1
|
Ratajczak W, Walczakiewicz K, Laszczyńska M, Chmielowiec K, Palma J, Drozd A, Lubkowska A, Sipak O. The profile of oxidative stress markers (arachidonic and linoleic acid derivatives) in patients with benign prostatic hyperplasia in relation to metabolic syndrome. Aging (Albany NY) 2025; 17:116-130. [PMID: 39773533 PMCID: PMC11810064 DOI: 10.18632/aging.206187] [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: 08/08/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025]
Abstract
So far, it has been proven that benign prostatic hyperplasia (BPH) is strongly associated with inflammation resulting from, i.a. the presence of infectious agent, autoimmune disease, aging process and lipid disorders associated with metabolic syndrome (MetS). We analyzed the association between serum eicosanoides (HETE, HODE, lipoxins, prostaglandin, and leucotrien) in aging man with benign prostatic hyperplasia (BPH) and healthy controls. The study involved 219 men (with BPH, n = 144; healthy controls, n = 75). We assessed the content arachidonic and linoleic acid derivatives in the serum samples of the study participants using liquid chromatography (HPLC). The levels of: RvE1 (p < 0.001); LXA4 5S,6R,15R (p = 0.001); 10S,17R-DiDHA (p < 0.001); MaR1 (p = 0.002); 9S-HODE (p < 0.05); 15S-HETE (p < 0.05); 12S-HETE (p < 0.001); 5-oxoETE (p < 0.05) and 5-HETE (p < 0.001) were significantly higher in patients with BPH than in the control group. PGE2 (p = 0.007), LTB4 (p < 0.001), and 18RS-HEPE (p < 0.001) were significantly higher in control group. We also analyzed the relationship between LXA4 5S,6R,15R serum levels of oxidative stress markers and concomitance of MetS. We noticed a relationship between levels and MetS (F1216 = 6.114965, p = 0.01). Our research results suggest that pro-inflammatory mediators and suppressors of inflammation are involved in the development of BPH, but their exact contribution has yet to be investigated.
Collapse
Affiliation(s)
- Weronika Ratajczak
- Department of Functional Diagnostics and Physical Medicine, Pomeranian Medical University, Żołnierska, Szczecin 71-210, Poland
| | | | - Maria Laszczyńska
- Department of Nursing, State University of Applied Sciences, Leśna, Koszalin 75-582, Poland
| | - Krzysztof Chmielowiec
- Department of Hygiene and Epidemiology, Collegium Medicum, University of Zielona Góra, Zyty St., Zielona Góra 65-046, Poland
| | - Joanna Palma
- Department of Biochemical Sciences, Pomeranian Medical University, Broniewskiego, Szczecin 71-460, Poland
| | - Arleta Drozd
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, Broniewskiego, Szczecin 71-460, Poland
| | - Anna Lubkowska
- Department of Functional Diagnostics and Physical Medicine, Pomeranian Medical University, Żołnierska, Szczecin 71-210, Poland
| | - Olimpia Sipak
- Department of Obstetrics and Pathology of Pregnancy, Pomeranian Medical University, Żołnierska, Szczecin 71-210, Poland
| |
Collapse
|
2
|
Liu X, Zhang J, Sun W, Cao J, Ma Z. COX-2 in lung cancer: Mechanisms, development, and targeted therapies. Chronic Dis Transl Med 2024; 10:281-292. [PMID: 39429482 PMCID: PMC11483542 DOI: 10.1002/cdt3.120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/13/2024] [Accepted: 02/22/2024] [Indexed: 10/22/2024] Open
Abstract
Lung cancer (LC) is the leading cause of cancer-related death worldwide, with non-small cell lung cancer (NSCLC) comprising 85% of all cases. COX-2, an enzyme induced significantly under stress conditions, catalyzes the conversion of free arachidonic acid into prostaglandins. It exhibits high expression in various tumors and is closely linked to LC progression. COX-2 functions as a pivotal driver in cancer pathogenesis by promoting prostaglandin E2 synthesis and facilitating tumor cell occurrence and development. Furthermore, COX-2 holds potential as a predictive marker for early-stage NSCLC, guiding targeted therapy in patients with early COX-2 overexpression. Additionally, combining COX-2 inhibitors with diverse treatment modalities enhances tumor therapeutic efficacy, minimizes adverse effects on healthy tissues, and improves overall patient survival rates posttreatment. In conclusion, combined therapy targeting COX-2 presents a promising novel strategy for NSCLC treatment, offering avenues for improving prognosis and effective tumor treatment. This review provides novel insights and ideas for developing new treatment strategies to improve the prognosis of NSCLC.
Collapse
Affiliation(s)
- Xueqi Liu
- Department of Respiratory MedicinePostgraduate Training Base of Jinzhou Medical University in the General Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Junli Zhang
- Department of Respiratory MedicineGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Wenwu Sun
- Department of Respiratory MedicineGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Jianping Cao
- Department of Respiratory MedicineGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| | - Zhuang Ma
- Department of Respiratory MedicineGeneral Hospital of Northern Theater CommandShenyangLiaoningChina
| |
Collapse
|
3
|
Meng YW, Liu JY. Pathological and pharmacological functions of the metabolites of polyunsaturated fatty acids mediated by cyclooxygenases, lipoxygenases, and cytochrome P450s in cancers. Pharmacol Ther 2024; 256:108612. [PMID: 38369063 DOI: 10.1016/j.pharmthera.2024.108612] [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: 10/30/2023] [Revised: 01/19/2024] [Accepted: 02/05/2024] [Indexed: 02/20/2024]
Abstract
Oxylipins have garnered increasing attention because they were consistently shown to play pathological and/or pharmacological roles in the development of multiple cancers. Oxylipins are the metabolites of polyunsaturated fatty acids via both enzymatic and nonenzymatic pathways. The enzymes mediating the metabolism of PUFAs include but not limited to lipoxygenases (LOXs), cyclooxygenases (COXs), and cytochrome P450s (CYPs) pathways, as well as the down-stream enzymes. Here, we systematically summarized the pleiotropic effects of oxylipins in different cancers through pathological and pharmacological aspects, with specific reference to the enzyme-mediated oxylipins. We discussed the specific roles of oxylipins on cancer onset, growth, invasion, and metastasis, as well as the expression changes in the associated metabolic enzymes and the associated underlying mechanisms. In addition, we also discussed the clinical application and potential of oxylipins and related metabolic enzymes as the targets for cancer prevention and treatment. We found the specific function of most oxylipins in cancers, especially the underlying mechanisms and clinic applications, deserves and needs further investigation. We believe that research on oxylipins will provide not only more therapeutic targets for various cancers but also dietary guidance for both cancer patients and healthy humans.
Collapse
Affiliation(s)
- Yi-Wen Meng
- CNTTI of the Institute of Life Sciences & Department of Anesthesia of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China; Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing 400016, China
| | - Jun-Yan Liu
- CNTTI of the Institute of Life Sciences & Department of Anesthesia of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China; Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing 400016, China; College of Pharmacy, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
4
|
Sieminska J, Kolmert J, Zurita J, Benkestock K, Revol-Cavalier J, Niklinski J, Reszec J, Dahlén SE, Ciborowski M, Wheelock CE. A single extraction 96-well method for LC-MS/MS quantification of urinary eicosanoids, steroids and drugs. Prostaglandins Other Lipid Mediat 2024; 170:106789. [PMID: 37879396 DOI: 10.1016/j.prostaglandins.2023.106789] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 10/27/2023]
Abstract
Urinary eicosanoid concentrations reflect inflammatory processes in multiple diseases and have been used as biomarkers of disease as well as suggested for patient stratification in precision medicine. However, implementation of urinary eicosanoid profiling in large-scale analyses is restricted due to sample preparation limits. Here we demonstrate a single solid-phase extraction of 300 µL urine in 96-well-format for prostaglandins, thromboxanes, isoprostanes, cysteinyl-leukotriene E4 and the linoleic acid-derived dihydroxy-octadecenoic acids (9,10- and 12,13-DiHOME). A simultaneous screening protocol was also developed for cortisol/cortisone and 7 exogenous steroids as well as 3 cyclooxygenase inhibitors. Satisfactory performance for quantification of eicosanoids with an appropriate internal standard was demonstrated for intra-plate analyses (CV = 8.5-15.1%) as well as for inter-plate (n = 35) from multiple studies (CV = 22.1-34.9%). Storage stability was evaluated at - 20 °C, and polar tetranors evidenced a 50% decrease after 5 months, while the remaining eicosanoids evidenced no significant degradation. All eicosanoids were stable over 3.5-years in urine stored at - 80 °C. This method will facilitate the implementation of urinary eicosanoid quantification in large-scale screening.
Collapse
Affiliation(s)
- Julia Sieminska
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Metabolomics Laboratory, Clinical Research Center, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Johan Kolmert
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Javier Zurita
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Johanna Revol-Cavalier
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jacek Niklinski
- Department of Clinical Molecular Biology, Medical University of Bialystok, Waszyngtona 13, 15-269 Bialystok, Poland
| | - Joanna Reszec
- Department of Medical Patomorphology, Medical University of Bialystok, Waszyngtona 13, 15-269 Bialystok, Poland
| | - Sven-Erik Dahlén
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Michal Ciborowski
- Metabolomics Laboratory, Clinical Research Center, Medical University of Bialystok, 15-276 Bialystok, Poland.
| | - Craig E Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
5
|
Geng S, Zhan H, Cao L, Geng L, Ren X. Targeting PTGES/PGE2 axis enhances sensitivity of colorectal cancer cells to 5-fluorouracil. Biochem Cell Biol 2023; 101:501-512. [PMID: 37358009 DOI: 10.1139/bcb-2023-0101] [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] [Indexed: 06/27/2023] Open
Abstract
Insensitivity and resistance to 5-fluorouracil (5FU) remain as major hurdles for effective and durable 5FU-based chemotherapy in colorectal cancer (CRC) patients. In this study, we identified prostaglandin E synthase (PTGES)/prostaglandin E2 (PGE2) axis as an important regulator for 5FU sensitivity in CRC cells. We found that PTGES expression and PGE2 production are elevated in CRC cells in comparison to normal colorectal epithelial cells. Depletion of PTGES significantly enhanced the inhibitory effect of 5FU on CRC cell viability that was fully reverted by exogenous supplement of PGE2. Inhibition of PTGES enzymatic function, by either inducing loss-of-function mutant or treatment with selective inhibitors, phenocopied the PTGES depletion in terms of 5FU sensitization. Mechanistically, PTGES/PGE2 axis modulates glycolysis in CRC cells, thereby regulating the 5FU sensitivity. Importantly, high PTGES expression is correlated with poor prognosis in 5FU-treated CRC patients. Thus, our study defines PTGES/PGE2 axis as a novel therapeutic target for enhancing the efficacy of 5FU-based chemotherapy in CRC.
Collapse
Affiliation(s)
- Song Geng
- Department of Colorectal Hernia Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Hao Zhan
- Department of Colorectal Hernia Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Lianmeng Cao
- Department of Gastrointestinal Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Longlong Geng
- Department of Colorectal Hernia Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xiang Ren
- Department of Colorectal Hernia Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| |
Collapse
|
6
|
Bülbül B, Ding K, Zhan CG, Çiftçi G, Yelekçi K, Gürboğa M, Özakpınar ÖB, Aydemir E, Baybağ D, Şahin F, Kulabaş N, Helvacıoğlu S, Charehsaz M, Tatar E, Özbey S, Küçükgüzel İ. Novel 1,2,4-triazoles derived from Ibuprofen: synthesis and in vitro evaluation of their mPGES-1 inhibitory and antiproliferative activity. Mol Divers 2023; 27:2185-2215. [PMID: 36331786 DOI: 10.1007/s11030-022-10551-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Some novel triazole-bearing ketone and oxime derivatives were synthesized from Ibuprofen. In vitro cytotoxic activities of all synthesized molecules against five cancer lines (human breast cancer MCF-7, human lung cancer A549, human prostate cancer PC-3, human cervix cancer HeLa, and human chronic myelogenous leukemia K562 cell lines) were evaluated by MTT assay. In addition, mouse embryonic fibroblast cells (NIH/3T3) were also evaluated to determine the selectivity. Compounds 18, 36, and 45 were found to be the most cytotoxic, and their IC50 values were in the range of 17.46-68.76 µM, against the tested cancer cells. According to the results, compounds 7 and 13 demonstrated good anti-inflammatory activity against the microsomal enzyme prostaglandin E2 synthase-1 (mPGES-1) enzyme at IC50 values of 13.6 and 4.95 µM. The low cytotoxicity and non-mutagenity of these compounds were found interesting. Also, these compounds significantly prevented tube formation in angiogenesis studies. In conclusion, the anti-inflammatory and angiogenesis inhibitory activities of these compounds without toxicity suggested that they may be promising agents in anti-inflammatory treatment and they may be supportive agents for the cancer treatment.
Collapse
Affiliation(s)
- Bahadır Bülbül
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Düzce University, Konuralp, Düzce, Turkey
- Department of Pharmaceutical Chemistry, Institute of Health Sciences, Marmara University, Dragos, Kartal, 34865, Istanbul, Turkey
| | - Kai Ding
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA
- Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA
| | - Chang-Guo Zhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA
- Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA
| | - Gamze Çiftçi
- Department of Bioinformatics and Genetics, Faculty of Engineering and Natural Sciences, Kadir Has University, Istanbul, Turkey
| | - Kemal Yelekçi
- Department of Bioinformatics and Genetics, Faculty of Engineering and Natural Sciences, Kadir Has University, Istanbul, Turkey
| | - Merve Gürboğa
- Department of Biochemistry, Faculty of Pharmacy, Marmara University, Haydarpaşa, 34668, Istanbul, Turkey
| | - Özlem Bingöl Özakpınar
- Department of Biochemistry, Faculty of Pharmacy, Marmara University, Haydarpaşa, 34668, Istanbul, Turkey
| | - Esra Aydemir
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Kayışdağı, Istanbul, Turkey
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Biruni University, Zeytinburnu, 34010, Turkey
| | - Deniz Baybağ
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Kayışdağı, Istanbul, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Kayışdağı, Istanbul, Turkey
| | - Necla Kulabaş
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Başıbüyük, 34854, Istanbul, Turkey
| | - Sinem Helvacıoğlu
- Department of Toxicology, Faculty of Pharmacy, Yeditepe University, Ataşehir, 34750, Istanbul, Turkey
| | - Mohammad Charehsaz
- Department of Toxicology, Faculty of Pharmacy, Yeditepe University, Ataşehir, 34750, Istanbul, Turkey
| | - Esra Tatar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Başıbüyük, 34854, Istanbul, Turkey
| | - Süheyla Özbey
- Department of Physics Engineering, Faculty of Engineering, Hacettepe University, Beytepe, 06800, Ankara, Turkey
| | - İlkay Küçükgüzel
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Başıbüyük, 34854, Istanbul, Turkey.
| |
Collapse
|
7
|
Proestler E, Donzelli J, Nevermann S, Breitwieser K, Koch LF, Best T, Fauth M, Wickström M, Harter PN, Kogner P, Lavieu G, Larsson K, Saul MJ. The multiple functions of miR-574-5p in the neuroblastoma tumor microenvironment. Front Pharmacol 2023; 14:1183720. [PMID: 37731742 PMCID: PMC10507178 DOI: 10.3389/fphar.2023.1183720] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/07/2023] [Indexed: 09/22/2023] Open
Abstract
Neuroblastoma is the most common extracranial solid tumor in childhood and arises from neural crest cells of the developing sympathetic nervous system. Prostaglandin E2 (PGE2) has been identified as a key pro-inflammatory mediator of the tumor microenvironment (TME) that promotes neuroblastoma progression. We report that the interaction between the microRNA miR-574-5p and CUG-binding protein 1 (CUGBP1) induces the expression of microsomal prostaglandin E2 synthase 1 (mPGES-1) in neuroblastoma cells, which contributes to PGE2 biosynthesis. PGE2 in turn specifically induces the sorting of miR-574-5p into small extracellular vesicles (sEV) in neuroblastoma cell lines. sEV are one of the major players in intercellular communication in the TME. We found that sEV-derived miR-574-5p has a paracrine function in neuroblastoma. It acts as a direct Toll-like receptor 7/8 (TLR7/8) ligand and induces α-smooth muscle actin (α-SMA) expression in fibroblasts, contributing to fibroblast differentiation. This is particularly noteworthy as it has an opposite function to that in the TME of lung carcinoma, another PGE2 dependent tumor type. Here, sEV-derived miR-574-5p has an autokrine function that inhibits PGE2 biosynthesis in lung cancer cells. We report that the tetraspanin composition on the surface of sEV is associated with the function of sEV-derived miR-574-5p. This suggests that the vesicles do not only transport miRs, but also appear to influence their mode of action.
Collapse
Affiliation(s)
- Eva Proestler
- Fachbereich Biologie, Technische Universität Darmstadt, Darmstadt, Germany
| | - Julia Donzelli
- Fachbereich Biologie, Technische Universität Darmstadt, Darmstadt, Germany
| | - Sheila Nevermann
- Fachbereich Biologie, Technische Universität Darmstadt, Darmstadt, Germany
| | - Kai Breitwieser
- Fachbereich Biologie, Technische Universität Darmstadt, Darmstadt, Germany
| | - Leon F. Koch
- Fachbereich Biologie, Technische Universität Darmstadt, Darmstadt, Germany
| | - Tatjana Best
- Fachbereich Biologie, Technische Universität Darmstadt, Darmstadt, Germany
- Merck KGaA, Darmstadt, Germany
| | - Maria Fauth
- Fachbereich Biologie, Technische Universität Darmstadt, Darmstadt, Germany
- Prolytic GmbH, a Kymos Company, Frankfurt, Germany
| | - Malin Wickström
- Childhood Cancer Research Unit, Department of Children’s and Women’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Patrick N. Harter
- Institute of Neurology (Edinger-Institute), University Hospital Frankfurt, Goethe University, Frankfurt am Main, Frankfurt, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Frankfurt, Germany
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Children’s and Women’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Grégory Lavieu
- INSERM U1316, UMR7057, Centre National de la Recherche Scientifique (CNRS), Université Paris Cité, Paris, France
| | - Karin Larsson
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Meike J. Saul
- Fachbereich Biologie, Technische Universität Darmstadt, Darmstadt, Germany
| |
Collapse
|
8
|
Babaoglu ZY, Kilic D. Virtual screening, molecular simulations and bioassays: Discovering novel microsomal prostaglandin E Synthase-1 (mPGES-1) inhibitors. Comput Biol Med 2023; 155:106616. [PMID: 36780799 DOI: 10.1016/j.compbiomed.2023.106616] [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: 06/14/2022] [Revised: 01/05/2023] [Accepted: 01/28/2023] [Indexed: 02/05/2023]
Abstract
Microsomal prostaglandin E synthase-1 (mPGES-1) is an inducible prostaglandin E synthase expressed following exposure to pro-inflammatory stimuli. The mPGES-1 enzyme represents a new target for the therapeutic treatment of acute and chronic inflammatory disorders and cancer. In the present study, compounds from the ZINC15 database with an indole scaffold were docked at the mPGES-1 binding site using Glide (high-throughput virtual screening [HTVS], standard precision [SP] and extra precision [XP]), and the stabilities of the complexes were determined by molecular simulation studies. Following HTVS, the top 10% compounds were retained and further screened by SP. Again, the top 10% of these compounds were retained. Finally, the Glide XP scores of the compounds were determined, 20% were analyzed, and the Prime MM-GBSA total free binding energies of the compounds were calculated. The molecular simulations (100 ns) of the reference ligand, LVJ, and the two best-scoring compounds were performed with the Desmond program to analyze the dynamics of the target protein-ligand complexes. In human lung cells treated with the hit compounds, cell viability by colorimetric method and PGE2 levels by immunoassay method were determined. These in vitro experiments demonstrated that the two indole-containing hit compounds are potential novel inhibitors of mPGES-1 and are, therefore, potential therapeutic agents for cancer/inflammation therapies. Moreover, the compounds are promising lead mPGES-1 inhibitors for novel molecule design.
Collapse
Affiliation(s)
| | - Deryanur Kilic
- Department of Chemistry, Faculty of Science, Atatürk University, Erzurum, Turkey.
| |
Collapse
|
9
|
Stewart MJ, Weaver LM, Ding K, Kyomuhangi A, Loftin CD, Zheng F, Zhan CG. Analgesic effects of a highly selective mPGES-1 inhibitor. Sci Rep 2023; 13:3326. [PMID: 36849491 PMCID: PMC9971260 DOI: 10.1038/s41598-023-30164-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 02/16/2023] [Indexed: 03/01/2023] Open
Abstract
The growing opioid use and overdose crisis in the US is closely related to the abuse of pain medications. Particularly for postoperative pain (POP), ~ 310 million major surgeries are performed globally per year. Most patients undergoing surgical procedures experience acute POP, and ~ 75% of those with POP report the severity as moderate, severe, or extreme. Opioid analgesics are the mainstay for POP management. It is highly desirable to develop a truly effective and safe non-opioid analgesic to treat POP and other forms of pain. Notably, microsomal prostaglandin E2 (PGE2) synthase-1 (mPGES-1) was once proposed as a potentially promising target for a next generation of anti-inflammatory drugs based on studies in mPGES-1 knockouts. However, to the best of our knowledge, no studies have ever been reported to explore whether mPGES-1 is also a potential target for POP treatment. In this study, we demonstrate for the first time that a highly selective mPGES-1 inhibitor can effectively relieve POP as well as other forms of pain through blocking the PGE2 overproduction. All the data have consistently demonstrated that mPGES-1 is a truly promising target for treatment of POP as well as other forms of pain.
Collapse
Affiliation(s)
- Madeline J. Stewart
- grid.266539.d0000 0004 1936 8438Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536 USA ,grid.266539.d0000 0004 1936 8438Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536 USA
| | - Lauren M. Weaver
- grid.266539.d0000 0004 1936 8438Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536 USA ,grid.266539.d0000 0004 1936 8438Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536 USA
| | - Kai Ding
- grid.266539.d0000 0004 1936 8438Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536 USA
| | - Annet Kyomuhangi
- grid.266539.d0000 0004 1936 8438Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536 USA ,grid.266539.d0000 0004 1936 8438Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536 USA
| | - Charles D. Loftin
- grid.266539.d0000 0004 1936 8438Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536 USA
| | - Fang Zheng
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA. .,Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA.
| | - Chang-Guo Zhan
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA. .,Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY, 40536, USA.
| |
Collapse
|
10
|
Synthesis, in vitro and in silico studies on novel 3-aryloxymethyl-5-[(2-oxo-2-arylethyl)sulfanyl]-1,2,4-triazoles and their oxime derivatives as potent inhibitors of mPGES-1. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
11
|
Guryleva MV, Penzar DD, Chistyakov DV, Mironov AA, Favorov AV, Sergeeva MG. Investigation of the Role of PUFA Metabolism in Breast Cancer Using a Rank-Based Random Forest Algorithm. Cancers (Basel) 2022; 14:cancers14194663. [PMID: 36230586 PMCID: PMC9562210 DOI: 10.3390/cancers14194663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 09/15/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Polyunsaturated fatty acids (PUFAs) and their derivatives, oxylipins, are a constant focus of cancer research due to the relationship between cancer and processes of energy metabolism and inflammation, where a PUFA system is an active player. Only recently have methods been developed that allow for studying such complex systems. Using the Rank-based Random Forest (RF) model, we show that PUFA metabolism genes are critical for the pathogenesis of breast cancer (BC); BC subtypes differ in PUFA metabolism gene expression. The enrichment of BC subtypes with various genes associated with oxylipin signaling pathways indicates a different contribution of these compounds to the biology of subtypes. Abstract Polyunsaturated fatty acid (PUFA) metabolism is currently a focus in cancer research due to PUFAs functioning as structural components of the membrane matrix, as fuel sources for energy production, and as sources of secondary messengers, so called oxylipins, important players of inflammatory processes. Although breast cancer (BC) is the leading cause of cancer death among women worldwide, no systematic study of PUFA metabolism as a system of interrelated processes in this disease has been carried out. Here, we implemented a Boruta-based feature selection algorithm to determine the list of most important PUFA metabolism genes altered in breast cancer tissues compared with in normal tissues. A rank-based Random Forest (RF) model was built on the selected gene list (33 genes) and applied to predict the cancer phenotype to ascertain the PUFA genes involved in cancerogenesis. It showed high-performance of dichotomic classification (balanced accuracy of 0.94, ROC AUC 0.99) We also retrieved a list of the important PUFA genes (46 genes) that differed between molecular subtypes at the level of breast cancer molecular subtypes. The balanced accuracy of the classification model built on the specified genes was 0.82, while the ROC AUC for the sensitivity analysis was 0.85. Specific patterns of PUFA metabolic changes were obtained for each molecular subtype of breast cancer. These results show evidence that (1) PUFA metabolism genes are critical for the pathogenesis of breast cancer; (2) BC subtypes differ in PUFA metabolism genes expression; and (3) the lists of genes selected in the models are enriched with genes involved in the metabolism of signaling lipids.
Collapse
Affiliation(s)
- Mariia V. Guryleva
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Dmitry D. Penzar
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Dmitry V. Chistyakov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Correspondence: ; Tel.: +7-495-939-4332
| | - Andrey A. Mironov
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
- Kharkevich Institute of Information Transmission Problems, Russian Academy of Sciences, 127051 Moscow, Russia
| | - Alexander V. Favorov
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
- School of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Marina G. Sergeeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| |
Collapse
|
12
|
Prostanoid Metabolites as Biomarkers in Human Disease. Metabolites 2022; 12:metabo12080721. [PMID: 36005592 PMCID: PMC9414732 DOI: 10.3390/metabo12080721] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Prostaglandins (PGD2, PGE2, PGF2α), prostacyclin (PGI2), and thromboxane A2 (TXA2) together form the prostanoid family of lipid mediators. As autacoids, these five primary prostanoids propagate intercellular signals and are involved in many physiological processes. Furthermore, alterations in their biosynthesis accompany a wide range of pathological conditions, which leads to substantially increased local levels during disease. Primary prostanoids are chemically instable and rapidly metabolized. Their metabolites are more stable, integrate the local production on a systemic level, and their analysis in various biological matrices yields valuable information under different pathological settings. Therefore, prostanoid metabolites may be used as diagnostic, predictive, or prognostic biomarkers in human disease. Although their potential as biomarkers is great and extensive research has identified major prostanoid metabolites that serve as target analytes in different biofluids, the number of studies that correlate prostanoid metabolite levels to disease outcome is still limited. We review the metabolism of primary prostanoids in humans, summarize the levels of prostanoid metabolites in healthy subjects, and highlight existing biomarker studies. Since analysis of prostanoid metabolites is challenging because of ongoing metabolism and limited half-lives, an emphasis of this review lies on the reliable measurement and interpretation of obtained levels.
Collapse
|
13
|
Donzelli J, Proestler E, Riedel A, Nevermann S, Hertel B, Guenther A, Gattenlöhner S, Savai R, Larsson K, Saul MJ. Small extracellular vesicle-derived miR-574-5p regulates PGE2-biosynthesis via TLR7/8 in lung cancer. J Extracell Vesicles 2021; 10:e12143. [PMID: 34596365 PMCID: PMC8485338 DOI: 10.1002/jev2.12143] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/20/2021] [Accepted: 08/19/2021] [Indexed: 12/30/2022] Open
Abstract
Intercellular communication plays an essential role in lung cancer (LC). One of the major players in cell-cell-communication is small extracellular vesicles (sEV). SEV trigger various biological responses by transporting cellular cargo to target cells. One essential sEV component are microRNAs (miRs), whose transport has recently attracted increasing research interest. We report that prostaglandin E2 (PGE2 ), a key inflammatory lipid mediator, specifically induces the sorting of miR-574-5p in sEV of A549 and 2106T cells. We found that sEV-derived miR-574-5p activates Toll-like receptors (TLR) 7/8, thereby decreasing PGE2 -levels. In contrast, intracellular miR-574-5p induces PGE2 -biosynthesis. Consequently, the combination of intracellular and sEV-derived miR-574-5p controls PGE2 -levels via a feedback loop. This was only observed in adeno- but not in squamous cell carcinoma, indicating a cell-specific response to sEV-derived miRs, which might be due to unique tetraspanin compositions. Hence, we describe a novel function of miR-574-5p unique to adenocarcinoma. Intracellular miR-574-5p induces PGE2 and thus the secretion of sEV-derived miR-574-5p, which in turn decreases PGE2 -biosynthesis in recipient cells.
Collapse
Affiliation(s)
- Julia Donzelli
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| | - Eva Proestler
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| | - Anna Riedel
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| | - Sheila Nevermann
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| | - Brigitte Hertel
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| | - Andreas Guenther
- Department of Internal MedicineMember of the German Centre for Lung Research (DZL)Member of Cardio‐Pulmonary Institute (CPI)Justus Liebig UniversityGiessenGermany
| | | | - Rajkumar Savai
- Department of Internal MedicineMember of the German Centre for Lung Research (DZL)Member of Cardio‐Pulmonary Institute (CPI)Justus Liebig UniversityGiessenGermany
- Department of Lung Development and RemodellingMember of the DZLMember of CPIMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
- Lung Microenvironmental Niche in CancerogenesisInstitute for Lung Health (ILH)Justus Liebig UniversityGiessenGermany
| | - Karin Larsson
- Rheumatology UnitDepartment of MedicineKarolinska University HospitalStockholmSweden
| | - Meike J. Saul
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| |
Collapse
|
14
|
Markosyan N, Smyth EM, Patrignani P, Ricciotti E. Editorial: Eicosanoids in Cancer. Front Pharmacol 2021; 12:765214. [PMID: 34566667 PMCID: PMC8458895 DOI: 10.3389/fphar.2021.765214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 11/23/2022] Open
Affiliation(s)
- Nune Markosyan
- Department of Medicine, Abramson Family Cancer Research Institute, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States
| | - Emer M Smyth
- Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, United States
| | - Paola Patrignani
- Department of Neuroscience, Imaging and Clinical Sciences, School of Medicine, CAST (Center for Advanced Studies and Technology), "G. d'Annunzio" University, Chieti, Italy
| | - Emanuela Ricciotti
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
15
|
Jiang X, Renkema H, Smeitink J, Beyrath J. Sonlicromanol's active metabolite KH176m normalizes prostate cancer stem cell mPGES-1 overexpression and inhibits cancer spheroid growth. PLoS One 2021; 16:e0254315. [PMID: 34242345 PMCID: PMC8270194 DOI: 10.1371/journal.pone.0254315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Aggressiveness of cancers, like prostate cancer, has been found to be associated with elevated expression of the microsomal prostaglandin E synthase-1 (mPGES-1). Here, we investigated whether KH176m (the active metabolite of sonlicromanol), a recently discovered selective mPGES-1 inhibitor, could affect prostate cancer cells-derived spheroid growth. We demonstrated that KH176m suppressed mPGES-1 expression and growth of DU145 (high mPGES-1 expression)-derived spheroids, while it had no effect on the LNCaP cell line, which has low mPGES-1 expression. By addition of exogenous PGE2, we found that the effect of KH176m on mPGES-1 expression and spheroid growth is due to the inhibition of a PGE2-driven positive feedback control-loop of mPGES-1 transcriptional regulation. Cancer stem cells (CSCs) are a subset of cancer cells exhibiting the ability of self-renewal, plasticity, and initiating and maintaining tumor growth. Our data shows that mPGES-1 is specifically expressed in this CSCs subpopulation (CD44+CD24-). KH176m inhibited the expression of mPGES-1 and reduced the growth of spheroids derived from the CSC. Based on the results obtained we propose selective mPGES-1 targeting by the sonlicromanol metabolite KH176m as a potential novel treatment approach for cancer patients with high mPGES-1 expression.
Collapse
Affiliation(s)
- Xiaolan Jiang
- Khondrion BV, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
16
|
Di Micco S, Terracciano S, Ruggiero D, Potenza M, Vaccaro MC, Fischer K, Werz O, Bruno I, Bifulco G. Identification of 2-(thiophen-2-yl)acetic Acid-Based Lead Compound for mPGES-1 Inhibition. Front Chem 2021; 9:676631. [PMID: 34046398 PMCID: PMC8144515 DOI: 10.3389/fchem.2021.676631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/07/2021] [Indexed: 11/13/2022] Open
Abstract
We report the implementation of our in silico/synthesis pipeline by targeting the glutathione-dependent enzyme mPGES-1, a valuable macromolecular target in both cancer therapy and inflammation therapy. Specifically, by using a virtual fragment screening approach of aromatic bromides, straightforwardly modifiable by the Suzuki-Miyaura reaction, we identified 3-phenylpropanoic acid and 2-(thiophen-2-yl)acetic acid to be suitable chemical platforms to develop tighter mPGES-1 inhibitors. Among these, compounds 1c and 2c showed selective inhibitory activity against mPGES-1 in the low micromolar range in accordance with molecular modeling calculations. Moreover, 1c and 2c exhibited interesting IC50 values on A549 cell lines compared to CAY10526, selected as reference compound. The most promising compound 2c induced the cycle arrest in the G0/G1 phase at 24 h of exposure, whereas at 48 and 72 h, it caused an increase of subG0/G1 fraction, suggesting an apoptosis/necrosis effect.
Collapse
Affiliation(s)
- Simone Di Micco
- European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
| | | | - Dafne Ruggiero
- Dipartimento di Farmacia, University degli Studi di Salerno, Fisciano, Italy
| | - Marianna Potenza
- Dipartimento di Farmacia, University degli Studi di Salerno, Fisciano, Italy
| | - Maria C Vaccaro
- Dipartimento di Farmacia, University degli Studi di Salerno, Fisciano, Italy
| | - Katrin Fischer
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Oliver Werz
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Ines Bruno
- Dipartimento di Farmacia, University degli Studi di Salerno, Fisciano, Italy
| | - Giuseppe Bifulco
- Dipartimento di Farmacia, University degli Studi di Salerno, Fisciano, Italy
| |
Collapse
|
17
|
Mechanism of action and potential applications of selective inhibition of microsomal prostaglandin E synthase-1-mediated PGE 2 biosynthesis by sonlicromanol's metabolite KH176m. Sci Rep 2021; 11:880. [PMID: 33441600 PMCID: PMC7806836 DOI: 10.1038/s41598-020-79466-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/08/2020] [Indexed: 01/29/2023] Open
Abstract
Increased prostaglandin E2 (PGE2) levels were detected in mitochondrial disease patient cells harboring nuclear gene mutations in structural subunits of complex I, using a metabolomics screening approach. The increased levels of this principal inflammation mediator normalized following exposure of KH176m, an active redox-modulator metabolite of sonlicromanol (KH176). We next demonstrated that KH176m selectively inhibited lipopolysaccharide (LPS) or interleukin-1β (IL-1β)-induced PGE2 production in control skin fibroblasts. Comparable results were obtained in the mouse macrophage-like cell line RAW264.7. KH176m selectively inhibited mPGES-1 activity, as well as the inflammation-induced expression of mPGES-1. Finally, we showed that the effect of KH176m on mPGES-1 expression is due to the inhibition of a PGE2-driven positive feedback control-loop of mPGES-1 transcriptional regulation. Based on the results obtained we discuss potential new therapeutic applications of KH176m and its clinical stage parent drug candidate sonlicromanol in mitochondrial disease and beyond.
Collapse
|
18
|
Monteleone NJ, Lutz CS. miR-708-5p enhances erlotinib/paclitaxel efficacy and overcomes chemoresistance in lung cancer cells. Oncotarget 2020; 11:4699-4721. [PMID: 33473256 PMCID: PMC7771713 DOI: 10.18632/oncotarget.27840] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/23/2020] [Indexed: 12/16/2022] Open
Abstract
Lung cancer is a collection of aggressive tumors generally not diagnosed until late-stage, resulting in high mortality rates. The vast majority of non-small cell lung cancer (NSCLC) patients undergo combinatory chemotherapeutic treatment, which initially reduces tumor growth, but frequently becomes ineffective due to toxicity and resistance. Researchers have identified multiple signaling pathways involved in lung cancer chemoresistance, including cyclooxygenase-2 (COX-2)/microsomal prostaglandin E synthase-1 (mPGES-1) derived prostaglandin E2 (PGE2). While COX-2 inhibitors have shown promise in the clinic, their use is limited due to severe side effects. One novel approach to effectively suppress COX-2 signaling is through microRNA (miRNA). MiRNAs are small-noncoding RNAs commonly misexpressed in cancer. One tumor suppressive miRNA, miR-708-5p, has been shown to repress pro-resistant signaling pathways, including COX-2 and mPGES-1. Here, we demonstrate that chemotherapies reduce COX-2 expression, possibly through induction of miR-708-5p. Moreover, combination treatment of erlotinib (ERL) or paclitaxel (PAC) with miR-708-5p enhances COX-2 and mPGES-1 protein suppression. We also show that combination chemotherapeutic and miR-708-5p treatment intensifies the anti-proliferative and pro-apoptotic effects of ERL and PAC. We also created ERL and PAC resistant lung cancer cell lines, which have increased COX-2 expression and diminished miR-708-5p levels compared to naïve lung cancer cells. While ERL and PAC treatments do not alter resistant cell phenotype alone, combination treatment with miR-708-5p partially restores the chemotherapies' anti-proliferative effects and fully restores their pro-apoptotic qualities. These data suggest miR-708-5p may have potential combinatory therapeutic value to more efficaciously treat lung tumors while overcoming chemoresistance.
Collapse
Affiliation(s)
- Nicholas J Monteleone
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical & Health Sciences, New Jersey Medical School, School of Graduate Studies, Newark, NJ 07103, USA
| | - Carol S Lutz
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical & Health Sciences, New Jersey Medical School, School of Graduate Studies, Newark, NJ 07103, USA
| |
Collapse
|
19
|
Daouk R, Bahmad HF, Saleh E, Monzer A, Ballout F, Kadara H, Abou-Kheir W. Genome-wide gene expression analysis of a murine model of prostate cancer progression: Deciphering the roles of IL-6 and p38 MAPK as potential therapeutic targets. PLoS One 2020; 15:e0237442. [PMID: 32790767 PMCID: PMC7425932 DOI: 10.1371/journal.pone.0237442] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Prostate cancer (PCa) is the most commonly diagnosed cancer and the second leading cause of cancer-related deaths among adult males globally. The poor prognosis of PCa is largely due to late diagnosis of the disease when it has already progressed to an advanced stage marked by androgen-independence, thus necessitating new strategies for early detection and treatment. We construe that these direly needed advances are limited by our poor understanding of early events in the progression of PCa and that would thus represent ideal targets for early intervention. To begin to fill this void, we interrogated molecular "oncophenotypes" that embody the transition of PCa from an androgen-dependent (AD) to-independent (AI) state. METHODS To accomplish this aim, we used our previously established AD and AI murine PCa cell lines, PLum-AD and PLum-AI, respectively, which recapitulate primary and progressive PCa morphologically and molecularly. We statistically surveyed global gene expressions in these cell lines by microarray analysis. Differential profiles were functionally interrogated by pathways, gene set enrichment and topological gene network analyses. RESULTS Gene expression analysis of PLum-AD and PLum-AI transcriptomes (n = 3 each), revealed 723 differentially expressed genes (392 upregulated and 331 downregulated) in PLum-AI compared to PLum-AD cells. Gene set analysis demonstrated enrichment of biological functions and pathways in PLum-AI cells that are central to tumor aggressiveness including cell migration and invasion facilitated by epithelial-to-mesenchymal transition (EMT). Further analysis demonstrated that the p38 mitogen-activated protein kinase (MAPK) was predicted to be significantly activated in the PLum-AI cells, whereas gene sets previously associated with favorable response to the p38 inhibitor SB203580 were attenuated (i.e., inversely enriched) in the PLum-AI cells, suggesting that these aggressive cells may be therapeutically vulnerable to p38 inhibition. Gene set and gene-network analysis also alluded to activation of other signaling networks particularly those associated with enhanced EMT, inflammation and immune function/response including, but not limited to Tnf, IL-6, Mmp 2, Ctgf, and Ptges. Accordingly, we chose SB203580 and IL-6 to validate their effect on PLum-AD and PLum-AI. Some of the common genes identified in the gene-network analysis were validated at the molecular and functional level. Additionally, the vulnerability to SB203580 and the effect of IL-6 were also validated on the stem/progenitor cell population using the sphere formation assay. CONCLUSIONS In summary, our study highlights pathways associated with an augmented malignant phenotype in AI cells and presents new high-potential targets to constrain the aggressive malignancy seen in the castration-resistant PCa.
Collapse
Affiliation(s)
- Reem Daouk
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hisham F. Bahmad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL, United States of America
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States of America
| | - Eman Saleh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Alissar Monzer
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Farah Ballout
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
20
|
DREAM-in-CDM Approach and Identification of a New Generation of Anti-inflammatory Drugs Targeting mPGES-1. Sci Rep 2020; 10:10187. [PMID: 32576928 PMCID: PMC7311425 DOI: 10.1038/s41598-020-67283-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 06/01/2020] [Indexed: 11/28/2022] Open
Abstract
Microsomal prostaglandin E2 synthase-1 (mPGES-1) is known as an ideal target for next generation of anti-inflammatory drugs without the side effects of currently available anti-inflammatory drugs. However, there has been no clinically promising mPGES-1 inhibitor identified through traditional drug discovery and development route. Here we report a new approach, called DREAM-in-CDM (Drug Repurposing Effort Applying Integrated Modeling-in vitro/vivo-Clinical Data Mining), to identify an FDA-approved drug suitable for use as an effective analgesic targeting mPGES-1. The DREAM-in-CDM approach consists of three steps: computational screening of FDA-approved drugs; in vitro and/or in vivo assays; and clinical data mining. By using the DREAM-in-CDM approach, lapatinib has been identified as a promising mPGES-1 inhibitor which may have significant anti-inflammatory effects to relieve various forms of pain and possibly treat various inflammation conditions involved in other inflammation-related diseases such as the lung inflammation caused by the newly identified COVID-19. We anticipate that the DREAM-in-CDM approach will be used to repurpose FDA-approved drugs for various new therapeutic indications associated with new targets.
Collapse
|
21
|
Chini MG, Giordano A, Potenza M, Terracciano S, Fischer K, Vaccaro MC, Colarusso E, Bruno I, Riccio R, Koeberle A, Werz O, Bifulco G. Targeting mPGES-1 by a Combinatorial Approach: Identification of the Aminobenzothiazole Scaffold to Suppress PGE 2 Levels. ACS Med Chem Lett 2020; 11:783-789. [PMID: 32435385 DOI: 10.1021/acsmedchemlett.9b00618] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/05/2020] [Indexed: 02/06/2023] Open
Abstract
Microsomal prostaglandin E2 synthase-1 (mPGES-1), the terminal enzyme responsible for the production of inducible prostaglandin E2, has become an attractive target for the treatment of inflammation and cancer pathologies. Starting from an aminobenzothiazole scaffold, used as an unprecedented chemical core for mPGES-1 inhibition, a Combinatorial Virtual Screening campaign was conducted, using the X-ray crystal structure of human mPGES-1. Two combinatorial libraries (6 × 104) were obtained by decorating the aminobenzothiazole scaffold with all acyl chlorides and boronates available at the Merck database. The scientific multidisciplinary approach included virtual screening workflow, synthesis, and biological evaluation and led to the identification of three novel aminobenzothiazoles 1, 3, and 13 acting as mPGES-1 inhibitors. The three disclosed hits are able to inhibit mPGES-1 in a cell-free system (IC50 = 1.4 ± 0.2, 0.7 ± 0.1, and 1.7 ± 0.2 μM, respectively), and all are endowed with antitumoral properties against A549 human cancer cell lines at micromolar concentrations (28.5 ± 1.1, 18.1 ± 0.8, and 19.2 ± 1.3 μM, respectively).
Collapse
Affiliation(s)
- Maria G. Chini
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, Pesche, Isernia, I-86090, Italy
| | - Assunta Giordano
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
- Institute of Biomolecular Chemistry (ICB), Consiglio Nazionale delle Ricerche (CNR), Via Campi Flegrei 34, I-80078, Pozzuoli, Napoli, Italy
| | - Marianna Potenza
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| | - Stefania Terracciano
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| | - Katrin Fischer
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Maria C. Vaccaro
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| | - Ester Colarusso
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| | - Ines Bruno
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| | - Raffaele Riccio
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| | - Andreas Koeberle
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
- Michael Popp Research Institute, University of Innsbruck, Innsbruck, Austria
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| |
Collapse
|
22
|
Bülbül B, Küçükgüzel İ. Microsomal Prostaglandin E2 Synthase-1 as a New Macromolecular Drug Target in the Prevention of Inflammation and Cancer. Anticancer Agents Med Chem 2020; 19:1205-1222. [PMID: 30827263 DOI: 10.2174/1871520619666190227174137] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/29/2019] [Accepted: 02/05/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cancer is one of the most life-threatening diseases worldwide. Since inflammation is considered to be one of the known characteristics of cancer, the activity of PGE2 has been paired with different tumorigenic steps such as increased tumor cell proliferation, resistance to apoptosis, increased invasiveness, angiogenesis and immunosuppression. OBJECTIVE It has been successfully demonstrated that inhibition of mPGES-1 prevented inflammation in preclinical studies. However, despite the crucial roles of mPGEs-1 and PGE2 in tumorigenesis, there is not much in vivo study on mPGES-1 inhibition in cancer therapy. The specificity of mPGEs-1 enzyme and its low expression level under normal conditions makes it a promising drug target with a low risk of side effects. METHODS A comprehensive literature search was performed for writing this review. An updated view on PGE2 biosynthesis, PGES isoenzyme family and its pharmacology and the latest information about inhibitors of mPGES-1 have been discussed. RESULTS In this study, it was aimed to highlight the importance of mPGES-1 and its inhibition in inflammationrelated cancer and other inflammatory conditions. Information about PGE2 biosynthesis, its role in inflammationrelated pathologies were also provided. We kept the noncancer-related inflammatory part short and tried to bring together promising molecules or scaffolds. CONCLUSION The information provided in this review might be useful to researchers in designing novel and potent mPGES-1 inhibitors for the treatment of cancer and inflammation.
Collapse
Affiliation(s)
- Bahadır Bülbül
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| | - İlkay Küçükgüzel
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| |
Collapse
|
23
|
Woolbright BL, Pilbeam CC, Taylor JA. Prostaglandin E2 as a therapeutic target in bladder cancer: From basic science to clinical trials. Prostaglandins Other Lipid Mediat 2020; 148:106409. [PMID: 31931078 DOI: 10.1016/j.prostaglandins.2020.106409] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/02/2019] [Accepted: 12/30/2019] [Indexed: 12/15/2022]
Abstract
Bladder cancer (BCa) is a common solid tumor marked by high rates of recurrence, especially in non-muscle invasive disease. Prostaglandin E2 (PGE2) is a ubiquitously present lipid mediator responsible for numerous physiological actions. Inhibition of cyclooxygenase (COX) enzymes by the non-steroidal anti-inflammatory (NSAID) class of drugs results in reduced PGE2 levels. NSAID usage has been associated with reductions in cancers such as BCa. Clinical trials using NSAIDs to prevent recurrence have had mixed results, but largely converge on issues with cardiotoxicity. The purpose of this review is to understand the basic science behind how and why inhibitors of PGE2 may be effective against BCa, and to explore alternate therapeutic modalities for addressing the role of PGE2 without the associated cardiotoxicity. We will address the role of PGE2 in a diverse array of cancer-related functions including stemness, immunosuppression, proliferation, cellular signaling and more.
Collapse
Affiliation(s)
| | - Carol C Pilbeam
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - John A Taylor
- Department of Urology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
24
|
Crispen PL, Kusmartsev S. Mechanisms of immune evasion in bladder cancer. Cancer Immunol Immunother 2019; 69:3-14. [PMID: 31811337 PMCID: PMC6949323 DOI: 10.1007/s00262-019-02443-4] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 11/27/2019] [Indexed: 12/16/2022]
Abstract
With the introduction of multiple new agents, the role of immunotherapy is rapidly expanding across all malignancies. Bladder cancer is known to be immunogenic and is responsive to immunotherapy including intravesical BCG and immune checkpoint inhibitors. Multiple trials have addressed the role of checkpoint inhibitors in advanced bladder cancer, including atezolizumab, avelumab, durvalumab, nivolumab and pembrolizumab (all targeting the PD1/PD-L1 pathway). While these trials have demonstrated promising results and improvements over existing therapies, less than half of patients with advanced disease demonstrate clinical benefit from checkpoint inhibitor therapy. Recent breakthroughs in cancer biology and immunology have led to an improved understanding of the influence of the tumor microenvironment on the host’s immune system. It appears that tumors promote the formation of highly immunosuppressive microenvironments preventing generation of effective anti-tumor immune response through multiple mechanisms. Therefore, reconditioning of the tumor microenvironment and restoration of the competent immune response is essential for achieving optimal efficacy of cancer immunotherapy. In this review, we aim to discuss the major mechanisms of immune evasion in bladder cancer and highlight novel pathways and molecular targets that may help to attenuate tumor-induced immune tolerance, overcome resistance to immunotherapy and improve clinical outcomes.
Collapse
Affiliation(s)
- Paul L Crispen
- Department of Urology, University of Florida, College of Medicine, 1200 Newell Dr, PO BOX 100247, Gainesville, FL, 32610, USA
| | - Sergei Kusmartsev
- Department of Urology, University of Florida, College of Medicine, 1200 Newell Dr, PO BOX 100247, Gainesville, FL, 32610, USA.
| |
Collapse
|
25
|
A review on mPGES-1 inhibitors: From preclinical studies to clinical applications. Prostaglandins Other Lipid Mediat 2019; 147:106383. [PMID: 31698145 DOI: 10.1016/j.prostaglandins.2019.106383] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/16/2019] [Accepted: 09/09/2019] [Indexed: 02/06/2023]
Abstract
Prostaglandin E2 (PGE2) is a lipid mediator of inflammation and cancer progression. It is mainly formed via metabolism of arachidonic acid by cyclooxygenases (COX) and the terminal enzyme microsomal prostaglandin E synthase-1 (mPGES-1). Widely used non-steroidal anti-inflammatory drugs (NSAIDs) inhibit COX activity, resulting in decreased PGE2 production and symptomatic relief. However, NSAIDs block the production of many other lipid mediators that have important physiological and resolving actions, and these drugs cause gastrointestinal bleeding and/or increase the risk for severe cardiovascular events. Selective inhibition of downstream mPGES-1 for reduction in only PGE2 biosynthesis is suggested as a safer therapeutic strategy. This review covers the recent advances in characterization of new mPGES-1 inhibitors in preclinical models and their future clinical applications.
Collapse
|
26
|
Zhou S, Zhou Z, Ding K, Yuan Y, Zheng F, Zhan CG. In Silico Observation of the Conformational Opening of the Glutathione-Binding Site of Microsomal Prostaglandin E2 Synthase-1. J Chem Inf Model 2019; 59:3839-3845. [DOI: 10.1021/acs.jcim.9b00289] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
27
|
Saul MJ, Emmerich AC, Steinhilber D, Suess B. Regulation of Eicosanoid Pathways by MicroRNAs. Front Pharmacol 2019; 10:824. [PMID: 31379585 PMCID: PMC6659501 DOI: 10.3389/fphar.2019.00824] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/26/2019] [Indexed: 01/07/2023] Open
Abstract
Over the last years, many microRNAs (miRNAs) have been identified that regulate the formation of bioactive lipid mediators such as prostanoids and leukotrienes. Many of these miRNAs are involved in complex regulatory circuits necessary for the fine-tuning of biological functions including inflammatory processes or cell growth. A better understanding of these networks will contribute to the development of novel therapeutic strategies for the treatment of inflammatory diseases and cancer. In this review, we provide an overview of the current knowledge of miRNA regulation in eicosanoid pathways with special focus on novel miRNA functions and regulatory circuits of leukotriene and prostaglandin biosynthesis.
Collapse
Affiliation(s)
- Meike J Saul
- Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Anne C Emmerich
- Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany.,Institute of Pharmaceutical Chemistry, Goethe Universität Frankfurt, Frankfurt, Germany
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry, Goethe Universität Frankfurt, Frankfurt, Germany
| | - Beatrix Suess
- Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| |
Collapse
|
28
|
Terzuoli E, Costanza F, Ciccone V, Ziche M, Morbidelli L, Donnini S. mPGES-1 as a new target to overcome acquired resistance to gefitinib in non-small cell lung cancer cell lines. Prostaglandins Other Lipid Mediat 2019; 143:106344. [PMID: 31207300 DOI: 10.1016/j.prostaglandins.2019.106344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/18/2019] [Accepted: 06/12/2019] [Indexed: 02/07/2023]
Abstract
Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) as gefitinib are standard treatment of non-small cell lung cancer (NSCLC), but resistance often occurs. This study demonstrates that NSCLC cells resistant to gefitinib (GR cells) displayed a significantly higher microsomal prostaglandin E synthase-1 (mPGES-1) expression and activity than parental cells. Overexpression of mPGES-1/prostaglandin E-2 (PGE-2) signaling in GR cells was associated with acquisition of mesenchymal and stem-like cell properties, nuclear EGFR translocation and tolerance to cisplatin. mPGES-1 inhibition reduced mesenchymal and stem-like properties, and nuclear EGFR translocation in GR cells. Consistently, inhibition of mPGES-1 activity enhanced sensitivity to cisplatin and responsiveness to gefitinib in GR cells. We propose the mPGES-1/PGE-2 signaling as a potential target for treating aggressive and resistant lung cancers.
Collapse
Affiliation(s)
- Erika Terzuoli
- Department of Medical and Surgical Sciences and Neurosciences, University of Siena, Via A. Moro, 2, 53100, Siena, Italy.
| | - Filomena Costanza
- Department of Life Sciences, University of Siena, Via A. Moro, 2, 53100, Siena, Italy.
| | - Valerio Ciccone
- Department of Life Sciences, University of Siena, Via A. Moro, 2, 53100, Siena, Italy.
| | - Marina Ziche
- Department of Medical and Surgical Sciences and Neurosciences, University of Siena, Via A. Moro, 2, 53100, Siena, Italy.
| | - Lucia Morbidelli
- Department of Life Sciences, University of Siena, Via A. Moro, 2, 53100, Siena, Italy.
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, Via A. Moro, 2, 53100, Siena, Italy.
| |
Collapse
|
29
|
Bergqvist F, Ossipova E, Idborg H, Raouf J, Checa A, Englund K, Englund P, Khoonsari PE, Kultima K, Wheelock CE, Larsson K, Korotkova M, Jakobsson PJ. Inhibition of mPGES-1 or COX-2 Results in Different Proteomic and Lipidomic Profiles in A549 Lung Cancer Cells. Front Pharmacol 2019; 10:636. [PMID: 31231223 PMCID: PMC6567928 DOI: 10.3389/fphar.2019.00636] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/17/2019] [Indexed: 12/23/2022] Open
Abstract
Pharmacological inhibition of microsomal prostaglandin E synthase (mPGES)-1 for selective reduction in prostaglandin E2 (PGE2) biosynthesis is protective in experimental models of cancer and inflammation. Targeting mPGES-1 is envisioned as a safer alternative to traditional non-steroidal anti-inflammatory drugs (NSAIDs). Herein, we compared the effects of mPGES-1 inhibitor Compound III (CIII) with the cyclooxygenase (COX)-2 inhibitor NS-398 on protein and lipid profiles in interleukin (IL)-1β-induced A549 lung cancer cells using mass spectrometry. Inhibition of mPGES-1 decreased PGE2 production and increased PGF2α and thromboxane B2 (TXB2) formation, while inhibition of COX-2 decreased the production of all three prostanoids. Our proteomics results revealed that CIII downregulated multiple canonical pathways including eIF2, eIF4/P70S6K, and mTOR signaling, compared to NS-398 that activated these pathways. Moreover, pathway analysis predicted that CIII increased cell death of cancer cells (Z = 3.8, p = 5.1E-41) while NS-398 decreased the same function (Z = -5.0, p = 6.5E-35). In our lipidomics analyses, we found alterations in nine phospholipids between the two inhibitors, with a stronger alteration in the lysophospholipid (LPC) profile with NS-398 compared to CIII. Inhibition of mPGES-1 increased the concentration of sphinganine and dihydroceramide (C16:0DhCer), while inhibition of COX-2 caused a general decrease in most ceramides, again suggesting different effects on cell death between the two inhibitors. We showed that CIII decreased proliferation and potentiated the cytotoxic effect of the cytostatic drugs cisplatin, etoposide, and vincristine when investigated in a live cell imaging system. Our results demonstrate differences in protein and lipid profiles after inhibition of mPGES-1 or COX-2 with important implications on the therapeutic potential of mPGES-1 inhibitors as adjuvant treatment in cancer. We encourage further investigations to illuminate the clinical benefit of mPGES-1 inhibitors in cancer.
Collapse
Affiliation(s)
- Filip Bergqvist
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Elena Ossipova
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Helena Idborg
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Joan Raouf
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Antonio Checa
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Karin Englund
- Department of Analytical Chemistry, Stockholm University, Stockholm, Sweden
| | - Petter Englund
- Department of Analytical Chemistry, Stockholm University, Stockholm, Sweden
| | - Payam Emami Khoonsari
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Kim Kultima
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Craig E Wheelock
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Karin Larsson
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Marina Korotkova
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Per-Johan Jakobsson
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| |
Collapse
|
30
|
Huang H, Aladelokun O, Ideta T, Giardina C, Ellis LM, Rosenberg DW. Inhibition of PGE 2/EP4 receptor signaling enhances oxaliplatin efficacy in resistant colon cancer cells through modulation of oxidative stress. Sci Rep 2019; 9:4954. [PMID: 30894570 PMCID: PMC6427013 DOI: 10.1038/s41598-019-40848-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 02/22/2019] [Indexed: 12/15/2022] Open
Abstract
The platinum-based chemotherapeutic agent, oxaliplatin, is used to treat advanced colorectal cancer (CRC). Unfortunately, nearly all patients acquire resistance to oxaliplatin after long-term use, limiting its therapeutic efficacy. Since COX-2 and PGE2 signaling can impact colon cancer cell proliferation and survival, we examined how this pathway was affected in an oxaliplatin resistant colon cancer cell line. PGE2 levels were significantly elevated in oxaliplatin-resistant HT29 cells (OXR) compared to naïve parental HT29 cells (PAR). This increase was associated with elevated COX-2 (17.9-fold; P = 0.008) and reduced 15-hydroxyprostaglandin dehydrogenase (2.9-fold; P < 0.0001) expression. RNAi knockdown of microsomal prostaglandin E synthase-1, the rate-limiting enzyme in PGE2 synthesis, sensitized OXR cells to oxaliplatin. Downstream effects of PGE2 in OXR cells were also examined. Selective inhibition of the EP4 PGE2 receptor by the small molecule inhibitor, L-161,982 enhanced oxaliplatin-induced apoptosis in OXR cells. L-161,982 also reduced expression of the colonic stem cell markers, CD133 and CD44, and inhibited tumor sphere formation. The accumulation of intracellular reactive oxygen species (ROS), a key component of oxaliplatin cytotoxicity, was significantly increased by EP4 inhibition (2.4 -fold; P < 0.0001). Overall, our findings uncover an important role for the COX-2/PGE2/EP4 signaling axis in oxaliplatin resistance via regulation of oxidative stress.
Collapse
Affiliation(s)
- Huakang Huang
- Center for Molecular Oncology, University of Connecticut Health, 263 Farmington Ave, Farmington, CT, USA
| | - Oladimeji Aladelokun
- Center for Molecular Oncology, University of Connecticut Health, 263 Farmington Ave, Farmington, CT, USA
| | - Takayasu Ideta
- Center for Molecular Oncology, University of Connecticut Health, 263 Farmington Ave, Farmington, CT, USA
| | - Charles Giardina
- Department of Cell and Molecular Biology, University of Connecticut, Storrs, CT, USA
| | - Lee M Ellis
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Holcombe Boulevard, Houston, Texas, USA
| | - Daniel W Rosenberg
- Center for Molecular Oncology, University of Connecticut Health, 263 Farmington Ave, Farmington, CT, USA.
| |
Collapse
|
31
|
Lukic A, Wahlund CJ, Gómez C, Brodin D, Samuelsson B, Wheelock CE, Gabrielsson S, Rådmark O. Exosomes and cells from lung cancer pleural exudates transform LTC4 to LTD4, promoting cell migration and survival via CysLT1. Cancer Lett 2019; 444:1-8. [DOI: 10.1016/j.canlet.2018.11.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 02/06/2023]
|
32
|
Sala A, Proschak E, Steinhilber D, Rovati GE. Two-pronged approach to anti-inflammatory therapy through the modulation of the arachidonic acid cascade. Biochem Pharmacol 2018; 158:161-173. [DOI: 10.1016/j.bcp.2018.10.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/09/2018] [Indexed: 12/11/2022]
|
33
|
Madrigal‐Martínez A, Constâncio V, Lucio‐Cazaña FJ, Fernández‐Martínez AB. PROSTAGLANDIN E
2
stimulates cancer‐related phenotypes in prostate cancer PC3 cells through cyclooxygenase‐2. J Cell Physiol 2018; 234:7548-7559. [DOI: 10.1002/jcp.27515] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 09/10/2018] [Indexed: 12/15/2022]
Affiliation(s)
| | - Vera Constâncio
- Departamento de Biología de Sistemas Universidad de Alcalá Madrid Spain
| | | | | |
Collapse
|
34
|
Tian Y, Yang T, Yu S, Liu C, He M, Hu C. Prostaglandin E2 increases migration and proliferation of human glioblastoma cells by activating transient receptor potential melastatin 7 channels. J Cell Mol Med 2018; 22:6327-6337. [PMID: 30338939 PMCID: PMC6237613 DOI: 10.1111/jcmm.13931] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 08/30/2018] [Indexed: 12/26/2022] Open
Abstract
Recent studies showed that both prostaglandin E2 (PGE2) and transient receptor potential melastatin 7 (TRPM7) play important roles in migration and proliferation of human glioblastoma cells. In this study, we tested the association between PGE2 and TRPM7. We found that PGE2 increased TRPM7 currents in HEK293 and human glioblastoma A172 cells. The PGE2 EP3 receptor antagonist L‐798106 abrogated the PGE2 stimulatory effect, while EP3 agonist 17‐phenyl trinor prostaglandin E2 (17‐pt‐PGE2) mimicked the effect of PEG2 on TRPM7. The TRPM7 phosphotransferase activity‐deficient mutation, K1646R had no effect on PGE2 induced increase of TRPM7 currents. Inhibition of protein kinase A (PKA) activity by Rp‐cAMP increased TRPM7 currents. TRPM7 PKA phosphorylation site mutation S1269A abolished the PGE2 effect on TRPM7 currents. PGE2 increased both mRNA and membrane protein expression of TRPM7 in A172 cells. Knockdown of TRPM7 by shRNA abrogated the PGE2 stimulated migration and proliferation of A172 cells. Blockage of TRPM7 with 2‐aminoethoxydiphenyl borate (2‐APB) or NS8593 had a similar effect as TRPM7‐shRNA. In conclusion, our results demonstrate that PGE2 activates TRPM7 via EP3/PKA signalling pathway, and that PGE2 enhances migration and proliferation of human glioblastoma cells by up‐regulation of the TRPM7 channel.
Collapse
Affiliation(s)
- Yafei Tian
- Department of Physiology and Biophysics, School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Tingting Yang
- Department of Physiology and Biophysics, School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Shuntai Yu
- Department of Physiology and Biophysics, School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Cuiyun Liu
- Department of Physiology and Biophysics, School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Min He
- Department of Physiology and Biophysics, School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Changlong Hu
- Department of Physiology and Biophysics, School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Garg R, Blando JM, Perez CJ, Lal P, Feldman MD, Smyth EM, Ricciotti E, Grosser T, Benavides F, Kazanietz MG. COX-2 mediates pro-tumorigenic effects of PKCε in prostate cancer. Oncogene 2018; 37:4735-4749. [PMID: 29765153 PMCID: PMC6195867 DOI: 10.1038/s41388-018-0318-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/22/2018] [Accepted: 04/20/2018] [Indexed: 12/31/2022]
Abstract
The pro-oncogenic kinase PKCε is overexpressed in human prostate cancer and cooperates with loss of the tumor suppressor Pten for the development of prostatic adenocarcinoma. However, the effectors driving PKCε-mediated phenotypes remain poorly defined. Here, using cellular and mouse models, we showed that PKCε overexpression acts synergistically with Pten loss to promote NF-κB activation and induce cyclooxygenase-2 (COX-2) expression, phenotypic traits which are also observed in human prostate tumors. Targeted disruption of PKCε from prostate cancer cells impaired COX-2 induction and PGE2 production. Notably, COX-2 inhibitors selectively killed prostate epithelial cells overexpressing PKCε, and this ability was greatly enhanced by Pten loss. Long-term COX-2 inhibition markedly reduced adenocarcinoma formation, as well as angiogenesis in a mouse model of prostate-specific PKCε expression and Pten loss. Overall, our results provide strong evidence for the involvement of the canonical NF-κB pathway and its target gene COX2 as PKCε effectors, and highlight the potential of PKCε as a useful biomarker for the use of COX inhibition for chemopreventive and/or chemotherapeutic purposes in prostate cancer.
Collapse
Affiliation(s)
- Rachana Garg
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jorge M Blando
- Department of Immunology, Immunopathology Laboratory, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Carlos J Perez
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX, 78957, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Priti Lal
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael D Feldman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Emer M Smyth
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Emanuela Ricciotti
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Tilo Grosser
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Fernando Benavides
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX, 78957, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Marcelo G Kazanietz
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
36
|
Gupta A, Chaudhary N, Aparoy P. MM-PBSA and per-residue decomposition energy studies on 7-Phenyl-imidazoquinolin-4(5H)-one derivatives: Identification of crucial site points at microsomal prostaglandin E synthase-1 (mPGES-1) active site. Int J Biol Macromol 2018; 119:352-359. [PMID: 30031079 DOI: 10.1016/j.ijbiomac.2018.07.050] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 12/29/2022]
Abstract
The huge therapeutic potential and the market share of painkillers are well-known. Due to the side effects associated with traditional NSAIDs and selective cyclooxygenase (COX-2) inhibitors, a new generation of painkillers is the need of the hour. In this regard, microsomal prostaglandin E synthase-1 (mPGES-1) offers great potential as an alternative drug target against inflammatory disorders. The present study is aimed at identifying the amino acids crucial in effective inhibitor binding at the mPGES-1 active site by performing molecular dynamics based studies on a series of 7-Phenyl-imidazoquinolin-4(5H)-one derivatives. Molecular dynamics (MD) simulations, MM-PBSA, per-residue energy decomposition and Dimensionality Reduction through Covariance matrix Transformation for Identification of Differences in dynamics (DIRECT-ID) analysis were performed to get insights into the structural details that can aid in novel drug design against mPGES-1. The high correlations of electrostatic and polar energy terms with biological activity highlight their importance and applicability in in silico screening studies. Further, per-residue energy decomposition studies revealed that Lys42, Arg52, Arg122, Pro124, Ser127, Val128 and Thr131 were contributing more towards inhibitor binding energy. The results clearly show that MM-PBSA can act as a filter in virtual screening experiments and can play major role in facilitating various mPGES-1 drug discovery studies.
Collapse
Affiliation(s)
- Ashish Gupta
- Centre for Computational Biology and Bioinformatics, School of Life Sciences, Central University of Himachal Pradesh, Dharamshala, Himachal Pradesh 176215, India
| | - Neha Chaudhary
- Centre for Computational Biology and Bioinformatics, School of Life Sciences, Central University of Himachal Pradesh, Dharamshala, Himachal Pradesh 176215, India
| | - Polamarasetty Aparoy
- Centre for Computational Biology and Bioinformatics, School of Life Sciences, Central University of Himachal Pradesh, Dharamshala, Himachal Pradesh 176215, India.
| |
Collapse
|
37
|
Yoo HJ, Kim M, Kim M, Kang M, Jung KJ, Hwang SM, Jee SH, Lee JH. Analysis of metabolites and metabolic pathways in breast cancer in a Korean prospective cohort: the Korean Cancer Prevention Study-II. Metabolomics 2018; 14:85. [PMID: 30830383 DOI: 10.1007/s11306-018-1382-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 06/05/2018] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Since blood is in contact with all tissues in the body and is considered to dynamically reflect the body's pathophysiological status, serum metabolomics changes are important and have diagnostic value in early cancer detection. OBJECTIVES In this prospective study, we investigated the application of metabolomics to differentiate subjects with incident breast cancer (BC) from subjects who remained free of cancer during a mean follow-up period of 7 years with the aim of identifying valuable biomarkers for BC. METHODS Baseline serum samples from 84 female subjects with incident BC (BC group) and 88 cancer-free female subjects (control group) were used. Metabolic alterations associated with BC were investigated via metabolomics analysis of the baseline serum samples using ultra-performance liquid chromatography-linear-trap quadrupole-Orbitrap mass spectrometry. RESULTS A total of 57 metabolites were identified through the metabolic analysis. Among them, 20 metabolite levels were significantly higher and 22 metabolite levels were significantly lower in the BC group than in the control group at baseline. Ten metabolic pathways, including amino acid metabolism, arachidonic acid (AA) metabolism, fatty acid metabolism, linoleic acid metabolism, and retinol metabolism, showed significant differences between the BC group and the control group. Logistic regression revealed that the incidence of BC was affected by leucine, AA, prostaglandin (PG)J2, PGE2, and γ-linolenic acid (GLA). CONCLUSIONS This prospective study showed the clinical relevance of dysregulation of various metabolisms on the incidence of BC. Additionally, leucine, AA, PGJ2, PGE2, and GLA were identified as independent variables affecting the incidence of BC.
Collapse
Affiliation(s)
- Hye Jin Yoo
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, South Korea
| | - Minjoo Kim
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, South Korea
| | - Minkyung Kim
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, South Korea
| | - Minsik Kang
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, South Korea
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Keum Ji Jung
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, South Korea
| | - Se-Mi Hwang
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, South Korea
| | - Sun Ha Jee
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, South Korea
| | - Jong Ho Lee
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, South Korea.
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, South Korea.
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| |
Collapse
|
38
|
Inhibition of Microsomal Prostaglandin E Synthase-1 in Cancer-Associated Fibroblasts Suppresses Neuroblastoma Tumor Growth. EBioMedicine 2018; 32:84-92. [PMID: 29804818 PMCID: PMC6021299 DOI: 10.1016/j.ebiom.2018.05.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/26/2018] [Accepted: 05/04/2018] [Indexed: 12/20/2022] Open
Abstract
Despite recent progress in diagnosis and treatment, survival for children with high-risk metastatic neuroblastoma is still poor. Prostaglandin E2 (PGE2)-driven inflammation promotes tumor growth, immune suppression, angiogenesis and resistance to established cancer therapies. In neuroblastoma, cancer-associated fibroblasts (CAFs) residing in the tumor microenvironment are the primary source of PGE2. However, clinical targeting of PGE2 with current non-steroidal anti-inflammatory drugs or cyclooxygenase inhibitors has been limited due to risk of adverse side effects. By specifically targeting microsomal prostaglandin E synthase-1 (mPGES-1) activity with a small molecule inhibitor we could block CAF-derived PGE2 production leading to reduced tumor growth, impaired angiogenesis, inhibited CAF migration and infiltration, reduced tumor cell proliferation and a favorable shift in the M1/M2 macrophage ratio. In this study, we provide proof-of-principle of the benefits of targeting mPGES-1 in neuroblastoma, applicable to a wide variety of tumors. This non-toxic single drug treatment targeting infiltrating stromal cells opens up for combination treatment options with established cancer therapies. Prostaglandin E2 nourishes neuroblastoma tumor growth via cancer-associated fibroblasts. mPGES-1 inhibitor limits tumor growth, angiogenesis, infiltration of cancer-associated fibroblasts and immune suppression. mPGES-1 constitutes a drug target for neuroblastoma treatment.
Cancer is the leading cause of death in children in high-income countries and the survival rate has almost been unchanged during the last decade. Further treatment intensification to improve survival rate may further increase the risk of side-effects. Therapies targeting the microenvironment have been suggested to improve survival and quality of life for these children. High-risk neuroblastomas present an immunosuppressive microenvironment and infiltrating cancer-associated fibroblasts are responsible for oncogenic prostaglandin E2 production. Here we show that selective inhibition of prostaglandin E2 biosynthesis and its role in the crosstalk between cells of the microenvironment provides a promising therapeutic strategy in neuroblastoma.
Collapse
|
39
|
Structure-based discovery of mPGES-1 inhibitors suitable for preclinical testing in wild-type mice as a new generation of anti-inflammatory drugs. Sci Rep 2018; 8:5205. [PMID: 29581541 PMCID: PMC5979965 DOI: 10.1038/s41598-018-23482-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/09/2018] [Indexed: 12/24/2022] Open
Abstract
Human mPGES-1 is recognized as a promising target for next generation of anti-inflammatory drugs without the side effects of currently available anti-inflammatory drugs, and various inhibitors have been reported in the literature. However, none of the reported potent inhibitors of human mPGES-1 has shown to be also a potent inhibitor of mouse or rat mPGES-1, which prevents using the well-established mouse/rat models of inflammation-related diseases for preclinical studies. Hence, despite of extensive efforts to design and discover various human mPGES-1 inhibitors, the promise of mPGES-1 as a target for the next generation of anti-inflammatory drugs has never been demonstrated in any wild-type mouse/rat model using an mPGES-1 inhibitor. Here we report discovery of a novel type of selective mPGES-1 inhibitors potent for both human and mouse mPGES-1 enzymes through structure-based rational design. Based on in vivo studies using wild-type mice, the lead compound is indeed non-toxic, orally bioavailable, and more potent in decreasing the PGE2 (an inflammatory marker) levels compared to the currently available drug celecoxib. This is the first demonstration in wild-type mice that mPGES-1 is truly a promising target for the next generation of anti-inflammatory drugs.
Collapse
|
40
|
Structure-based discovery of mPGES-1 inhibitors suitable for preclinical testing in wild-type mice as a new generation of anti-inflammatory drugs. Sci Rep 2018. [PMID: 29581541 DOI: 10.1038/s41598-41018-23482-41594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023] Open
Abstract
Human mPGES-1 is recognized as a promising target for next generation of anti-inflammatory drugs without the side effects of currently available anti-inflammatory drugs, and various inhibitors have been reported in the literature. However, none of the reported potent inhibitors of human mPGES-1 has shown to be also a potent inhibitor of mouse or rat mPGES-1, which prevents using the well-established mouse/rat models of inflammation-related diseases for preclinical studies. Hence, despite of extensive efforts to design and discover various human mPGES-1 inhibitors, the promise of mPGES-1 as a target for the next generation of anti-inflammatory drugs has never been demonstrated in any wild-type mouse/rat model using an mPGES-1 inhibitor. Here we report discovery of a novel type of selective mPGES-1 inhibitors potent for both human and mouse mPGES-1 enzymes through structure-based rational design. Based on in vivo studies using wild-type mice, the lead compound is indeed non-toxic, orally bioavailable, and more potent in decreasing the PGE2 (an inflammatory marker) levels compared to the currently available drug celecoxib. This is the first demonstration in wild-type mice that mPGES-1 is truly a promising target for the next generation of anti-inflammatory drugs.
Collapse
|
41
|
Terzuoli E, Donnini S, Finetti F, Nesi G, Villari D, Hanaka H, Radmark O, Giachetti A, Ziche M. Linking microsomal prostaglandin E Synthase-1/PGE-2 pathway with miR-15a and -186 expression: Novel mechanism of VEGF modulation in prostate cancer. Oncotarget 2018; 7:44350-44364. [PMID: 27322147 PMCID: PMC5190102 DOI: 10.18632/oncotarget.10051] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 06/01/2016] [Indexed: 12/29/2022] Open
Abstract
Prostaglandin E-2 (PGE-2) promotes tumor angiogenesis via paracrine secretion of pro-angiogenic growth factors, such as vascular endothelial growth factor (VEGF). Since miRNAs regulate several cell processes, including angiogenesis, we sought to determine whether they would influence PGE-2-induced VEGF. We compared DU145 and PC3 prostate cancer cells bearing the mPGES-1 enzyme (mPGES-1+/+) and producing PGE-2, with those in which the enzyme was silenced or deleted (mPGES-1-/-). We demonstrated that mPGES-1/PGE-2 signaling decreased Dicer expression and miRNA biogenesis. Genome-wide sequencing of miRNAs revealed that miR-15a and miR-186, associated with expression of VEGF and hypoxia inducible factor-1α (HIF-1α), were down-regulated in mPGES-1+/+ cells. As a consequence, mPGES-1+/+ tumor cells expressed high levels of VEGF and HIF-1α, induced endothelial cells activation and formed highly vascularized tumors. Mir-186 mimic inhibited VEGF expression in mPGES-1+/+ tumor xenografts and reduced tumor growth. In human prostate cancer specimens, mPGES-1 was over-expressed in tumors with high Gleason score, elevated expression of VEGF and HIF-1α, high microvessel density and decreased expression of Dicer, miR15a and miR-186. Thus, clear evidence for regulating miRNA processing and VEGF output by intrinsic PGE-2 production provides a means to distinguish between aggressive and indolent prostate tumors and suggests a potential target for controlling tumor progression.
Collapse
Affiliation(s)
- Erika Terzuoli
- Department of Life Sciences, University of Siena, 53100, Siena, Italy
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, 53100, Siena, Italy.,Istituto Toscano Tumori (ITT), 50136, Florence, Italy
| | - Federica Finetti
- Department of Life Sciences, University of Siena, 53100, Siena, Italy
| | - Gabriella Nesi
- Department of Surgery and Translational Medicine, University of Florence, 50136, Florence, Italy
| | - Donata Villari
- Department of Clinical and Experimental Medicine, University of Florence, 50136, Florence, Italy
| | - Hiromi Hanaka
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, S-171 77, Stockholm, Sweden
| | - Olof Radmark
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, S-171 77, Stockholm, Sweden
| | - Antonio Giachetti
- Department of Life Sciences, University of Siena, 53100, Siena, Italy
| | - Marina Ziche
- Department of Life Sciences, University of Siena, 53100, Siena, Italy.,Istituto Toscano Tumori (ITT), 50136, Florence, Italy
| |
Collapse
|
42
|
Alves Rico SR, Abbasi AZ, Ribeiro G, Ahmed T, Wu XY, de Oliveira Silva D. Diruthenium(ii,iii) metallodrugs of ibuprofen and naproxen encapsulated in intravenously injectable polymer-lipid nanoparticles exhibit enhanced activity against breast and prostate cancer cells. NANOSCALE 2017; 9:10701-10714. [PMID: 28678269 DOI: 10.1039/c7nr01582h] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
A unique class of diruthenium(ii,iii) metallodrugs containing non-steroidal anti-inflammatory drug (NSAID), Ru2(NSAID), have been reported to show anticancer activity in glioma models in vitro and in vivo. This work reports the encapsulation of the lead metallodrug of ibuprofen (HIbp), [Ru2(Ibp)4Cl] or RuIbp, and also of the new analogue of naproxen (HNpx), [Ru2(Npx)4Cl] or RuNpx, in novel intravenously (i.v.) injectable solid polymer-lipid nanoparticles (SPLNs). A rationally selected composition of lipids/polymers rendered nearly spherical Ru2(NSAID)-SPLNs with a mean size of 120 nm and zeta potential of about -20 mV. The Ru2(NSAID)-SPLNs are characterized by spectroscopic techniques and the composition in terms of ruthenium-drug species is analyzed by mass spectrometry. The metallodrug-loaded nanoparticles showed high drug loading (17-18%) with ∼100% drug loading efficiency, and good colloidal stability in serum at body temperature. Fluorescence-labeled SPLNs were taken up by the cancer cells in a time- and energy-dependent manner as analyzed by confocal microscopy and fluorescence spectrometry. The Ru2(NSAID)-SPLNs showed enhanced cytotoxicity (IC50 at 60-100 μmol L-1 ) in relation to the corresponding Ru2(NSAID) metallodrugs in breast (EMT6 and MDA-MB-231) and prostate (DU145) cancer cells in vitro. The cell viability of both metallodrug nanoformulations is also compared with those of the parent NSAIDs, HIbp and HNpx, and their corresponding NSAID-SPLNs. In vivo and ex vivo fluorescence imaging revealed good biodistribution and high tumor accumulation of fluorescence-labeled SPLNs following i.v. injection in an orthotopic breast tumor model. The enhanced anticancer activity of the metallodrug-loaded SPLNs in these cell lines can be associated with the advantages of the nanoformulations, assigned mainly to the stability of the colloidal nanoparticles suitable for i.v. injection and enhanced cellular uptake. The findings of this work encourage future in vivo efficacy studies to further exploit the potential of the novel Ru2(NSAID)-SPLN nanoformulations for clinical application.
Collapse
Affiliation(s)
- Samara R Alves Rico
- Laboratory for Synthetic and Structural Inorganic Chemistry - Bioinorganic and Metallodrugs, Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes, 748, B2 T, 05508-000, São Paulo, SP, Brazil.
| | | | | | | | | | | |
Collapse
|
43
|
Bugde P, Biswas R, Merien F, Lu J, Liu DX, Chen M, Zhou S, Li Y. The therapeutic potential of targeting ABC transporters to combat multi-drug resistance. Expert Opin Ther Targets 2017; 21:511-530. [DOI: 10.1080/14728222.2017.1310841] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Piyush Bugde
- School of Science, Auckland University of Technology, Auckland, New Zealand
| | - Riya Biswas
- School of Science, Auckland University of Technology, Auckland, New Zealand
| | - Fabrice Merien
- School of Science, Auckland University of Technology, Auckland, New Zealand
- School of Science, AUT Roche Diagnostic Laboratory, Auckland University of Technology, Auckland, New Zealand
| | - Jun Lu
- School of Science, Auckland University of Technology, Auckland, New Zealand
- School of Interprofessional Health Studies, Auckland University of Technology, Auckland, New Zealand
| | - Dong-Xu Liu
- School of Science, Auckland University of Technology, Auckland, New Zealand
| | - Mingwei Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Shufeng Zhou
- Department of Biotechnology and Bioengineering, College of Chemical Engineering, Huaqiao University, Xiamen, China
| | - Yan Li
- School of Science, Auckland University of Technology, Auckland, New Zealand
- School of Interprofessional Health Studies, Auckland University of Technology, Auckland, New Zealand
| |
Collapse
|
44
|
Terzuoli E, Finetti F, Costanza F, Giachetti A, Ziche M, Donnini S. Linking of mPGES-1 and iNOS activates stem-like phenotype in EGFR-driven epithelial tumor cells. Nitric Oxide 2017; 66:17-29. [PMID: 28257996 DOI: 10.1016/j.niox.2017.02.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 02/21/2017] [Accepted: 02/27/2017] [Indexed: 12/19/2022]
Abstract
Inflammatory prostaglandin E-2 (PGE-2) favors cancer progression in epithelial tumors characterized by persistent oncogene input. However, its effects on tumor cell stemness are poorly understood at molecular level. Here we describe two epithelial tumor cells A431 and A459, originating from human lung and skin tumors, in which epithelial growth factor (EGF) induces sequential up-regulation of mPGES-1 and iNOS enzymes, producing an inflammatory intracellular milieu. We demonstrated that concerted action of EGF, mPGES-1 and iNOS causes sharp changes in cell phenotype demonstrated by acquisition of stem-cell features and activation of the epithelial-mesenchymal transition (EMT). When primed with EGF, epithelial tumor cells transfected with mPGES-1 or iNOS to ensure steady enzyme levels display major stem-like and EMT markers, such as reduction in E-cadherin with a concomitant rise in vimentin, ALDH-1, CD133 and ALDH activity. Tumorsphere studies with these cells show increased sphere number and size, enhanced migratory and clonogenic capacity and sharp changes in EMT markers, indicating activation of this process. The concerted action of the enzymes forms a well-orchestrated cascade where expression of iNOS depends on overexpression of mPGES-1. Indeed, we show that through its downstream effectors (PGE-2, PKA, PI3K/Akt), mPGES-1 recruits non-canonical transcription factors, thus facilitating iNOS production. In conclusion, we propose that the initial event leading to tumor stem-cell activation may be a leveraged intrinsic mechanism in which all players are either inherent constituents (EGF) or highly inducible proteins (mPGES-1, iNOS) of tumor cells. We suggest that incipient tumor aggressiveness may be moderated by reducing pivotal input of mPGES-1.
Collapse
Affiliation(s)
- Erika Terzuoli
- Department of Life Sciences, University of Siena, 53100 Siena, Italy.
| | - Federica Finetti
- Department of Life Sciences, University of Siena, 53100 Siena, Italy.
| | - Filomena Costanza
- Department of Life Sciences, University of Siena, 53100 Siena, Italy.
| | - Antonio Giachetti
- Department of Life Sciences, University of Siena, 53100 Siena, Italy.
| | - Marina Ziche
- Department of Life Sciences, University of Siena, 53100 Siena, Italy; Istituto Toscano Tumori (ITT), 50136 Florence, Italy.
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, 53100 Siena, Italy; Istituto Toscano Tumori (ITT), 50136 Florence, Italy.
| |
Collapse
|
45
|
Dovizio M, Sacco A, Patrignani P. Curbing tumorigenesis and malignant progression through the pharmacological control of the wound healing process. Vascul Pharmacol 2017; 89:1-11. [PMID: 28089842 DOI: 10.1016/j.vph.2017.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 12/21/2016] [Accepted: 01/09/2017] [Indexed: 01/13/2023]
Abstract
The prevention of cancer development and its progression is an urgent unmet medical need. Novel knowledge on the biology of cancer has evidenced that genetic changes occurring within cancer cells contribute, but are not sufficient, for tumor promotion and progression. The results of clinical studies and experimental animal models have suggested pursuing new avenues for the prevention of cancer development in the early stages, by using drugs that modulate platelet responses and those interfering with the synthesis and action of the mediators of inflammation. In fact, malignant tumors often develop at sites of chronic injury associated with platelet activation and chronic inflammation. In this review, we cover the evidence supporting this hypothesis and the rationale for the pharmacological treatment with antiplatelet agents, including low-dose aspirin, and antiinflammatory drugs to curb tumorigenesis and malignant progression. The evidence for a chemopreventive effect of low-dose aspirin against colorectal cancer (CRC) has been recently found appropriate by the U.S. Preventive Services Task Force, which recommends the use of the drug for primary prevention of cardiovascular disease and CRC.
Collapse
Affiliation(s)
- Melania Dovizio
- Section of Cardiovascular and Pharmacological Sciences, Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy; CeSI-MeT (Centro Scienze dell'Invecchiamento e Medicina Traslazionale), "G. d'Annunzio" University, Chieti, Italy
| | - Angela Sacco
- Section of Cardiovascular and Pharmacological Sciences, Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy; CeSI-MeT (Centro Scienze dell'Invecchiamento e Medicina Traslazionale), "G. d'Annunzio" University, Chieti, Italy
| | - Paola Patrignani
- Section of Cardiovascular and Pharmacological Sciences, Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy; CeSI-MeT (Centro Scienze dell'Invecchiamento e Medicina Traslazionale), "G. d'Annunzio" University, Chieti, Italy.
| |
Collapse
|
46
|
Gupta A, Aparoy P. Insights into the structure activity relationship of mPGES-1 inhibitors: Hints for better inhibitor design. Int J Biol Macromol 2016; 88:624-32. [DOI: 10.1016/j.ijbiomac.2016.03.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 03/19/2016] [Accepted: 03/19/2016] [Indexed: 10/22/2022]
|
47
|
Inada M, Takita M, Yokoyama S, Watanabe K, Tominari T, Matsumoto C, Hirata M, Maru Y, Maruyama T, Sugimoto Y, Narumiya S, Uematsu S, Akira S, Murphy G, Nagase H, Miyaura C. Direct Melanoma Cell Contact Induces Stromal Cell Autocrine Prostaglandin E2-EP4 Receptor Signaling That Drives Tumor Growth, Angiogenesis, and Metastasis. J Biol Chem 2015; 290:29781-93. [PMID: 26475855 DOI: 10.1074/jbc.m115.669481] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Indexed: 01/11/2023] Open
Abstract
The stromal cells associated with tumors such as melanoma are significant determinants of tumor growth and metastasis. Using membrane-bound prostaglandin E synthase 1 (mPges1(-/-)) mice, we show that prostaglandin E2 (PGE2) production by host tissues is critical for B16 melanoma growth, angiogenesis, and metastasis to both bone and soft tissues. Concomitant studies in vitro showed that PGE2 production by fibroblasts is regulated by direct interaction with B16 cells. Autocrine activity of PGE2 further regulates the production of angiogenic factors by fibroblasts, which are key to the vascularization of both primary and metastatic tumor growth. Similarly, cell-cell interactions between B16 cells and host osteoblasts modulate mPGES-1 activity and PGE2 production by the osteoblasts. PGE2, in turn, acts to stimulate receptor activator of NF-κB ligand expression, leading to osteoclast differentiation and bone erosion. Using eicosanoid receptor antagonists, we show that PGE2 acts on osteoblasts and fibroblasts in the tumor microenvironment through the EP4 receptor. Metastatic tumor growth and vascularization in soft tissues was abrogated by an EP4 receptor antagonist. EP4-null Ptger4(-/-) mice do not support B16 melanoma growth. In vitro, an EP4 receptor antagonist modulated PGE2 effects on fibroblast production of angiogenic factors. Our data show that B16 melanoma cells directly influence host stromal cells to generate PGE2 signals governing neoangiogenesis and metastatic growth in bone via osteoclast erosive activity as well as angiogenesis in soft tissue tumors.
Collapse
Affiliation(s)
- Masaki Inada
- From the Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan, the Global Innovation Research Organization, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan
| | - Morichika Takita
- From the Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan, the Department of Pharmacology, Tokyo Women's Medical University, Tokyo 162-8666, Japan
| | - Satoshi Yokoyama
- From the Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan
| | - Kenta Watanabe
- From the Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan
| | - Tsukasa Tominari
- From the Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan, the Global Innovation Research Organization, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan
| | - Chiho Matsumoto
- From the Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan
| | - Michiko Hirata
- From the Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan
| | - Yoshiro Maru
- the Department of Pharmacology, Tokyo Women's Medical University, Tokyo 162-8666, Japan
| | - Takayuki Maruyama
- the Minase Research Institutes, Ono Pharmaceutical Co. Ltd, Osaka 618-8585, Japan
| | - Yukihiko Sugimoto
- the Department of Pharmaceutical Biochemistry, Graduate School of Pharmaceutical Science, Kumamoto University, Kumamoto 862-0973, Japan
| | - Shuh Narumiya
- the Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Satoshi Uematsu
- the Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan, the Department of Mucosal Immunology, School of Medicine, Chiba University, Chiba 260-8670, Japan, the Division of Innate Immune, Regulation, International Research, and Development, Center for Mucosal Vaccines, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Shizuo Akira
- the Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Gillian Murphy
- the Global Innovation Research Organization, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan, the Department of Oncology, University of Cambridge, Cancer Research UK, Cambridge Institute, Li Ka Shing Centre, Cambridge CB2 0RE, United Kingdom, and
| | - Hideaki Nagase
- the Global Innovation Research Organization, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan, the Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, United Kingdom
| | - Chisato Miyaura
- From the Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan, the Global Innovation Research Organization, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan,
| |
Collapse
|
48
|
Finetti F, Terzuoli E, Giachetti A, Santi R, Villari D, Hanaka H, Radmark O, Ziche M, Donnini S. mPGES-1 in prostate cancer controls stemness and amplifies epidermal growth factor receptor-driven oncogenicity. Endocr Relat Cancer 2015; 22:665-78. [PMID: 26113609 PMCID: PMC4526795 DOI: 10.1530/erc-15-0277] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/23/2015] [Indexed: 02/06/2023]
Abstract
There is evidence that an inflammatory microenvironment is associated with the development and progression of prostate cancer (PCa), although the determinants of intrinsic inflammation in PCa cells are not completely understood. Here we investigated whether expression of intrinsic microsomal PGE synthase-1 (mPGES-1) enhanced aggressiveness of PCa cells and might be critical for epidermal growth factor receptor (EGFR)-mediated tumour progression. In PCa, overexpression of EGFR promotes metastatic invasion and correlates with a high Gleason score, while prostaglandin E2 (PGE2) has been reported to modulate oncogenic EGFR-driven oncogenicity. Immunohistochemical studies revealed that mPGES-1 in human prostate tissues is correlated with EGFR expression in advanced tumours. In DU145 and PC-3 cell lines expressing mPGES-1 (mPGES-1(SC) cells), we demonstrate that silencing or 'knock down' of mPGES-1 (mPGES-1(KD)) or pharmacological inhibition by MF63 strongly attenuates overall oncogenic drive. Indeed, mPGES-1(SC) cells express stem-cell-like features (high CD44, β1-integrin, Nanog and Oct4 and low CD24 and α6-integrin) as well as mesenchymal transition markers (high vimentin, high fibronectin, low E-cadherin). They also show increased capacity to survive irrespective of anchorage condition, and overexpress EGFR compared to mPGES-1(KD) cells. mPGES-1 expression correlates with increased in vivo tumour growth and metastasis. Although EGFR inhibition reduces mPGES-1(SC) and mPGES-1(KD) cell xenograft tumour growth, we show that mPGES-1/PGE2 signalling sensitizes tumour cells to EGFR inhibitors. We propose mPGES-1 as a possible new marker of tumour aggressiveness in PCa.
Collapse
Affiliation(s)
- Federica Finetti
- Department of Life SciencesUniversity of Siena, Via Aldo Moro 2, 53100 Siena, ItalyDepartment of Surgery and Translational MedicineUniversity of Florence, Largo Brambilla 3, 50134 Firenze, ItalyDepartment of Clinical and Experimental MedicineUniversity of Florence, Viale Pieraccini 18, 50139 Firenze, ItalyDepartment of Medical Biochemistry and BiophysicsKarolinska Institutet, SE-171 77 Stockholm, SwedenIstituto Toscano Tumori (ITT)Firenze, Italy
| | - Erika Terzuoli
- Department of Life SciencesUniversity of Siena, Via Aldo Moro 2, 53100 Siena, ItalyDepartment of Surgery and Translational MedicineUniversity of Florence, Largo Brambilla 3, 50134 Firenze, ItalyDepartment of Clinical and Experimental MedicineUniversity of Florence, Viale Pieraccini 18, 50139 Firenze, ItalyDepartment of Medical Biochemistry and BiophysicsKarolinska Institutet, SE-171 77 Stockholm, SwedenIstituto Toscano Tumori (ITT)Firenze, Italy
| | - Antonio Giachetti
- Department of Life SciencesUniversity of Siena, Via Aldo Moro 2, 53100 Siena, ItalyDepartment of Surgery and Translational MedicineUniversity of Florence, Largo Brambilla 3, 50134 Firenze, ItalyDepartment of Clinical and Experimental MedicineUniversity of Florence, Viale Pieraccini 18, 50139 Firenze, ItalyDepartment of Medical Biochemistry and BiophysicsKarolinska Institutet, SE-171 77 Stockholm, SwedenIstituto Toscano Tumori (ITT)Firenze, Italy
| | - Raffaella Santi
- Department of Life SciencesUniversity of Siena, Via Aldo Moro 2, 53100 Siena, ItalyDepartment of Surgery and Translational MedicineUniversity of Florence, Largo Brambilla 3, 50134 Firenze, ItalyDepartment of Clinical and Experimental MedicineUniversity of Florence, Viale Pieraccini 18, 50139 Firenze, ItalyDepartment of Medical Biochemistry and BiophysicsKarolinska Institutet, SE-171 77 Stockholm, SwedenIstituto Toscano Tumori (ITT)Firenze, Italy
| | - Donata Villari
- Department of Life SciencesUniversity of Siena, Via Aldo Moro 2, 53100 Siena, ItalyDepartment of Surgery and Translational MedicineUniversity of Florence, Largo Brambilla 3, 50134 Firenze, ItalyDepartment of Clinical and Experimental MedicineUniversity of Florence, Viale Pieraccini 18, 50139 Firenze, ItalyDepartment of Medical Biochemistry and BiophysicsKarolinska Institutet, SE-171 77 Stockholm, SwedenIstituto Toscano Tumori (ITT)Firenze, Italy
| | - Hiromi Hanaka
- Department of Life SciencesUniversity of Siena, Via Aldo Moro 2, 53100 Siena, ItalyDepartment of Surgery and Translational MedicineUniversity of Florence, Largo Brambilla 3, 50134 Firenze, ItalyDepartment of Clinical and Experimental MedicineUniversity of Florence, Viale Pieraccini 18, 50139 Firenze, ItalyDepartment of Medical Biochemistry and BiophysicsKarolinska Institutet, SE-171 77 Stockholm, SwedenIstituto Toscano Tumori (ITT)Firenze, Italy
| | - Olof Radmark
- Department of Life SciencesUniversity of Siena, Via Aldo Moro 2, 53100 Siena, ItalyDepartment of Surgery and Translational MedicineUniversity of Florence, Largo Brambilla 3, 50134 Firenze, ItalyDepartment of Clinical and Experimental MedicineUniversity of Florence, Viale Pieraccini 18, 50139 Firenze, ItalyDepartment of Medical Biochemistry and BiophysicsKarolinska Institutet, SE-171 77 Stockholm, SwedenIstituto Toscano Tumori (ITT)Firenze, Italy
| | - Marina Ziche
- Department of Life SciencesUniversity of Siena, Via Aldo Moro 2, 53100 Siena, ItalyDepartment of Surgery and Translational MedicineUniversity of Florence, Largo Brambilla 3, 50134 Firenze, ItalyDepartment of Clinical and Experimental MedicineUniversity of Florence, Viale Pieraccini 18, 50139 Firenze, ItalyDepartment of Medical Biochemistry and BiophysicsKarolinska Institutet, SE-171 77 Stockholm, SwedenIstituto Toscano Tumori (ITT)Firenze, Italy Department of Life SciencesUniversity of Siena, Via Aldo Moro 2, 53100 Siena, ItalyDepartment of Surgery and Translational MedicineUniversity of Florence, Largo Brambilla 3, 50134 Firenze, ItalyDepartment of Clinical and Experimental MedicineUniversity of Florence, Viale Pieraccini 18, 50139 Firenze, ItalyDepartment of Medical Biochemistry and BiophysicsKarolinska Institutet, SE-171 77 Stockholm, SwedenIstituto Toscano Tumori (ITT)Firenze, Italy
| | - Sandra Donnini
- Department of Life SciencesUniversity of Siena, Via Aldo Moro 2, 53100 Siena, ItalyDepartment of Surgery and Translational MedicineUniversity of Florence, Largo Brambilla 3, 50134 Firenze, ItalyDepartment of Clinical and Experimental MedicineUniversity of Florence, Viale Pieraccini 18, 50139 Firenze, ItalyDepartment of Medical Biochemistry and BiophysicsKarolinska Institutet, SE-171 77 Stockholm, SwedenIstituto Toscano Tumori (ITT)Firenze, Italy Department of Life SciencesUniversity of Siena, Via Aldo Moro 2, 53100 Siena, ItalyDepartment of Surgery and Translational MedicineUniversity of Florence, Largo Brambilla 3, 50134 Firenze, ItalyDepartment of Clinical and Experimental MedicineUniversity of Florence, Viale Pieraccini 18, 50139 Firenze, ItalyDepartment of Medical Biochemistry and BiophysicsKarolinska Institutet, SE-171 77 Stockholm, SwedenIstituto Toscano Tumori (ITT)Firenze, Italy
| |
Collapse
|
49
|
Larsson K, Jakobsson PJ. Inhibition of microsomal prostaglandin E synthase-1 as targeted therapy in cancer treatment. Prostaglandins Other Lipid Mediat 2015; 120:161-5. [PMID: 26100239 DOI: 10.1016/j.prostaglandins.2015.06.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 05/22/2015] [Accepted: 06/02/2015] [Indexed: 11/29/2022]
|
50
|
COX/mPGES-1/PGE2 pathway depicts an inflammatory-dependent high-risk neuroblastoma subset. Proc Natl Acad Sci U S A 2015; 112:8070-5. [PMID: 26080408 DOI: 10.1073/pnas.1424355112] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The majority of solid tumors are presented with an inflammatory microenvironment. Proinflammatory lipid mediators including prostaglandin E2 (PGE2) contribute to the establishment of inflammation and have been linked to tumor growth and aggressiveness. Here we show that high-risk neuroblastoma with deletion of chromosome 11q represents an inflammatory subset of neuroblastomas. Analysis of enzymes involved in the production of proinflammatory lipid mediators showed that 11q-deleted neuroblastoma tumors express high levels of microsomal prostaglandin E synthase-1 (mPGES-1) and elevated levels of PGE2. High mPGES-1 expression also corresponded to poor survival of neuroblastoma patients. Investigation of the tumor microenvironment showed high infiltration of tumor-promoting macrophages with high expression of the M2-polarization markers CD163 and CD206. mPGES-1-expressing cells in tumors from different subtypes of neuroblastoma showed differential expression of one or several cancer-associated fibroblast markers such as vimentin, fibroblast activation protein α, α smooth muscle actin, and PDGF receptor β. Importantly, inhibition of PGE2 production with diclofenac, a nonselective COX inhibitor, resulted in reduced tumor growth in an in vivo model of 11q-deleted neuroblastoma. Collectively, these results suggest that PGE2 is involved in the tumor microenvironment of specific neuroblastoma subgroups and indicate that therapeutic strategies using existing anti-inflammatory drugs in combination with current treatment should be considered for certain neuroblastomas.
Collapse
|