1
|
Zihlif M, Hameduh T, Bulatova N, Hammad H. Alteration in the expression of the chemotherapy resistance‑related genes in response to chronic and acute hypoxia in pancreatic cancer. Biomed Rep 2023; 19:88. [PMID: 37901880 PMCID: PMC10603373 DOI: 10.3892/br.2023.1670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 09/08/2023] [Indexed: 10/31/2023] Open
Abstract
Pancreatic cancer is currently one of the least curable types of human cancer and remains a key health problem. One of the most important characteristics of pancreatic cancer is its ability to grow under hypoxic conditions. Hypoxia is associated with resistance of cancer cells to radiotherapy and chemotherapy. It is a major contributor to pancreatic cancer genetic instability, which local and systemic resistance that may result in poor clinical outcome. Accordingly, identifying gene expression changes in cancer resistance genes that occur under hypoxic conditions may identify a new therapeutic target. The aim of the present study was to explore the association between hypoxia and resistance to chemotherapy and determine the alteration in the expression of cancer resistance-related genes in the presence of hypoxia. Pancreatic cancer cells (PANC-1) were exposed to 8 h hypoxic episodes (<1% oxygen) three times/week for a total of 20 episodes (chronic hypoxia) or 72 h hypoxic episodes twice/week for a total of 10 episodes (acute hypoxia). The alterations in gene expression were examined using reverse transcription-quantitative PCR array compared with normoxic cells. Chemoresistance of hypoxic cells toward doxorubicin was assessed using MTT cell proliferation assay. Both chronic and acute hypoxia induced chemoresistance toward doxorubicin in PANC-1 pancreatic cancer cell line. The greatest changes occurred in estrogen Receptor Alpha Gene (ESR1) and ETS Like-1 protein (ELK1) pathways, in nucleic transcription factor Peroxisome proliferator-activated receptors (PPARs) and in a cell cycle inhibitor cyclin dependent kinase inhibitor 1A (CDKN1A). The present study demonstrated that exposing cells to prolonged hypoxia results in different gene expression changes involving pleotropic pathways that serve a role in inducing resistance in pancreatic cancer.
Collapse
Affiliation(s)
- Malek Zihlif
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Tareq Hameduh
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Nailya Bulatova
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Hana Hammad
- Department of Biology, School of Science, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
2
|
Wang N, He X, Zhao J, Jiang H, Cheng X, Xia Y, Eric Xu H, He Y. Structural basis of leukotriene B4 receptor 1 activation. Nat Commun 2022; 13:1156. [PMID: 35241677 PMCID: PMC8894450 DOI: 10.1038/s41467-022-28820-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/07/2022] [Indexed: 12/18/2022] Open
Abstract
Leukotriene B4 receptor 1 (BLT1) plays crucial roles in the acute inflammatory responses and is a valuable target for anti-inflammation treatment, however, the mechanism by which leukotriene B4 (LTB4) activates receptor remains unclear. Here, we report the cryo-electron microscopy (cryo-EM) structure of the LTB4 -bound human BLT1 in complex with a Gi protein in an active conformation at resolution of 2.91 Å. In combination of molecule dynamics (MD) simulation, docking and site-directed mutagenesis, our structure reveals that a hydrogen-bond network of water molecules and key polar residues is the key molecular determinant for LTB4 binding. We also find that the displacement of residues M1013.36 and I2717.39 to the center of receptor, which unlock the ion lock of the lower part of pocket, is the key mechanism of receptor activation. In addition, we reveal a binding site of phosphatidylinositol (PI) and discover that the widely open ligand binding pocket may contribute the lack of specificity and efficacy for current BLT1-targeting drug design. Taken together, our structural analysis provides a scaffold for understanding BLT1 activation and a rational basis for designing anti-leukotriene drugs. In the paper, Dr. Wang et al reported a cryo-EM structure of the human leukotriene B4 receptor 1 (BLT1) in complex with its native ligand leukotriene B4 (LTB4) in an active conformation complexed with Gi protein. The structure reveals the molecule determinant of LTB4 binding and the mechanism of receptor activation. These structural information will boost the understanding of LTB4-BLT1 signaling and provide a rational basis for designing novel anti-leukotriene drugs.
Collapse
Affiliation(s)
- Na Wang
- Laboratory of Receptor Structure and Signaling, The HIT Center for Life Sciences, Harbin Institute of Technology, Harbin, 150001, China
| | - Xinheng He
- The CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jing Zhao
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Hualiang Jiang
- The CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xi Cheng
- The CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yu Xia
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - H Eric Xu
- The CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuanzheng He
- Laboratory of Receptor Structure and Signaling, The HIT Center for Life Sciences, Harbin Institute of Technology, Harbin, 150001, China.
| |
Collapse
|
3
|
Alizamani E, Ghorbanzadeh B, Naserzadeh R, Mansouri MT. Montelukast, a cysteinyl leukotriene receptor antagonist, exerts local antinociception in animal model of pain through the L-arginine/nitric oxide/cyclic GMP/K ATP channel pathway and PPARγ receptors. Int J Neurosci 2021; 131:1004-1011. [PMID: 32408781 DOI: 10.1080/00207454.2020.1769618] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 04/11/2020] [Accepted: 04/17/2020] [Indexed: 01/06/2023]
Abstract
OBJECTIVE The leukotrienes are inflammatory mediators. In the present study, the analgesic role of local montelukast, a cysteinyl leukotriene receptor antagonist, and the possible involvement of L-arginine/NO/cGMP/KATP channel pathway and PPARγ receptors was assessed in the formalin test in rats. METHODS AND RESULTS The local administration of montelukast into the hind paw produced dose-related analgesia during both phases of the formalin test. Furthermore, pre-treatment with L-NAME, methylene blue, and glibenclamide prevented montelukast (10 μg/paw)-induced antinociception in both early and late phases of the test. Moreover, the local L-arginine and diazoxide before the sub-effective dose of montelukast (3 μg/paw) produced an analgesic effect. Also, local GW-9662 blocked antinociception induced by montelukast plus pioglitazone (10 μg/paw). CONCLUSION In conclusion, montelukast produced peripheral analgesia through PPARγ receptors and activation of the L-arginine/NO/cGMP/KATP channel pathway, with potential for a new topical analgesic drug.
Collapse
Affiliation(s)
- Ehsan Alizamani
- Department of Pharmacology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Behnam Ghorbanzadeh
- Department of Pharmacology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
- Pain Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Reza Naserzadeh
- Department of Pharmacology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | | |
Collapse
|
4
|
Targeting Leukotrienes as a Therapeutic Strategy to Prevent Comorbidities Associated with Metabolic Stress. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1274:55-69. [PMID: 32894507 DOI: 10.1007/978-3-030-50621-6_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Leukotrienes (LTs) are potent lipid mediators that exert a variety of functions, ranging from maintaining the tone of the homeostatic immune response to exerting potent proinflammatory effects. Therefore, LTs are essential elements in the development and maintenance of different chronic diseases, such as asthma, arthritis, and atherosclerosis. Due to the pleiotropic effects of LTs in the pathogenesis of inflammatory diseases, studies are needed to discover potent and specific LT synthesis inhibitors and LT receptor antagonists. Even though most clinical trials using LT inhibitors or antagonists have failed due to low efficacy and/or toxicity, new drug development strategies are driving the discovery for LT inhibitors to prevent inflammatory diseases. A newly important detrimental role for LTs in comorbidities associated with metabolic stress has emerged in the last few years and managing LT production and/or actions could represent an exciting new strategy to prevent or treat inflammatory diseases associated with metabolic disorders. This review is intended to shed light on the synthesis and actions of leukotrienes, the most common drugs used in clinical trials, and discuss the therapeutic potential of preventing LT function in obesity, diabetes, and hyperlipidemia.
Collapse
|
5
|
Hirakata T, Matsuda A, Yokomizo T. Leukotriene B 4 receptors as therapeutic targets for ophthalmic diseases. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158756. [PMID: 32535236 DOI: 10.1016/j.bbalip.2020.158756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/01/2020] [Accepted: 06/04/2020] [Indexed: 11/26/2022]
Abstract
Leukotriene B4 (LTB4) is an inflammatory lipid mediator produced from arachidonic acid by multiple reactions catalyzed by two enzymes 5-lipoxygenase (5-LOX) and LTA4 hydrolase (LTA4H). The two receptors for LTB4 have been identified: a high-affinity receptor, BLT1, and a low-affinity receptor, BLT2. Our group identified 12(S)-hydroxy-5Z,8E,10E-heptadecatrienoic acid (12-HHT) as a high-affinity BLT2 ligand. Numerous studies have revealed critical roles for LTB4 and its receptors in various systemic diseases. Recently, we also reported the roles of LTB4, BLT1 and BLT2 in the murine ophthalmic disease models of mice including cornea wound, allergic conjunctivitis, and age-related macular degeneration. Moreover, other groups revealed the evidence of the ocular function of LTB4. In the present review, we introduce the roles of LTB4 and its receptors both in ophthalmic diseases and systemic inflammatory diseases. LTB4 and its receptors are putative novel therapeutic targets for systemic and ophthalmic diseases.
Collapse
Affiliation(s)
- Toshiaki Hirakata
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, Japan; Department of Biochemistry, Juntendo University Graduate School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, Japan
| | - Akira Matsuda
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, Japan
| | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
6
|
Zhang Z, Zhao G, Liu L, He J, Darwazeh R, Liu H, Chen H, Zhou C, Guo Z, Sun X. Bexarotene Exerts Protective Effects Through Modulation of the Cerebral Vascular Smooth Muscle Cell Phenotypic Transformation by Regulating PPARγ/FLAP/LTB 4 After Subarachnoid Hemorrhage in Rats. Cell Transplant 2019; 28:1161-1172. [PMID: 31010302 PMCID: PMC6767892 DOI: 10.1177/0963689719842161] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) play an important role after a subarachnoid hemorrhage (SAH). The changes in VSMCs following bexarotene treatment after SAH are unknown. In the present study, neurological impairment, decreased cerebral cortical blood flow and transformation of cerebral VSMCs from a contractile to a synthetic phenotype were observed after SAH. Bexarotene reduced neurological impairment, improved cerebral cortical blood flow, inhibited VSMC phenotypic transformation and suppressed the expression of 5-lipoxygenase-activating protein (FLAP) and leukotriene B4 (LTB4), which was partly reversed by GW9662, an inhibitor of peroxisome proliferator-activated receptor gamma (PPARγ). Mechanistically, sh-PPARγ-mediated phenotypic transformation of VSMCs was partially suppressed by MK886, an antagonist of FLAP. Therefore, we conclude that bexarotene reduced neurological impairment, improved cerebral cortical blood flow and inhibited the VSMC phenotypic transformation after SAH, which was achieved by activating PPARγ-mediated inhibition of FLAP/LTB4 in VSMCs.
Collapse
Affiliation(s)
- Zhaosi Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guosheng Zhao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liu Liu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junchi He
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Rami Darwazeh
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Han Liu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Chen
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chao Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zongduo Guo
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Scarpati M, Qi Y, Govind S, Singh S. A combined computational strategy of sequence and structural analysis predicts the existence of a functional eicosanoid pathway in Drosophila melanogaster. PLoS One 2019; 14:e0211897. [PMID: 30753230 PMCID: PMC6372189 DOI: 10.1371/journal.pone.0211897] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 01/22/2019] [Indexed: 02/07/2023] Open
Abstract
This study reports on a putative eicosanoid biosynthesis pathway in Drosophila melanogaster and challenges the currently held view that mechanistic routes to synthesize eicosanoid or eicosanoid-like biolipids do not exist in insects, since to date, putative fly homologs of most mammalian enzymes have not been identified. Here we use systematic and comprehensive bioinformatics approaches to identify most of the mammalian eicosanoid synthesis enzymes. Sensitive sequence analysis techniques identified candidate Drosophila enzymes that share low global sequence identities with their human counterparts. Twenty Drosophila candidates were selected based upon (a) sequence identity with human enzymes of the cyclooxygenase and lipoxygenase branches, (b) similar domain architecture and structural conservation of the catalytic domain, and (c) presence of potentially equivalent functional residues. Evaluation of full-length structural models for these 20 top-scoring Drosophila candidates revealed a surprising degree of conservation in their overall folds and potential analogs for functional residues in all 20 enzymes. Although we were unable to identify any suitable candidate for lipoxygenase enzymes, we report structural homology models of three fly cyclooxygenases. Our findings predict that the D. melanogaster genome likely codes for one or more pathways for eicosanoid or eicosanoid-like biolipid synthesis. Our study suggests that classical and/or novel eicosanoids mediators must regulate biological functions in insects–predictions that can be tested with the power of Drosophila genetics. Such experimental analysis of eicosanoid biology in a simple model organism will have high relevance to human development and health.
Collapse
Affiliation(s)
- Michael Scarpati
- Brooklyn College of the City University of New York, Brooklyn, New York, United States of America
- PhD program in Biology, Graduate Center of the City University of New York, New York, New York, United States of America
| | - Yan Qi
- Brooklyn College of the City University of New York, Brooklyn, New York, United States of America
- PhD program in Biology, Graduate Center of the City University of New York, New York, New York, United States of America
| | - Shubha Govind
- PhD program in Biology, Graduate Center of the City University of New York, New York, New York, United States of America
- PhD program in Biochemistry, Graduate Center of the City University of New York, New York, New York, United States of America
- The City College of the City University of New York, New York, New York, United States of America
| | - Shaneen Singh
- Brooklyn College of the City University of New York, Brooklyn, New York, United States of America
- PhD program in Biology, Graduate Center of the City University of New York, New York, New York, United States of America
- PhD program in Biochemistry, Graduate Center of the City University of New York, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
8
|
Merchant N, Bhaskar LV, Momin S, Sujatha P, Reddy AB, Nagaraju GP. 5-Lipoxygenase: Its involvement in gastrointestinal malignancies. Crit Rev Oncol Hematol 2018; 127:50-55. [DOI: 10.1016/j.critrevonc.2018.05.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/16/2018] [Accepted: 05/14/2018] [Indexed: 12/11/2022] Open
|
9
|
Arachidonic Acid Metabolite as a Novel Therapeutic Target in Breast Cancer Metastasis. Int J Mol Sci 2017; 18:ijms18122661. [PMID: 29292756 PMCID: PMC5751263 DOI: 10.3390/ijms18122661] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/02/2017] [Accepted: 12/06/2017] [Indexed: 12/14/2022] Open
Abstract
Metastatic breast cancer (BC) (also referred to as stage IV) spreads beyond the breast to the bones, lungs, liver, or brain and is a major contributor to the deaths of cancer patients. Interestingly, metastasis is a result of stroma-coordinated hallmarks such as invasion and migration of the tumor cells from the primary niche, regrowth of the invading tumor cells in the distant organs, proliferation, vascularization, and immune suppression. Targeted therapies, when used as monotherapies or combination therapies, have shown limited success in decreasing the established metastatic growth and improving survival. Thus, novel therapeutic targets are warranted to improve the metastasis outcomes. We have been actively investigating the cytochrome P450 4 (CYP4) family of enzymes that can biosynthesize 20-hydroxyeicosatetraenoic acid (20-HETE), an important signaling eicosanoid involved in the regulation of vascular tone and angiogenesis. We have shown that 20-HETE can activate several intracellular protein kinases, pro-inflammatory mediators, and chemokines in cancer. This review article is focused on understanding the role of the arachidonic acid metabolic pathway in BC metastasis with an emphasis on 20-HETE as a novel therapeutic target to decrease BC metastasis. We have discussed all the significant investigational mechanisms and put forward studies showing how 20-HETE can promote angiogenesis and metastasis, and how its inhibition could affect the metastatic niches. Potential adjuvant therapies targeting the tumor microenvironment showing anti-tumor properties against BC and its lung metastasis are discussed at the end. This review will highlight the importance of exploring tumor-inherent and stromal-inherent metabolic pathways in the development of novel therapeutics for treating BC metastasis.
Collapse
|
10
|
Bhatt L, Roinestad K, Van T, Springman E. Recent advances in clinical development of leukotriene B4 pathway drugs. Semin Immunol 2017; 33:65-73. [DOI: 10.1016/j.smim.2017.08.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 05/04/2017] [Accepted: 08/08/2017] [Indexed: 12/23/2022]
|
11
|
Polvani S, Tarocchi M, Tempesti S, Galli A. Nuclear receptors and pathogenesis of pancreatic cancer. World J Gastroenterol 2014; 20:12062-12081. [PMID: 25232244 PMCID: PMC4161795 DOI: 10.3748/wjg.v20.i34.12062] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 04/03/2014] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with a median overall survival time of 5 mo and the five years survival less than 5%, a rate essentially unchanged over the course of the years. A well defined progression model of accumulation of genetic alterations ranging from single point mutations to gross chromosomal abnormalities has been introduced to describe the origin of this disease. However, due to the its subtle nature and concurring events PDAC cure remains elusive. Nuclear receptors (NR) are members of a large superfamily of evolutionarily conserved ligand-regulated DNA-binding transcription factors functionally involved in important cellular functions ranging from regulation of metabolism, to growth and development. Given the nature of their ligands, NR are very tempting drug targets and their pharmacological modulation has been widely exploited for the treatment of metabolic and inflammatory diseases. There are now clear evidences that both classical ligand-activated and orphan NR are involved in the pathogenesis of PDAC from its very early stages; nonetheless many aspects of their role are not fully understood. The purpose of this review is to highlight the striking connections that link peroxisome proliferator activated receptors, retinoic acid receptors, retinoid X receptor, androgen receptor, estrogen receptors and the orphan NR Nur, chicken ovalbumin upstream promoter transcription factor II and the liver receptor homologue-1 receptor to PDAC development, connections that could lead to the identification of novel therapies for this disease.
Collapse
|
12
|
Knab LM, Grippo PJ, Bentrem DJ. Involvement of eicosanoids in the pathogenesis of pancreatic cancer: The roles of cyclooxygenase-2 and 5-lipoxygenase. World J Gastroenterol 2014; 20:10729-10739. [PMID: 25152576 PMCID: PMC4138453 DOI: 10.3748/wjg.v20.i31.10729] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 01/30/2014] [Accepted: 04/09/2014] [Indexed: 02/06/2023] Open
Abstract
The interplay between inflammation and cancer progression is a growing area of research. A combination of clinical, epidemiological, and basic science investigations indicate that there is a relationship between inflammatory changes in the pancreas and neoplastic progression. Diets high in ω-6 polyunsaturated fatty acids provide increased substrate for arachidonic acid metabolism by cyclooxygenase-2 (COX-2) and 5-lipoxygenase (5-LOX) to form eicosanoids. These eicosanoids directly contribute to pancreatic cancer cell proliferation. Both COX-2 and 5-LOX are upregulated in multiple cancer types, including pancreatic cancer. In vitro studies using pancreatic cancer cell lines have demonstrated upregulation of COX-2 and 5-LOX at both the mRNA and protein levels. When COX-2 and 5-LOX are blocked via a variety of mechanisms, cancer cell proliferation is abrogated both in vitro and in vivo. The mechanism of COX-2 has been shown to include effects on apoptosis as well as angiogenesis. 5-LOX has been implicated in apoptosis. The use of COX-2 and 5-LOX inhibitors in clinical studies in patients with pancreatic cancer has been limited. Patient enrollment has been restricted to those with advanced disease which makes evaluation of these drugs as chemopreventive agents difficult. COX-2 and 5-LOX expression have been shown to be present during the early neoplastic changes of pancreatic cancer, well before progression to invasive disease. This indicates that the ideal role for these interventions is early in the disease process as preventive agents, perhaps in patients with chronic pancreatitis or hereditary pancreatitis.
Collapse
|
13
|
Lu J, Liu L, Zhu Y, Zhang Y, Wu Y, Wang G, Zhang D, Xu J, Xie X, Ke R, Han D, Li S, Feng W, Xie M, Liu Y, Fang P, Shi H, He P, Liu Y, Sun X, Li M. PPAR-γ inhibits IL-13-induced collagen production in mouse airway fibroblasts. Eur J Pharmacol 2014; 737:133-9. [PMID: 24858619 DOI: 10.1016/j.ejphar.2014.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 05/02/2014] [Accepted: 05/07/2014] [Indexed: 01/15/2023]
Abstract
Interleukin-13 (IL-13) plays an important role in extracellular matrix production of airway remodeling in asthma. Activation of PPAR-γ has been shown to inhibit the occurrence of airway fibrosis in asthma, yet it remains unknown whether the effect of PPAR-γ on suppression of airway fibrosis is associated with the inhibition of IL-13 signaling. In the present study, primary cultured airway fibroblasts were stimulated with IL-13, and JAK inhibitor, PDGF receptor blocker and MEK inhibitor were applied to investigate the involvement of these pathways in IL-13-induced collagen production. Our results demonstrate that IL-13 dose- and time-dependently induced collagen production in primary cultured mouse airway fibroblasts; this effect was blocked by inhibition of JAK/STAT6 signal pathway. IL-13 also stimulated JAK/STAT6-dependent PDGF production, elevation of PDGF in turn activated ERK1/2 MAPK and caused collagen production. Activation of PPAR-γ by rosiglitazone reduced IL-13-induced collagen expression by suppression of STAT6-driven PDGF production. Our results indicate that activation of JAK/STAT6 signal and subsequent PDGF generation and ERK1/2 MAPK activation mediate IL-13-induced collagen production in airway fibroblasts. This study suggests that activation of PPAR-γ might be a novel strategy for the treatment of asthma partially by inhibition of airway fibrosis.
Collapse
Affiliation(s)
- Jiamei Lu
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Lu Liu
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Yanting Zhu
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Yonghong Zhang
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Yuanyuan Wu
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Guizuo Wang
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Dexin Zhang
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Jing Xu
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Xinming Xie
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Rui Ke
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Dong Han
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Shaojun Li
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Wei Feng
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Mei Xie
- Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Yun Liu
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Ping Fang
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Hongyang Shi
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Ping He
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Yuan Liu
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Xiuzhen Sun
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China
| | - Manxiang Li
- Respiratory Diseases Research Center, Xi׳an Jiaotong University, Shaanxi, Xi׳an 710004, PR China; Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi׳an Jiaotong University, No. 157, West 5th Road, Shaanxi, Xi׳an 710004, PR China.
| |
Collapse
|
14
|
Youssef J, Badr M. Peroxisome proliferator-activated receptors and cancer: challenges and opportunities. Br J Pharmacol 2012; 164:68-82. [PMID: 21449912 DOI: 10.1111/j.1476-5381.2011.01383.x] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs), members of the nuclear hormone receptor superfamily, function as transcription factors and modulators of gene expression. These actions allow PPARs to regulate a variety of biological processes and to play a significant role in several diseases and conditions. The current literature describes frequently opposing and paradoxical roles for the three PPAR isotypes, PPARα, PPARβ/δ and PPARγ, in cancer. While some studies have implicated PPARs in the promotion and development of cancer, others, in contrast, have presented evidence for a protective role for these receptors against cancer. In some tissues, the expression level of these receptors and/or their activation correlates with a positive outcome against cancer, while, in other tissue types, their expression and activation have the opposite effect. These disparate findings raise the possibility of (i) PPAR receptor-independent effects, including effects on receptors other than PPARs by the utilized ligands; (ii) cancer stage-specific effect; and/or (iii) differences in essential ligand-related pharmacokinetic considerations. In this review, we highlight the latest available studies on the role of the various PPAR isotypes in cancer in several major organs and present challenges as well as promising opportunities in the field.
Collapse
Affiliation(s)
- Jihan Youssef
- University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | | |
Collapse
|
15
|
Greene ER, Huang S, Serhan CN, Panigrahy D. Regulation of inflammation in cancer by eicosanoids. Prostaglandins Other Lipid Mediat 2011; 96:27-36. [PMID: 21864702 PMCID: PMC4051344 DOI: 10.1016/j.prostaglandins.2011.08.004] [Citation(s) in RCA: 223] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 08/05/2011] [Accepted: 08/08/2011] [Indexed: 12/12/2022]
Abstract
Inflammation in the tumor microenvironment is now recognized as one of the hallmarks of cancer. Endogenously produced lipid autacoids, locally acting small molecule lipid mediators, play a central role in inflammation and tissue homeostasis, and have recently been implicated in cancer. A well-studied group of autacoid mediators that are the products of arachidonic acid metabolism include: the prostaglandins, leukotrienes, lipoxins and cytochrome P450 (CYP) derived bioactive products. These lipid mediators are collectively referred to as eicosanoids and are generated by distinct enzymatic systems initiated by cyclooxygenases (COX 1 and 2), lipoxygenases (5-LOX, 12-LOX, 15-LOXa, 15-LOXb), and cytochrome P450s, respectively. These pathways are the target of approved drugs for the treatment of inflammation, pain, asthma, allergies, and cardiovascular disorders. Beyond their potent anti-inflammatory and anti-cancer effects, non-steroidal anti-inflammatory drugs (NSAIDs) and COX-2 specific inhibitors have been evaluated in both preclinical tumor models and clinical trials. Eicosanoid biosynthesis and actions can also be directly influenced by nutrients in the diet, as evidenced by the emerging role of omega-3 fatty acids in cancer prevention and treatment. Most research dedicated to using eicosanoids to inhibit tumor-associated inflammation has focused on the COX and LOX pathways. Novel experimental approaches that demonstrate the anti-tumor effects of inhibiting cancer-associated inflammation currently include: eicosanoid receptor antagonism, overexpression of eicosanoid metabolizing enzymes, and the use of endogenous anti-inflammatory lipid mediators. Here we review the actions of eicosanoids on inflammation in the context of tumorigenesis. Eicosanoids may represent a missing link between inflammation and cancer and thus could serve as therapeutic target(s) for inhibiting tumor growth.
Collapse
Affiliation(s)
- Emily R. Greene
- Vascular Biology Program, Children’s Hospital Boston, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Sui Huang
- Institute for Biocomplexity and Informatics, University of Calgary, Calgary, Canada
| | - Charles N. Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA.
| | - Dipak Panigrahy
- Vascular Biology Program, Children’s Hospital Boston, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| |
Collapse
|
16
|
Steinhilber D, Fischer AS, Metzner J, Steinbrink SD, Roos J, Ruthardt M, Maier TJ. 5-lipoxygenase: underappreciated role of a pro-inflammatory enzyme in tumorigenesis. Front Pharmacol 2010; 1:143. [PMID: 21833182 PMCID: PMC3153017 DOI: 10.3389/fphar.2010.00143] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2010] [Accepted: 12/10/2010] [Indexed: 01/14/2023] Open
Abstract
Leukotrienes constitute a group of bioactive lipids generated by the 5-lipoxygenase (5-LO) pathway. An increasing body of evidence supports an acute role for 5-LO products already during the earliest stages of pancreatic, prostate, and colorectal carcinogenesis. Several pieces of experimental data form the basis for this hypothesis and suggest a correlation between 5-LO expression and tumor cell viability. First, several independent studies documented an overexpression of 5-LO in primary tumor cells as well as in established cancer cell lines. Second, addition of 5-LO products to cultured tumor cells also led to increased cell proliferation and activation of anti-apoptotic signaling pathways. 5-LO antisense technology approaches demonstrated impaired tumor cell growth due to reduction of 5-LO expression. Lastly, pharmacological inhibition of 5-LO potently suppressed tumor cell growth by inducing cell cycle arrest and triggering cell death via the intrinsic apoptotic pathway. However, the documented strong cytotoxic off-target effects of 5-LO inhibitors, in combination with the relatively high concentrations of 5-LO products needed to achieve mitogenic effects in cell culture assays, raise concern over the assignment of the cause, and question the relationship between 5-LO products and tumorigenesis.
Collapse
Affiliation(s)
- Dieter Steinhilber
- Institute of Pharmaceutical Chemistry/ZAFES, Goethe University Frankfurt Frankfurt, Germany
| | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Eicosanoids, including prostaglandins and leukotrienes, are biologically active lipids that have been implicated in various pathological processes, such as inflammation and cancer. This Review highlights our understanding of the intricate roles of eicosanoids in epithelial-derived tumours and their microenvironment. The knowledge of how these lipids orchestrate the complex interactions between transformed epithelial cells and the surrounding stromal cells is crucial for understanding tumour evolution, progression and metastasis. Understanding the molecular mechanisms underlying the role of prostaglandins and other eicosanoids in cancer progression will help to develop more effective cancer chemopreventive and/or therapeutic agents.
Collapse
Affiliation(s)
- Dingzhi Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | | |
Collapse
|