDA-approved medications, clinical trials that use lipid-targeted medicines, and the integration of lipidomics with other omics technologies. This study emphasizes lipidomics as a possible tool in developing more effective treatment methods for GBM by exploring various lipid-centric techniques.
Collapse
Affiliation(s)
- Xiaohui Weng
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Michael Gonzalez
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Jeannes Angelia
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States
| | - Somayeh Piroozmand
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Saleh Jamehdor
- Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Sciences, University of Manitoba, Max Rady College of Medicine, Winnipeg, Manitoba, Canada
| | - Hamid Latifi-Navid
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran; School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.; Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Iran
| | - Mazaher Ahmadi
- Department of Analytical Chemistry, Faculty of Chemistry and Petroleum Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University Fullerton, Fullerton, CA 92831, United States.
| |
Collapse
|
2
|
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
|
3
|
Repici A, Ardizzone A, Filippone A, Colarossi C, Mare M, Raciti G, Mannino D, Cuzzocrea S, Paterniti I, Esposito E. Interleukin-21 Influences Glioblastoma Course: Biological Mechanisms and Therapeutic Potential. Cells 2023; 12:2284. [PMID: 37759505 PMCID: PMC10526836 DOI: 10.3390/cells12182284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Brain tumors represent a heterogeneous group of neoplasms involving the brain or nearby tissues, affecting populations of all ages with a high incidence worldwide. Among the primary brain tumors, the most aggressive and also the most common is glioblastoma (GB), a type of glioma that falls into the category of IV-grade astrocytoma. GB often leads to death within a few months after diagnosis, even if the patient is treated with available therapies; for this reason, it is important to continue to discover new therapeutic approaches to allow for a better survival rate of these patients. Immunotherapy, today, seems to be one of the most innovative types of treatment, based on the ability of the immune system to counteract various pathologies, including cancer. In this context, interleukin 21 (IL-21), a type I cytokine produced by natural killer (NK) cells and CD4+ T lymphocytes, appears to be a valid target for new therapies since this cytokine is involved in the activation of innate and adaptive immunity. To match this purpose, our review deeply evaluated how IL-21 could influence the progression of GB, analyzing its main biological processes and mechanisms while evaluating the potential use of the latest available therapies.
Collapse
Affiliation(s)
- Alberto Repici
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.R.); (A.A.); (A.F.); (D.M.); (S.C.); (E.E.)
| | - Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.R.); (A.A.); (A.F.); (D.M.); (S.C.); (E.E.)
| | - Alessia Filippone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.R.); (A.A.); (A.F.); (D.M.); (S.C.); (E.E.)
| | - Cristina Colarossi
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy; (C.C.); (M.M.)
| | - Marzia Mare
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy; (C.C.); (M.M.)
| | - Gabriele Raciti
- IOM Ricerca, Via Penninazzo 11, 95029 Viagrande, Italy;
- Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy
| | - Deborah Mannino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.R.); (A.A.); (A.F.); (D.M.); (S.C.); (E.E.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.R.); (A.A.); (A.F.); (D.M.); (S.C.); (E.E.)
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.R.); (A.A.); (A.F.); (D.M.); (S.C.); (E.E.)
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.R.); (A.A.); (A.F.); (D.M.); (S.C.); (E.E.)
| |
Collapse
|
4
|
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
|
5
|
Sluter MN, Li Q, Yasmen N, Chen Y, Li L, Hou R, Yu Y, Yang CY, Meibohm B, Jiang J. The inducible prostaglandin E synthase (mPGES-1) in neuroinflammatory disorders. Exp Biol Med (Maywood) 2023; 248:811-819. [PMID: 37515545 PMCID: PMC10468642 DOI: 10.1177/15353702231179926] [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] [Indexed: 07/31/2023] Open
Abstract
The cyclooxygenase (COX)/prostaglandin E2 (PGE2) signaling pathway has emerged as a critical target for anti-inflammatory therapeutic development in neurological diseases. However, medical use of COX inhibitors in the treatment of various neurological disorders has been limited due to well-documented cardiovascular and cerebrovascular complications. It has been widely proposed that modulation of downstream microsomal prostaglandin E synthase-1 (mPGES-1) enzyme may provide more specificity for inhibiting PGE2-elicited neuroinflammation. Heightened levels of mPGES-1 have been detected in a variety of brain diseases such as epilepsy, stroke, glioma, and neurodegenerative diseases. Subsequently, elevated levels of PGE2, the enzymatic product of mPGES-1, have been demonstrated to modulate a multitude of deleterious effects. In epilepsy, PGE2 participates in retrograde signaling to augment glutamate release at the synapse leading to neuronal death. The excitotoxic demise of neurons incites the activation of microglia, which can become overactive upon further stimulation by PGE2. A selective mPGES-1 inhibitor was able to reduce gliosis and the expression of proinflammatory cytokines in the hippocampus following status epilepticus. A similar mechanism has also been observed in stroke, where the overactivation of microglia by PGE2 upregulated the expression and secretion of proinflammatory cytokines. This intense activation of neuroinflammatory processes triggered the secondary injury commonly observed in stroke, and blockade of mPGES-1 reduced infarction size and edema, suppressed induction of proinflammatory cytokines, and improved post-stroke well-being and cognition. Furthermore, elevated levels of PGE2 have been shown to intensify the proliferation of glioma cells, mediate P-glycoprotein expression at the blood-brain barrier (BBB) and facilitate breakdown of the BBB. For these reasons, targeting mPGES-1, the central and inducible enzyme of the COX cascade, may provide a more specific therapeutic strategy for treating neuroinflammatory diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Ruida Hou
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ying Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Chao-Yie Yang
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jianxiong Jiang
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
6
|
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
|
7
|
Uram Ł, Markowicz J, Misiorek M, Filipowicz-Rachwał A, Wołowiec S, Wałajtys-Rode E. Celecoxib substituted biotinylated poly(amidoamine) G3 dendrimer as potential treatment for temozolomide resistant glioma therapy and anti-nematode agent. Eur J Pharm Sci 2020; 152:105439. [PMID: 32615261 DOI: 10.1016/j.ejps.2020.105439] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/24/2020] [Accepted: 06/28/2020] [Indexed: 02/01/2023]
Abstract
Glioblastoma multiforme (GBM) is a one of the most widely diagnosed and difficult to treat type of central nervous system tumors. Resection combined with radiotherapy and temozolomide (TMZ) chemotherapy prolongs patients' survival only for 12 - 15 months after diagnosis. Moreover, many patients develop TMZ resistance, thus important is search for a new therapy regimes including targeted drug delivery. Most types of GBM reveal increased expression of cyclooxygenase-2 (COX-2) and production of prostaglandin E2 (PGE2), that are considered as valuable therapeutic target. In these studies, the anti-tumor properties of the selective COX-2 inhibitor celecoxib (CXB) and biotinylated third generation of the poly(amidoamine) dendrimer substituted with 31 CXB residues (G3BC31) on TMZ -resistant U-118 MG glioma cell line were examined and compared with the effect of TMZ alone including viability, proliferation, migration and apoptosis, as well as the cellular expression of COX-2, ATP level, and PGE2 production. Confocal microscopy analysis with the fluorescently labeled G3BC31 analogue has shown that the compound was effectively accumulated in U-118 MG cells in time-dependent manner and its localization was confirmed in lysosomes but not nuclei. G3BC31 reveal much higher cytotoxicity for U-118 MG cells at relatively low concentrations in the range of 2-4 µM with compared to CBX alone, active at 50-100 µM. This was due to induction of apoptosis and inhibition of proliferation and migration. Observed effects were concomitant with reduction of PGE2 production but independent of COX-2 expression. We suggest that investigated conjugate may be a promising candidate for therapy of TMZ-resistant glioblastoma multiforme, although applicable in local treatment, since our previous study of G3BC31 did not demonstrate selectivity against glioma cells compared to normal human fibroblasts. However, it has to be pointed that in our in vivo studies conducted with model organism, Caenorhabditis elegans indicated high anti-nematode activity of G3BC31 in comparison with CXB alone that confirms of usefulness of that organism for estimation of anti-cancer drug toxicity.
Collapse
Affiliation(s)
- Łukasz Uram
- Faculty of Chemistry, Rzeszow University of Technology, 6 Powstancow Warszawy Ave, 35-959 Rzeszow, Poland.
| | - Joanna Markowicz
- Faculty of Chemistry, Rzeszow University of Technology, 6 Powstancow Warszawy Ave, 35-959 Rzeszow, Poland
| | - Maria Misiorek
- Faculty of Chemistry, Rzeszow University of Technology, 6 Powstancow Warszawy Ave, 35-959 Rzeszow, Poland
| | - Aleksandra Filipowicz-Rachwał
- Faculty of Medical Sciences, Rzeszow University of Information Technology and Management, 2 Sucharskiego Str, 35-225 Rzeszow, Poland
| | - Stanisław Wołowiec
- Centre for Innovative Research in Medical and Natural Sciences, Faculty of Medicine, University of Rzeszow, Warzywna 1a, 35-310 Rzeszow, Poland
| | - Elżbieta Wałajtys-Rode
- Department of Drug Technology and Biotechnology, Faculty of Chemistry, Warsaw University of Technology,75 Koszykowa Str, 00-664 Warsaw, Poland
| |
Collapse
|
8
|
Montecillo-Aguado M, Tirado-Rodriguez B, Tong Z, Vega OM, Morales-Martínez M, Abkenari S, Pedraza-Chaverri J, Huerta-Yepez S. Importance of the Role of ω-3 and ω-6 Polyunsaturated Fatty Acids in the Progression of Brain Cancer. Brain Sci 2020; 10:E381. [PMID: 32560280 PMCID: PMC7349634 DOI: 10.3390/brainsci10060381] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/28/2020] [Accepted: 06/09/2020] [Indexed: 12/21/2022] Open
Abstract
Brain cancer is one of the most malignant types of cancer in both children and adults. Brain cancer patients tend to have a poor prognosis and a high rate of mortality. Additionally, 20-40% of all other types of cancer can develop brain metastasis. Numerous pieces of evidence suggest that omega-3-polyunsaturated fatty acids (ω-PUFAs) could potentially be used in the prevention and therapy of several types of cancer. PUFAs and oxylipins are fundamental in preserving physiological events in the nervous system; it is, therefore, necessary to maintain a certain ratio of ω-3 to ω-6 for normal nervous system function. Alterations in PUFAs signaling are involved in the development of various pathologies of the nervous system, including cancer. It is well established that an omega-6-polyunsaturated fatty acid (ω-6 PUFA)-rich diet has a pro-tumoral effect, whereas the consumption of an ω-3 rich diet has an anti-tumoral effect. This review aims to offer a better understanding of brain cancer and PUFAs and to discuss the role and impact of PUFAs on the development of different types of brain cancer. Considering the difficulty of antitumor drugs in crossing the blood-brain barrier, the therapeutic role of ω-3/ω-6 PUFAs against brain cancer would be a good alternative to consider. We highlight our current understanding of the role of PUFAs and its metabolites (oxylipins) in different brain tumors, proliferation, apoptosis, invasion, angiogenesis, and immunosuppression by focusing on recent research in vitro and in vivo.
Collapse
Affiliation(s)
- Mayra Montecillo-Aguado
- Programa de Doctorado en Ciencias Biomédicas, Facultad de Medicina, Universidad Nacional Autónoma de Mexico (UNAM), Mexico City 04510, Mexico;
- Hospital Infantil de Mexico, Federico Gomez, Unidad de Investigacion en Enfermedades Oncologicas, Mexico City 06720, Mexico; (B.T.-R.); (M.M.-M.)
| | - Belen Tirado-Rodriguez
- Hospital Infantil de Mexico, Federico Gomez, Unidad de Investigacion en Enfermedades Oncologicas, Mexico City 06720, Mexico; (B.T.-R.); (M.M.-M.)
| | - Zhen Tong
- Molecular Toxicology Interdepartmental Program and Environmental Health Sciences, University of California, Los Angeles, CA 90095, USA;
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA 90095, USA; (O.M.V.); (S.A.)
| | - Owen M. Vega
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA 90095, USA; (O.M.V.); (S.A.)
| | - Mario Morales-Martínez
- Hospital Infantil de Mexico, Federico Gomez, Unidad de Investigacion en Enfermedades Oncologicas, Mexico City 06720, Mexico; (B.T.-R.); (M.M.-M.)
| | - Shaheen Abkenari
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA 90095, USA; (O.M.V.); (S.A.)
| | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City 04510, Mexico;
| | - Sara Huerta-Yepez
- Hospital Infantil de Mexico, Federico Gomez, Unidad de Investigacion en Enfermedades Oncologicas, Mexico City 06720, Mexico; (B.T.-R.); (M.M.-M.)
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles, CA 90095, USA; (O.M.V.); (S.A.)
| |
Collapse
|
9
|
Abstract
Prostaglandin E2 (PGE2) has been thought to be an important mediator of inflammation in peripheral tissues, but recent studies clearly show the involvement of PGE2 in inflammatory brain diseases. In some animal models of brain disease, the genetic disruption and chemical inhibition of cyclooxygenase (COX)-2 resulted in the reduction of PGE2 and amelioration of symptoms, and it had been thought that PGE2 produced by COX-2 may be involved in the progression of injuries. However, COX-2 produces not only PGE2, but also some other prostanoids, and thus the protective effects of COX-2 inhibition, as well as severe side effects, may be caused by the inhibition of prostanoids other than PGE2. Therefore, to elucidate the role of PGE2, studies of microsomal prostaglandin E synthase-1 (mPGES-1), an inducible terminal enzyme for PGE2 synthesis, have recently been an active area of research. Studies from mPGES-1 deficient mice provide compelling evidence for its role in a variety of inflammatory brain diseases, such as ischemic stroke, Alzheimer's disease and epilepsy, and clues for developing new therapeutic treatments for brain diseases by targeting mPGES-1. Considering that COX inhibitors may non-selectively suppress the production of many types of prostanoids that are essential for normal physiological functioning of the brain and peripheral tissues, as well as induce gastro-intestinal, renal and cardiovascular complications, mPGES-1 inhibitors are expected to be injury-selective and have fewer side-effects when treating human brain diseases. Thus, this paper focuses on recent studies that have demonstrated the involvement of mPGES-1 in pathological brain diseases.
Collapse
Affiliation(s)
- Yuri Ikeda-Matsuo
- Laboratory of Pharmacology, Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Hokuriku University
| |
Collapse
|
10
|
Mucignat-Caretta C, Denaro L, D'Avella D, Caretta A. Protein Kinase A Distribution Differentiates Human Glioblastoma from Brain Tissue. Cancers (Basel) 2017; 10:cancers10010002. [PMID: 29267253 PMCID: PMC5789352 DOI: 10.3390/cancers10010002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/13/2017] [Accepted: 12/13/2017] [Indexed: 12/17/2022] Open
Abstract
Brain tumor glioblastoma has no clear molecular signature and there is no effective therapy. In rodents, the intracellular distribution of the cyclic AMP (cAMP)-dependent protein kinase (Protein kinase A, PKA) R2Alpha subunit was previously shown to differentiate tumor cells from healthy brain cells. Now, we aim to validate this observation in human tumors. The distribution of regulatory (R1 and R2) and catalytic subunits of PKA was examined via immunohistochemistry and Western blot in primary cell cultures and biopsies from 11 glioblastoma patients. Data were compared with information obtained from 17 other different tumor samples. The R1 subunit was clearly detectable only in some samples. The catalytic subunit was variably distributed in the different tumors. Similar to rodent tumors, all human glioblastoma specimens showed perinuclear R2 distribution in the Golgi area, while it was undetectable outside the tumor. To test the effect of targeting PKA as a therapeutic strategy, the intracellular cyclic AMP concentration was modulated with different agents in four human glioblastoma cell lines. A significant increase in cell death was detected after increasing cAMP levels or modulating PKA activity. These data raise the possibility of targeting the PKA intracellular pathway for the development of diagnostic and/or therapeutic tools for human glioblastoma.
Collapse
Affiliation(s)
- Carla Mucignat-Caretta
- Department of Molecular Medicine, University of Padova, Padova 35131, Italy.
- Biostructures and Biosystems National Institute, Rome 00136, Italy.
| | - Luca Denaro
- Department of Neuroscience, University of Padova, Padova 35131, Italy.
| | - Domenico D'Avella
- Department of Neuroscience, University of Padova, Padova 35131, Italy.
| | - Antonio Caretta
- Biostructures and Biosystems National Institute, Rome 00136, Italy.
- Department of Food and Drug, University of Parma, Parma 43121, Italy.
| |
Collapse
|
11
|
Colquhoun A. Cell biology-metabolic crosstalk in glioma. Int J Biochem Cell Biol 2017; 89:171-181. [PMID: 28549626 DOI: 10.1016/j.biocel.2017.05.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 12/19/2022]
Abstract
The renewed interest in cancer metabolism in recent years has been fuelled by the identification of the involvement of key oncogenes and tumour suppressor genes in the control of metabolic pathways. Many of these alterations lead to dramatic changes in bioenergetics, biosynthesis and redox balance within tumour cells. The complex relationship between tumour cell metabolism and the tumour microenvironment has turned this field of biochemistry and cell biology into a challenging and exciting area for study. In the case of gliomas the involvement of altered metabolic pathways including glycolysis, oxidative phosphorylation and glutaminolysis are pointing the way to new possibilities for treatment. The tumour-promoting effects of inflammation are an emerging hallmark of cancer and the role of the eicosanoids in gliomas is an area of active research to elucidate the importance of individual eicosanoids in glioma cell proliferation, migration and immune escape. In this review, the different aspects of metabolic reprogramming which occur in gliomas are highlighted and their relationship to glioma cell biology and the wider tumour microenvironment is described.
Collapse
Affiliation(s)
- Alison Colquhoun
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
12
|
Jiang J, Qiu J, Li Q, Shi Z. Prostaglandin E2 Signaling: Alternative Target for Glioblastoma? Trends Cancer 2017; 3:75-78. [PMID: 28718447 PMCID: PMC5518646 DOI: 10.1016/j.trecan.2016.12.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 12/04/2016] [Accepted: 12/06/2016] [Indexed: 11/21/2022]
Abstract
Elevated cyclooxygenase-2 (COX-2) and the associated inflammation within the brain contribute to glioblastoma development. However, medical use of COX inhibitors in glioblastoma treatment has been limited due to their well-documented vascular toxicity and inconsistent outcomes from recent human studies. Prostaglandin E2 (PGE2) has emerged as a principal mediator for COX-2 cascade-driven gliomagenesis. Are PGE2 terminal synthases and receptors feasible therapeutic targets for glioblastoma?
Collapse
Affiliation(s)
- Jianxiong Jiang
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, OH 45267-0514, USA.
| | - Jiange Qiu
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, OH 45267-0514, USA; Department of Cell Biology and Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong 510632, China
| | - Qianqian Li
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, OH 45267-0514, USA
| | - Zhi Shi
- Department of Cell Biology and Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong 510632, China.
| |
Collapse
|
13
|
Chen Y, Liu H, Xu S, Wang T, Li W. Targeting microsomal prostaglandin E2synthase-1 (mPGES-1): the development of inhibitors as an alternative to non-steroidal anti-inflammatory drugs (NSAIDs). MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00278h] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
AA cascade and several key residues in the 3D structure of mPGES-1.
Collapse
Affiliation(s)
- Yuqing Chen
- Department of Medicinal Chemistry, School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| | | | - Shuang Xu
- Department of Medicinal Chemistry, School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| | - Tianlin Wang
- Department of Medicinal Chemistry, School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| |
Collapse
|
14
|
Feng H, Li Y, Yin Y, Zhang W, Hou Y, Zhang L, Li Z, Xie B, Gao WQ, Sarkaria JN, Raizer JJ, James CD, Parsa AT, Hu B, Cheng SY. Protein kinase A-dependent phosphorylation of Dock180 at serine residue 1250 is important for glioma growth and invasion stimulated by platelet derived-growth factor receptor α. Neuro Oncol 2014; 17:832-42. [PMID: 25468898 DOI: 10.1093/neuonc/nou323] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Accepted: 10/30/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Dedicator of cytokinesis 1 (Dock1 or Dock180), a bipartite guanine nucleotide exchange factor for Rac1, plays critical roles in receptor tyrosine kinase-stimulated cancer growth and invasion. Dock180 activity is required in cell migration cancer tumorigenesis promoted by platelet derived growth factor receptor (PDGFR) and epidermal growth factor receptor. METHODS To demonstrate whether PDGFRα promotes tumor malignant behavior through protein kinase A (PKA)-dependent serine phosphorylation of Dock180, we performed cell proliferation, viability, migration, immunoprecipitation, immunoblotting, colony formation, and in vivo tumorigenesis assays using established and short-term explant cultures of glioblastoma cell lines. RESULTS Stimulation of PDGFRα results in phosphorylation of Dock180 at serine residue 1250 (S1250), whereas PKA inhibitors H-89 and KT5720 oppose this phosphorylation. S1250 locates within the Rac1-binding Dock homology region 2 domain of Dock180, and its phosphorylation activates Rac1, p-Akt, and phosphorylated extracellular signal-regulated kinase 1/2, while promoting cell migration, in vitro. By expressing RNA interference (RNAi)-resistant wild-type Dock180, but not mutant Dock180 S1250L, we were able to rescue PDGFRα-associated signaling and biological activities in cultured glioblastoma multiforme (GBM) cells that had been treated with RNAi for suppression of endogenous Dock180. In addition, expression of the same RNAi-resistant Dock180 rescued an invasive phenotype of GBM cells following intracranial engraftment in immunocompromised mice. CONCLUSION These data describe an important mechanism by which PDGFRα promotes glioma malignant phenotypes through PKA-dependent serine phosphorylation of Dock180, and the data thereby support targeting the PDGFRα-PKA-Dock180-Rac1 axis for treating GBM with molecular profiles indicating PDGFRα signaling dependency.
Collapse
Affiliation(s)
- Haizhong Feng
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (H.F., W.Z., Y.H., L.Z., Z.L., W.-Q.G., S.-Y.C.); Department of Neurology, Northwestern Brain Tumor Institute, Center for Genetic Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (H.F., J.J.R., B.H., S.-Y.C.); Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.L.); Department of Neurological Surgery (Y.Y., B.X.); Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.H.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Department of Neurological Surgery, Northwestern Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (C.D.J., A.T.P.)
| | - Yanxin Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (H.F., W.Z., Y.H., L.Z., Z.L., W.-Q.G., S.-Y.C.); Department of Neurology, Northwestern Brain Tumor Institute, Center for Genetic Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (H.F., J.J.R., B.H., S.-Y.C.); Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.L.); Department of Neurological Surgery (Y.Y., B.X.); Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.H.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Department of Neurological Surgery, Northwestern Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (C.D.J., A.T.P.)
| | - Yuhua Yin
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (H.F., W.Z., Y.H., L.Z., Z.L., W.-Q.G., S.-Y.C.); Department of Neurology, Northwestern Brain Tumor Institute, Center for Genetic Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (H.F., J.J.R., B.H., S.-Y.C.); Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.L.); Department of Neurological Surgery (Y.Y., B.X.); Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.H.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Department of Neurological Surgery, Northwestern Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (C.D.J., A.T.P.)
| | - Weiwei Zhang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (H.F., W.Z., Y.H., L.Z., Z.L., W.-Q.G., S.-Y.C.); Department of Neurology, Northwestern Brain Tumor Institute, Center for Genetic Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (H.F., J.J.R., B.H., S.-Y.C.); Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.L.); Department of Neurological Surgery (Y.Y., B.X.); Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.H.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Department of Neurological Surgery, Northwestern Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (C.D.J., A.T.P.)
| | - Yanli Hou
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (H.F., W.Z., Y.H., L.Z., Z.L., W.-Q.G., S.-Y.C.); Department of Neurology, Northwestern Brain Tumor Institute, Center for Genetic Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (H.F., J.J.R., B.H., S.-Y.C.); Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.L.); Department of Neurological Surgery (Y.Y., B.X.); Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.H.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Department of Neurological Surgery, Northwestern Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (C.D.J., A.T.P.)
| | - Lei Zhang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (H.F., W.Z., Y.H., L.Z., Z.L., W.-Q.G., S.-Y.C.); Department of Neurology, Northwestern Brain Tumor Institute, Center for Genetic Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (H.F., J.J.R., B.H., S.-Y.C.); Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.L.); Department of Neurological Surgery (Y.Y., B.X.); Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.H.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Department of Neurological Surgery, Northwestern Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (C.D.J., A.T.P.)
| | - Zuoqing Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (H.F., W.Z., Y.H., L.Z., Z.L., W.-Q.G., S.-Y.C.); Department of Neurology, Northwestern Brain Tumor Institute, Center for Genetic Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (H.F., J.J.R., B.H., S.-Y.C.); Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.L.); Department of Neurological Surgery (Y.Y., B.X.); Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.H.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Department of Neurological Surgery, Northwestern Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (C.D.J., A.T.P.)
| | - Baoshu Xie
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (H.F., W.Z., Y.H., L.Z., Z.L., W.-Q.G., S.-Y.C.); Department of Neurology, Northwestern Brain Tumor Institute, Center for Genetic Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (H.F., J.J.R., B.H., S.-Y.C.); Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.L.); Department of Neurological Surgery (Y.Y., B.X.); Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.H.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Department of Neurological Surgery, Northwestern Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (C.D.J., A.T.P.)
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (H.F., W.Z., Y.H., L.Z., Z.L., W.-Q.G., S.-Y.C.); Department of Neurology, Northwestern Brain Tumor Institute, Center for Genetic Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (H.F., J.J.R., B.H., S.-Y.C.); Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.L.); Department of Neurological Surgery (Y.Y., B.X.); Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.H.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Department of Neurological Surgery, Northwestern Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (C.D.J., A.T.P.)
| | - Jann N Sarkaria
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (H.F., W.Z., Y.H., L.Z., Z.L., W.-Q.G., S.-Y.C.); Department of Neurology, Northwestern Brain Tumor Institute, Center for Genetic Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (H.F., J.J.R., B.H., S.-Y.C.); Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.L.); Department of Neurological Surgery (Y.Y., B.X.); Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.H.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Department of Neurological Surgery, Northwestern Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (C.D.J., A.T.P.)
| | - Jeffery J Raizer
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (H.F., W.Z., Y.H., L.Z., Z.L., W.-Q.G., S.-Y.C.); Department of Neurology, Northwestern Brain Tumor Institute, Center for Genetic Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (H.F., J.J.R., B.H., S.-Y.C.); Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.L.); Department of Neurological Surgery (Y.Y., B.X.); Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.H.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Department of Neurological Surgery, Northwestern Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (C.D.J., A.T.P.)
| | - C David James
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (H.F., W.Z., Y.H., L.Z., Z.L., W.-Q.G., S.-Y.C.); Department of Neurology, Northwestern Brain Tumor Institute, Center for Genetic Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (H.F., J.J.R., B.H., S.-Y.C.); Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.L.); Department of Neurological Surgery (Y.Y., B.X.); Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.H.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Department of Neurological Surgery, Northwestern Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (C.D.J., A.T.P.)
| | - Andrew T Parsa
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (H.F., W.Z., Y.H., L.Z., Z.L., W.-Q.G., S.-Y.C.); Department of Neurology, Northwestern Brain Tumor Institute, Center for Genetic Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (H.F., J.J.R., B.H., S.-Y.C.); Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.L.); Department of Neurological Surgery (Y.Y., B.X.); Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.H.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Department of Neurological Surgery, Northwestern Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (C.D.J., A.T.P.)
| | - Bo Hu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (H.F., W.Z., Y.H., L.Z., Z.L., W.-Q.G., S.-Y.C.); Department of Neurology, Northwestern Brain Tumor Institute, Center for Genetic Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (H.F., J.J.R., B.H., S.-Y.C.); Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.L.); Department of Neurological Surgery (Y.Y., B.X.); Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.H.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Department of Neurological Surgery, Northwestern Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (C.D.J., A.T.P.)
| | - Shi-Yuan Cheng
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (H.F., W.Z., Y.H., L.Z., Z.L., W.-Q.G., S.-Y.C.); Department of Neurology, Northwestern Brain Tumor Institute, Center for Genetic Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (H.F., J.J.R., B.H., S.-Y.C.); Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.L.); Department of Neurological Surgery (Y.Y., B.X.); Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (Y.H.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.N.S.); Department of Neurological Surgery, Northwestern Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois (C.D.J., A.T.P.)
| |
Collapse
|
15
|
Evans J, Ko Y, Mata W, Saquib M, Eldridge J, Cohen-Gadol A, Leaver HA, Wang S, Rizzo MT. Arachidonic acid induces brain endothelial cell apoptosis via p38-MAPK and intracellular calcium signaling. Microvasc Res 2014; 98:145-58. [PMID: 24802256 DOI: 10.1016/j.mvr.2014.04.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 04/23/2014] [Accepted: 04/27/2014] [Indexed: 01/19/2023]
Abstract
Arachidonic acid (AA), a bioactive fatty acid whose levels increase during neuroinflammation, contributes to cerebral vascular damage and dysfunction. However, the mode of injury and underlying signaling mechanisms remain unknown. Challenge of primary human brain endothelial cells (HBECs) with AA activated a stress response resulting in caspase-3 activation, poly(ADP-ribose) polymerase cleavage, and disruption of monolayer integrity. AA also induced loss of mitochondrial membrane potential and cytochrome c release consistent with activation of intrinsic apoptosis. HBEC stimulation with AA resulted in sustained p38-MAPK activation and subsequent phosphorylation of mitogen-activated protein kinase activated protein-2 (MAPKAP-2) kinase and heat shock protein-27 (Hsp27). Conversely, other unsaturated and saturated fatty acids had no effect. Pharmacological and RNA interference-mediated p38α or p38β suppression abrogated AA signaling to caspase-3 and Hsp27, suggesting involvement of both p38 isoforms in AA-induced HBEC apoptosis. Hsp27 silencing also blocked caspase-3 activation. AA stimulated intracellular calcium release, which was attenuated by inositol 1,4,5-trisphosphate (IP3) receptor antagonists. Blockade of intracellular calcium release decreased caspase-3 activation, but had no effect on AA-induced p38-MAPK activation. However, inhibition of p38-MAPK or blockade of intracellular calcium mobilization abrogated AA-induced cytochrome c release. AA-induced caspase-3 activation was abrogated by pharmacological inhibition of lipooxygenases. These findings support a previously unrecognized signaling cooperation between p38-MAPK/MAPKAP-2/Hsp27 and intracellular calcium release in AA-induced HBEC apoptosis and suggest its relevance to neurological disorders associated with vascular inflammation.
Collapse
Affiliation(s)
- Justin Evans
- Signal Transduction Laboratory, Methodist Research Institute, Indiana University Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - YooSeung Ko
- Signal Transduction Laboratory, Methodist Research Institute, Indiana University Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wilmer Mata
- Signal Transduction Laboratory, Methodist Research Institute, Indiana University Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Muhammad Saquib
- Signal Transduction Laboratory, Methodist Research Institute, Indiana University Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joel Eldridge
- Signal Transduction Laboratory, Methodist Research Institute, Indiana University Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Aaron Cohen-Gadol
- Goodman Campbell Brain and Spine, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H Anne Leaver
- Division of Clinical Neuroscience, Edinburgh University, Edinburgh, UK
| | - Shukun Wang
- Signal Transduction Laboratory, Methodist Research Institute, Indiana University Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maria Teresa Rizzo
- Signal Transduction Laboratory, Methodist Research Institute, Indiana University Health, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pharmacology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
16
|
Gomez I, Benyahia C, Louedec L, Leséche G, Jacob MP, Longrois D, Norel X. Decreased PGE₂ content reduces MMP-1 activity and consequently increases collagen density in human varicose vein. PLoS One 2014; 9:e88021. [PMID: 24505358 PMCID: PMC3914898 DOI: 10.1371/journal.pone.0088021] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 01/02/2014] [Indexed: 11/19/2022] Open
Abstract
UNLABELLED Varicose veins are elongated and dilated saphenous veins. Despite the high prevalence of this disease, its pathogenesis remains unclear. AIMS In this study, we investigated the control of matrix metalloproteinases (MMPs) expression by prostaglandin (PG)E₂ during the vascular wall remodeling of human varicose veins. METHODS AND RESULTS Varicose (small (SDv) and large diameter (LDv)) and healthy saphenous veins (SV) were obtained after surgery. Microsomal and cytosolic PGE-synthases (mPGES and cPGES) protein and mRNA responsible for PGE₂ metabolism were analyzed in all veins. cPGES protein was absent while its mRNA was weakly expressed. mPGES-2 expression was similar in the different saphenous veins. mPGES-1 mRNA and protein were detected in healthy veins and a significant decrease was found in LDv. Additionally, 15-hydroxyprostaglandin dehydrogenase (15-PGDH), responsible for PGE₂ degradation, was over-expressed in varicose veins. These variations in mPGES-1 and 15-PGDH density account for the decreased PGE₂ level observed in varicose veins. Furthermore, a significant decrease in PGE₂ receptor (EP4) levels was also found in SDv and LDv. Active MMP-1 and total MMP-2 concentrations were significantly decreased in varicose veins while the tissue inhibitors of metalloproteinases (TIMP -1 and -2), were significantly increased, probably explaining the increased collagen content found in LDv. Finally, the MMP/TIMP ratio is restored by exogenous PGE₂ in varicose veins and reduced in presence of an EP4 receptor antagonist in healthy veins. CONCLUSIONS In conclusion, PGE₂ could be responsible for the vascular wall thickening in human varicose veins. This mechanism could be protective, strengthening the vascular wall in order to counteract venous stasis.
Collapse
Affiliation(s)
- Ingrid Gomez
- INSERM, U698, Paris, France
- University Paris Nord, UMR-S698, Paris, France
| | - Chabha Benyahia
- INSERM, U698, Paris, France
- University Paris Nord, UMR-S698, Paris, France
| | | | - Guy Leséche
- INSERM, U698, Paris, France
- AP-HP CHU X. Bichat, Department of Vascular and Thoracic Surgery, University Paris Diderot, Sorbonne Paris-Cité, UMR-S698, Paris, France
| | | | - Dan Longrois
- INSERM, U698, Paris, France
- AP-HP CHU X. Bichat, Department of Anesthesia and Intensive Care, University Paris Diderot, Sorbonne Paris-Cité, UMR-S698, Paris, France
| | - Xavier Norel
- INSERM, U698, Paris, France
- University Paris Nord, UMR-S698, Paris, France
- * E-mail:
| |
Collapse
|
17
|
Gomez I, Foudi N, Longrois D, Norel X. The role of prostaglandin E2 in human vascular inflammation. Prostaglandins Leukot Essent Fatty Acids 2013; 89:55-63. [PMID: 23756023 DOI: 10.1016/j.plefa.2013.04.004] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 04/04/2013] [Accepted: 04/05/2013] [Indexed: 10/26/2022]
Abstract
Prostaglandins (PG) are the product of a cascade of enzymes such as cyclooxygenases and PG synthases. Among PG, PGE2 is produced by 3 isoforms of PGE synthase (PGES) and through activation of its cognate receptors (EP1-4), this PG is involved in the pathophysiology of vascular diseases. Some anti-inflammatory drugs (e.g. glucocorticoids, nonsteroidal anti-inflammatory drugs) interfere with its metabolism or effects. Vascular cells can initiate many of the responses associated with inflammation. In human vascular tissue, PGE2 is involved in many physiological processes, such as increasing vascular permeability, cell proliferation, cell migration and control of vascular smooth muscle tone. PGE2 has been shown to contribute to the pathogenesis of atherosclerosis, abdominal aortic aneurysm but also in physiologic/adaptive processes such as angiogenesis. Understanding the roles of PGE2 and its cognate receptors in vascular diseases could help to identify diagnostic and prognostic biomarkers. In addition, from these recent studies new promising therapeutic approaches like mPGES-1 inhibition and/or EP4-antagonism should be investigated.
Collapse
Affiliation(s)
- I Gomez
- INSERM, U698, Paris F-75018, France; University Paris Nord, UMR-S698, Paris F-75018, France
| | | | | | | |
Collapse
|
18
|
Feng H, Hu B, Vuori K, Sarkaria JN, Furnari FB, Cavenee WK, Cheng SY. EGFRvIII stimulates glioma growth and invasion through PKA-dependent serine phosphorylation of Dock180. Oncogene 2013; 33:2504-12. [PMID: 23728337 DOI: 10.1038/onc.2013.198] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 03/06/2013] [Accepted: 04/12/2013] [Indexed: 12/17/2022]
Abstract
Glioblastomas (GBMs), the most common and malignant brain tumors, are highly resistant to current therapies. The failure of targeted therapies against aberrantly activated oncogenic signaling, such as that of the EGFR-PI3K/Akt pathway, underscores the urgent need to understand alternative downstream pathways and to identify new molecular targets for the development of more effective treatments for gliomas. Here, we report that EGFRvIII (ΔEGFR/de2-7EGFR), a constitutively active EGFR mutant that is frequently co-overexpressed with EGFR in clinical GBM tumors, promotes glioma growth and invasion through protein kinase A (PKA)-dependent phosphorylation of Dock180, a bipartite guanine nucleotide exchange factor (GEF) for Rac1. We demonstrate that EGFRvIII induces serine phosphorylation of Dock180, stimulates Rac1 activation and glioma cell migration. Treatments of glioma cells using the PKA inhibitors H-89 and KT5720, overexpression of a PKA inhibitor (PKI), and in vitro PKA kinase assays show that EGFRvIII induction of serine phosphorylation of Dock180 is PKA-dependent. Significantly, PKA induces phosphorylation of Dock180 at amino acid residue S1250 that resides within its Rac1-activating DHR-2 domain. Expression of the Dock180(S1250L) mutant, but not wild type Dock180(WT), protein in EGFRvIII-expressing glioma cells inhibited receptor-stimulated cell proliferation, survival, migration in vitro and glioma tumor growth and invasion in vivo. Together, our findings describe a novel mechanism by which EGFRvIII drives glioma tumorigenesis and invasion through PKA-dependent phosphorylation of Dock180, thereby suggesting that targeting EGFRvIII-PKA-Dock180-Rac1 signaling axis could provide a novel pathway to develop potential therapeutic strategies for malignant gliomas.
Collapse
Affiliation(s)
- H Feng
- 1] Department of Neurology, Northwestern Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA [2] Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA [3] The Robert H Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA [4] Stem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - B Hu
- 1] Department of Neurology, Northwestern Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA [2] Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA [3] The Robert H Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - K Vuori
- Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - J N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - F B Furnari
- Ludwig Institute for Cancer Research, University of California-San Diego, School of Medicine, La Jolla, CA, USA
| | - W K Cavenee
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - S-Y Cheng
- 1] Department of Neurology, Northwestern Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA [2] Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA [3] The Robert H Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
19
|
Zhou X, Li D, Resnick MB, Wands J, Cao W. NADPH oxidase NOX5-S and nuclear factor κB1 mediate acid-induced microsomal prostaglandin E synthase-1 expression in Barrett's esophageal adenocarcinoma cells. Mol Pharmacol 2013; 83:978-90. [PMID: 23439561 DOI: 10.1124/mol.112.083287] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The mechanisms of progression from Barrett's esophagus (BE) to esophageal adenocarcinoma (EA) are not known. Cycloxygenase-2 (COX-2)-derived prostaglandin E₂ (PGE₂) has been shown to be important in esophageal tumorigenesis. We have shown that COX-2 mediates acid-induced PGE₂ production. The prostaglandin E synthase (PGES) responsible for acid-induced PGE2 production in BE, however, is not known. We found that microsomal PGES1 (mPGES1), mPGES2, and cytosolic PGES (cPGES) were present in FLO EA cells. Pulsed acid treatment significantly increased mPGES1 mRNA and protein levels but had little or no effect on mPGES2 or cPGES mRNA. Knockdown of mPGES1 by mPGES1 small interfering RNA (siRNA) blocked acid-induced increase in PGE2 production and thymidine incorporation. Knockdown of NADPH oxidase, NOX5-S, a variant lacking calcium-binding domains, by NOX5 siRNA significantly inhibited acid-induced increase in mPGES1 expression, thymidine incorporation, and PGE2 production. Overexpression of NOX5-S significantly increased the luciferase activity in FLO cells transfected with a nuclear factor κB (NF-κB) in vivo activation reporter plasmid pNF-κB-Luc. Knockdown of NF-κB1 p50 by p50 siRNA significantly decreased acid-induced increase in mPGES1 expression, thymidine incorporation, and PGE₂ production. Two novel NF-κB binding elements, GGAGTCTCCC and CGGGACACCC, were identified in the mPGES1 gene promoter. We conclude that mPGES1 mediates acid-induced increase in PGE₂ production and cell proliferation. Acid-induced mPGES1 expression depends on activation of NOX5-S and NF-κB1 p50. Microsomal PGES1 may be a potential target to prevent or treat EA.
Collapse
Affiliation(s)
- Xiaoxu Zhou
- Department of Medicine, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | | | | | | | | |
Collapse
|
20
|
Gomes RN, Colquhoun A. E series prostaglandins alter the proliferative, apoptotic and migratory properties of T98G human glioma cells in vitro. Lipids Health Dis 2012; 11:171. [PMID: 23231886 PMCID: PMC3547780 DOI: 10.1186/1476-511x-11-171] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 12/04/2012] [Indexed: 12/02/2022] Open
Abstract
Background In many types of cancer, prostaglandin E2 (PGE2) is associated with tumour related processes including proliferation, migration, angiogenesis and apoptosis. However in gliomas the role of this prostanoid is poorly understood. Here, we report on the proliferative, migratory, and apoptotic effects of PGE1, PGE2 and Ibuprofen (IBP) observed in the T98G human glioma cell line in vitro. Methods T98G human glioma cells were treated with IBP, PGE1 or PGE2 at varying concentrations for 24–72 hours. Cell proliferation, mitotic index and apoptotic index were determined for each treatment. Caspase-9 and caspase-3 activity was measured using fluorescent probes in live cells (FITC-LEHD-FMK and FITC-DEVD-FMK respectively). The migratory capacity of the cells was quantified using a scratch migration assay and a transwell migration assay. Results A significant decrease was seen in cell number (54%) in the presence of 50 μM IBP. Mitotic index and bromodeoxyuridine (BrdU) incorporation were also decreased 57% and 65%, respectively, by IBP. The apoptotic index was increased (167%) and the in situ activity of caspase-9 and caspase-3 was evident in IBP treated cells. The inhibition of COX activity by IBP also caused a significant inhibition of cell migration in the monolayer scratch assay (74%) and the transwell migration assay (36%). In contrast, the presence of exogenous PGE1 or PGE2 caused significant increases in cell number (37% PGE1 and 45% PGE2). When mitotic index was measured no change was found for either PG treatment. However, the BrdU incorporation rate was significantly increased by PGE1 (62%) and to a greater extent by PGE2 (100%). The apoptotic index was unchanged by exogenous PGs. The addition of exogenous PGs caused an increase in cell migration in the monolayer scratch assay (43% PGE1 and 44% PGE2) and the transwell migration assay (28% PGE1 and 68% PGE2). Conclusions The present study demonstrated that treatments which alter PGE1 and PGE2 metabolism influence the proliferative and apoptotic indices of T98G glioma cells. The migratory capacity of the cells was also significantly affected by the change in prostaglandin metabolism. Modifying PG metabolism remains an interesting target for future studies in gliomas.
Collapse
Affiliation(s)
- Renata N Gomes
- Department of Cell and Developmental Biology, University of São Paulo, São Paulo, SP, Brazil
| | | |
Collapse
|
21
|
Venza I, Visalli M, Fortunato C, Ruggeri M, Ratone S, Caffo M, Caruso G, Alafaci C, Tomasello F, Teti D, Venza M. PGE2 induces interleukin-8 derepression in human astrocytoma through coordinated DNA demethylation and histone hyperacetylation. Epigenetics 2012; 7:1315-30. [PMID: 23051921 PMCID: PMC3499332 DOI: 10.4161/epi.22446] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We have recently reported that in astrocytoma cells the expression of interleukin-8 (IL-8) is upregulated by prostaglandin E2 (PGE2). Unfortunately, the exact mechanism by which this happens has not been clarified yet. Here, we have investigated whether IL-8 activation by PGE2 involves changes in DNA methylation and/or histone modifications in 46 astrocytoma specimens, two astrocytoma cell lines and normal astrocytic cells. The DNA methylation status of the IL-8 promoter was analyzed by bisulphite sequencing and by methylation DNA immunoprecipitation analysis. The involvement of DNA methyltransferases (DNMTs) and histone deacetylases (HDACs), as well as histone acetylation levels, was assayed by chromatin immunoprecipitation. IL-8 expression at promoter, mRNA and protein level was explored by transfection, real-time PCR and enzyme immunoassay experiments in cells untreated or treated with PGE2, 5-aza-2'-deoxycytidine (5-aza-dC) and HDAC inhibitors, alone or in combination. EMSA was performed with crude cell extracts or recombinant protein. We observed that PGE2 induced IL-8 activation through: (1) demethylation of the single CpG site 5 located at position -83 within the binding region for CEBP-β in the IL-8 promoter; (2) C/EBP-β and p300 co-activator recruitment; (3) H3 acetylation enhancement and (4) reduction in DNMT1, DNMT3a, HDAC2 and HDAC3 association to CpG site 5 region. Treatment with 5-aza-dC or HDAC inhibitors of class I HDACs strengthened either basal or PGE2-mediated IL-8 expression. These findings have elucidated an orchestrated mechanism triggered by PGE2 whereby concurrent association of site-specific demethylation and histone H3 hyperacetylation resulted in derepression of IL-8 gene expression in human astrocytoma.
Collapse
Affiliation(s)
- Isabella Venza
- Department of Surgical Specialities, University of Messina, Messina, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Davidson J, Rotondo D, Rizzo MT, Leaver HA. Therapeutic implications of disorders of cell death signalling: membranes, micro-environment, and eicosanoid and docosanoid metabolism. Br J Pharmacol 2012; 166:1193-210. [PMID: 22364602 DOI: 10.1111/j.1476-5381.2012.01900.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Disruptions of cell death signalling occur in pathological processes, such as cancer and degenerative disease. Increased knowledge of cell death signalling has opened new areas of therapeutic research, and identifying key mediators of cell death has become increasingly important. Early triggering events in cell death may provide potential therapeutic targets, whereas agents affecting later signals may be more palliative in nature. A group of primary mediators are derivatives of the highly unsaturated fatty acids (HUFAs), particularly oxygenated metabolites such as prostaglandins. HUFAs, esterified in cell membranes, act as critical signalling molecules in many pathological processes. Currently, agents affecting HUFA metabolism are widely prescribed in diseases involving disordered cell death signalling. However, partly due to rapid metabolism, their role in cell death signalling pathways is poorly characterized. Recently, HUFA-derived mediators, the resolvins/protectins and endocannabinoids, have added opportunities to target selective signals and pathways. This review will focus on the control of cell death by HUFA, eicosanoid (C20 fatty acid metabolites) and docosanoid (C22 metabolites), HUFA-derived lipid mediators, signalling elements in the micro-environment and their potential therapeutic applications. Further therapeutic approaches will involve cell and molecular biology, the multiple hit theory of disease progression and analysis of system plasticity. Advances in the cell biology of eicosanoid and docosanoid metabolism, together with structure/function analysis of HUFA-derived mediators, will be useful in developing therapeutic agents in pathologies characterized by alterations in cell death signalling.
Collapse
Affiliation(s)
- J Davidson
- SIPBS, Strathclyde University, Glasgow, UK
| | | | | | | |
Collapse
|
23
|
KOEBERLE ANDREAS, WERZ OLIVER. Microsomal Prostaglandin E2 Synthase-1. ANTI-INFLAMMATORY DRUG DISCOVERY 2012. [DOI: 10.1039/9781849735346-00001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The prostanoids and leukotrienes (LTs) formed from arachidonic acid (AA) via the cyclooxygenase (COX)-1/2 and 5-lipoxygenase (5-LO) pathway, respectively, mediate inflammatory responses, chronic tissue remodelling, cancer, asthma and autoimmune disorders, but also possess homeostatic functions in the gastrointestinal tract, uterus, brain, kidney, vasculature and host defence. Based on the manifold functions of these eicosanoids, the clinical use of non-steroidal anti-inflammatory drugs (NSAIDs), a class of drugs that block formation of all prostanoids, is hampered by severe side-effects including gastrointestinal injury, renal irritations and cardiovascular risks. Therefore, anti-inflammatory agents interfering with eicosanoid biosynthesis require a well-balanced pharmacological profile to minimize these on-target side-effects. Current anti-inflammatory research aims at identifying compounds that can suppress the massive formation of pro-inflammatory prostaglandin (PG)E2 without affecting homeostatic PGE2 and PGI2 synthesis. The inducible microsomal prostaglandin E2 synthase-1 (mPGES-1) is one promising target enzyme. We will give an overview about the structure, regulation and function of mPGES-1 and then present novel inhibitors of mPGES-1 that may possess a promising pharmacological profile.
Collapse
Affiliation(s)
- ANDREAS KOEBERLE
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy University Jena Philosophenweg 14, D-07743 Jena Germany
| | - OLIVER WERZ
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy University Jena Philosophenweg 14, D-07743 Jena Germany
| |
Collapse
|
24
|
Li W, Graeber MB. The molecular profile of microglia under the influence of glioma. Neuro Oncol 2012; 14:958-78. [PMID: 22573310 DOI: 10.1093/neuonc/nos116] [Citation(s) in RCA: 259] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Microglia, which contribute substantially to the tumor mass of glioblastoma, have been shown to play an important role in glioma growth and invasion. While a large number of experimental studies on functional attributes of microglia in glioma provide evidence for their tumor-supporting roles, there also exist hints in support of their anti-tumor properties. Microglial activities during glioma progression seem multifaceted. They have been attributed to the receptors expressed on the microglia surface, to glioma-derived molecules that have an effect on microglia, and to the molecules released by microglia in response to their environment under glioma control, which can have autocrine effects. In this paper, the microglia and glioma literature is reviewed. We provide a synopsis of the molecular profile of microglia under the influence of glioma in order to help establish a rational basis for their potential therapeutic use. The ability of microglia precursors to cross the blood-brain barrier makes them an attractive target for the development of novel cell-based treatments of malignant glioma.
Collapse
Affiliation(s)
- Wei Li
- Brain Tumor Research Laboratories, The Brain and Mind Research Institute, University of Sydney, 94 Mallett St, Camperdown, Sydney, NSW 2050, Australia
| | | |
Collapse
|
25
|
Venza M, Visalli M, Alafaci C, Caffo M, Caruso G, Salpietro FM, Tomasello F, Teti D. Interleukin-8 overexpression in astrocytomas is induced by prostaglandin E2 and is associated with the transcription factors CCAAT/enhancer-binding protein-β and CCAAT/enhancer-binding homologous protein. Neurosurgery 2011; 69:713-21; discussion 721. [PMID: 21471847 DOI: 10.1227/neu.0b013e31821954c6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The upregulation of microsomal prostaglandin E synthase-1 (mPGES-1) and the overexpression of interleukin-8 (IL-8) have been separately linked to glioma malignancy. OBJECTIVE To evaluate (1) the correlation between the mRNA levels of IL-8, mPGES-1, and the main transcription factors (TFs) activating the IL-8 promoter in human brain tumors of different grades; (2) the role of prostaglandin E2 (PGE2) on IL-8 activation and the expression of these TFs in tumor-derived cells; and (3) the biological impact of PGE2 treatment and mPGES-1 silencing on IL-8 synthesis and tumorigenesis. METHODS Quantitative real-time polymerase chain reaction, transfection experiments, and cell proliferation and apoptosis assays were performed. RESULTS Regardless of histological grade, a significant positive association between IL-8 expression and mPGES-1, CCAAT/enhancer-binding protein-β (C/EBP-β) and C/EBP Homologous Protein (CHOP) mRNA levels was found only in astrogliomas (P < .001). The correlation was not significant in the other brain tumors. PGE2-treated astroglioma cells showed a marked upregulation of IL-8, C/EBP-β, and CHOP, as well as increased proliferation and decreased apoptosis compared with untreated cells. mPGES-1-silenced astroglioma cells displayed decreased IL-8 synthesis, accompanied by reduced cell growth and an increased rate of apoptosis. The other brain tumor cells were unaffected either by PGE2 treatment or by mPGES-1 knockout. CONCLUSION (1) PGE2 is responsible for IL-8 overexpression, independently of the malignancy grade, in astrogliomas only. (2) C/EBP-β and CHOP may be involved in mediating PGE2-induced IL-8 activation in these tumors. (3) mPGES-1 inhibition may have potential as a form of adjuvant therapy for astrogliomas.
Collapse
Affiliation(s)
- Mario Venza
- Department of Neurosciences, Psychiatry and Anaesthesiology, Neurosurgical Clinic, University of Messina, Messina, Italy
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Liu G, Hosomi N, Hitomi H, Pelisch N, Fu H, Masugata H, Murao K, Ueno M, Matsumoto M, Nishiyama A. Angiotensin II induces human astrocyte senescence through reactive oxygen species production. Hypertens Res 2011; 34:479-83. [DOI: 10.1038/hr.2010.269] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
27
|
Rizzo MT. Cyclooxygenase-2 in oncogenesis. Clin Chim Acta 2010; 412:671-87. [PMID: 21187081 DOI: 10.1016/j.cca.2010.12.026] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 12/20/2010] [Accepted: 12/21/2010] [Indexed: 02/07/2023]
Abstract
Compelling experimental and clinical evidence supports the notion that cyclooxygenase-2, the inducible isoform of cyclooxygenase, plays a crucial role in oncogenesis. Clinical and epidemiological data indicate that aberrant regulation of cyclooxygenase-2 in certain solid tumors and hematological malignancies is associated with adverse clinical outcome. Moreover, findings extrapolated from experimental studies in cultured tumor cells and animal tumor models indicate that cyclooxygenase-2 critically influences all stages of tumor development from tumor initiation to tumor progression. Cyclooxygenase-2 elicits cell-autonomous effects on tumor cells resulting in stimulation of growth, increased cell survival, enhanced tumor cell invasiveness, stimulation of neovascularization, and tumor evasion from the host immune system. Additionally, the oncogenic effects of cyclooxygenase-2 stem from its unique ability to impact tumor cell surroundings and create a proinflammatory environment conducive for tumor development, growth and progression. The initial enthusiasm generated by the availability of cyclooxygenase-2 selective inhibitors for cancer prevention and therapy has been lessened by the severe cardiovascular adverse side effects associated with their long-term use, as well as by the mixed results of recent clinical trials evaluating the efficacy of cyclooxygenase-2 inhibitors in adjuvant chemotherapy. Therefore, our ability to efficiently target the oncogenic effects of cyclooxygenase-2 for therapeutic and preventive purposes strictly depends on a better understanding of the spatial and temporal aspects of its activation in tumor cells along with a clearer elucidation of the signaling networks whereby cyclooxygenase-2 affects tumor cells and their interactions with the tumor microenvironment. This knowledge has the potential of leading to the identification of novel cyclooxygenase-2-dependent molecular and signaling networks that can be exploited to improve cancer prevention and therapy.
Collapse
Affiliation(s)
- Maria Teresa Rizzo
- Signal Transduction Laboratory, Methodist Research Institute, Clarian Health and Department of Pharmacology, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
28
|
Abstract
The Bax protein (Bcl-2-associated X protein) is pivotal for the apoptotic process. Bax, which resides in an inactive form in the cytosol of healthy cells, is activated during the early stages of apoptosis and becomes associated with mitochondria through poorly understood mechanisms. In this study, we show that a family of bioactive lipids, namely prostaglandins, regulates Bax-dependent apoptosis. The prostaglandin E(2) (PGE(2)) or its derivative PGA(2) binds to Bax, induces its change of conformation, and thereby triggers apoptosis. A cysteine present in the loop between the two transmembrane α-helices of Bax, Cys126 is critical for its activation. PGD(2) inhibits PGE(2) binding to Bax and PGE(2)-induced apoptosis, as well as cell death induced by staurosporine and UV-B in various cell lines. This result is consistent with the fact that apoptosis is accompanied during these treatments by an increase in PGE(2). This process is distinct, yet cooperative, from that involving the BH3-only protein Bid. Our results establish that the PGE(2)/PGD(2) balance is involved in a new early mechanism of control in the activation of Bax during apoptosis.
Collapse
|
29
|
Glioma cell death: cell-cell interactions and signalling networks. Mol Neurobiol 2010; 42:89-96. [PMID: 20443079 DOI: 10.1007/s12035-010-8135-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Accepted: 04/05/2010] [Indexed: 01/03/2023]
Abstract
The prognosis for patients with malignant gliomas is poor, but improvements may emerge from a better understanding of the pathophysiology of glioma signalling. Recent therapeutic developments have implicated lipid signalling in glioma cell death. Stress signalling in glioma cell death involves mitochondria and endoplasmic reticulum. Lipid mediators also signal via extrinsic pathways in glioma cell proliferation, migration and interaction with endothelial and microglial cells. Glioma cell death and tumour regression have been reported using polyunsaturated fatty acids in animal models, human ex vivo explants, glioma cell preparations and in clinical case reports involving intratumoral infusion. Cell death signalling was associated with generation of reactive oxygen intermediates and mitochondrial and other signalling pathways. In this review, evidence for mitochondrial responses to stress signals, including polyunsaturated fatty acids, peroxidizing agents and calcium is presented. Additionally, evidence for interaction of glioma cells with primary brain endothelial cells is described, modulating human glioma peroxidative signalling. Glioma responses to potential therapeutic agents should be analysed in systems reflecting tumour connectivity and CNS structural and functional integrity. Future insights may also be derived from studies of signalling in glioma-derived tumour stem cells.
Collapse
|
30
|
Prostaglandin E2 activates cAMP response element-binding protein in glioma cells via a signaling pathway involving PKA-dependent inhibition of ERK. Prostaglandins Other Lipid Mediat 2009; 91:18-29. [PMID: 20015475 DOI: 10.1016/j.prostaglandins.2009.12.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Revised: 12/03/2009] [Accepted: 12/07/2009] [Indexed: 12/13/2022]
Abstract
Prostaglandin E(2) (PGE(2)) plays a critical role in influencing the biological behavior of tumor cells. We previously demonstrated that PGE(2) stimulates human glioma cell growth via activation of protein kinase A (PKA) type II. This study was undertaken to further elucidate the intracellular pathways activated by PGE(2) downstream to PKA. Stimulation of U87-MG glioma cells with PGE(2) increased phosphorylation of the cyclic-AMP response element (CRE) binding protein CREB at Ser-133 and CREB-driven transcription in a dose- and time-dependent manner. Expression of dominant CREB constructs that interfere with CREB phosphorylation at Ser-133 or with its binding to the CRE site markedly decreased PGE(2)-induced CREB activation. Inhibition of PKA by H-89 or expression of a dominant negative PKA construct attenuated PGE(2)-induced CREB activation. Moreover, inhibition of PKA type II decreased PGE(2)-induced CREB-dependent transcription by 45% compared to vehicle-treated cells. To investigate the involvement of additional signaling pathways, U87-MG cells were pretreated with wortmannin or LY294002 to inhibit the PI3-kinase/AKT pathway. Both inhibitors had no effect on PGE(2)-induced CREB phosphorylation and transcriptional activity, suggesting that PGE(2) activates CREB in a PI3-kinase/AKT independent manner. Challenge of U87-MG cells with PGE(2), at concentrations that induced maximal CREB activation, or with forskolin inhibited extracellular signal-regulated kinase (ERK) phosphorylation. Pretreatment of U87-MG cells with the ERK inhibitor PD98059, accentuated ERK inhibition and increased CREB phosphorylation at Ser-133 and CREB-driven transcription stimulated by PGE(2), suggesting that inhibition of ERK contributes to PGE(2)-induced CREB activation. Inhibition of ERK by PGE(2) or by forskolin was rescued by treatment of cells with H-89 or by the dominant negative PKA construct. Moreover, PGE(2) or forskolin inhibited phosphorylation of Raf-1 phosphorylation at Ser-338. Challenge of U87-MG cells with 11-deoxy-PGE(1) increased CREB-driven transcription and stimulated cell growth, while other PGE(2) analogues had no effect. Together our results reveal a novel signaling pathway whereby PGE(2) signals through PKA to inhibit ERK and increase CREB transcriptional activity.
Collapse
|
31
|
Mattila S, Tuominen H, Koivukangas J, Stenbäck F. The terminal prostaglandin synthases mPGES-1, mPGES-2, and cPGES are all overexpressed in human gliomas. Neuropathology 2009; 29:156-65. [DOI: 10.1111/j.1440-1789.2008.00963.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
32
|
Hutchinson AJ, Chou CL, Israel DD, Xu W, Regan JW. Activation of EP2 prostanoid receptors in human glial cell lines stimulates the secretion of BDNF. Neurochem Int 2009; 54:439-46. [PMID: 19428786 DOI: 10.1016/j.neuint.2009.01.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Revised: 12/27/2008] [Accepted: 01/22/2009] [Indexed: 10/21/2022]
Abstract
Prostaglandin E(2) (PGE(2)) is produced at high levels in the injured central nervous system, where it is generally considered a cytotoxic mediator of inflammation. The cellular actions of PGE(2) are mediated by G-protein signaling activated by prostanoid receptors termed EP(1), EP(2), EP(3) and EP(4). Recent studies have implicated the EP(2) prostanoid receptor to be in apparently conflicting roles promoting neuronal death in some model systems and the survival of neurons in others. Here we show that treatment of immortalized human microglia and CCF-STTG1 astrocytes with either PGE(2) or the EP(2) selective agonist butaprost stimulates the release of brain-derived neurotrophic factor (BDNF). Both cell lines express mRNA for the EP(2) receptor, whereas transcripts for the other subtypes are not detected. Pharmacological studies using PGE(2) and modulators of cyclic AMP signaling implicate this pathway in PGE(2)-stimulated BDNF release. These results indicate that EP(2) prostanoid receptor activation induces BDNF secretion through stimulation of cyclic AMP dependent signaling. Our findings provide a mechanism by which endogenous PGE(2) might contribute to either neurotoxicity or neuroprotection in the injured brain via the induction of BDNF release from microglial cells and astrocytes.
Collapse
Affiliation(s)
- Anthony J Hutchinson
- Program in Neuroscience, The University of Arizona, College of Pharmacy, Gould-Simpson 611, PO Box 210077, Tucson, AZ 85721, USA
| | | | | | | | | |
Collapse
|
33
|
Walker WS, Leaver HA. Immunologic and Stress Responses Following Video-Assisted Thoracic Surgery and Open Pulmonary Lobectomy in Early Stage Lung Cancer. Thorac Surg Clin 2007; 17:241-9, ix. [PMID: 17626402 DOI: 10.1016/j.thorsurg.2007.04.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Conventional open major surgery evokes an injury response involving endocrine, neural, and immunologic mechanisms. The immunologic responses are characterized by release of cytokines, inflammatory mediators, and acute-phase proteins and by adverse disturbances in immune cell function. The use of a minimal access approach strategy is associated with a significant reduction in the cytokine response, as exemplified by reduced interleukin-6 levels and a corresponding reduction in acute-phase protein generation with reduced C-reactive protein levels. Circulating immune cell function and numbers also are better preserved. These changes have been demonstrated in comparing open with video-assisted thoracoscopic surgery (VATS) lobectomy and, together with further investigation into local immune function, may offer some insight into the excellent survival data reported for VATS resection of stage I non-small cell lung cancer.
Collapse
Affiliation(s)
- William S Walker
- Department of Thoracic Surgery, Royal Infirmary of Edinburgh, Little France, Edinburgh, EH16 4SA, Scotland, UK.
| | | |
Collapse
|