1
|
Cheng L, Hu Z, Gu J, Li Q, Liu J, Liu M, Li J, Bi X. Exploring COX-Independent Pathways: A Novel Approach for Meloxicam and Other NSAIDs in Cancer and Cardiovascular Disease Treatment. Pharmaceuticals (Basel) 2024; 17:1488. [PMID: 39598398 PMCID: PMC11597362 DOI: 10.3390/ph17111488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
As a fundamental process of innate immunity, inflammation is associated with the pathologic process of various diseases and constitutes a prevalent risk factor for both cancer and cardiovascular disease (CVD). Studies have indicated that several non-steroidal anti-inflammatory drugs (NSAIDs), including Meloxicam, may prevent tumorigenesis, reduce the risk of carcinogenesis, improve the efficacy of anticancer therapies, and reduce the risk of CVD, in addition to controlling the body's inflammatory imbalances. Traditionally, most NSAIDs work by inhibiting cyclooxygenase (COX) activity, thereby blocking the synthesis of prostaglandins (PGs), which play a role in inflammation, cancer, and various cardiovascular conditions. However, long-term COX inhibition and reduced PGs synthesis can result in serious side effects. Recent studies have increasingly shown that some selective COX-2 inhibitors and NSAIDs, such as Meloxicam, may exert effects beyond COX inhibition. This emerging understanding prompts a re-evaluation of the mechanisms by which NSAIDs operate, suggesting that their benefits in cancer and CVD treatment may not solely depend on COX targeting. In this review, we will explore the potential COX-independent mechanisms of Meloxicam and other NSAIDs in addressing oncology and cardiovascular health.
Collapse
Affiliation(s)
- Lixia Cheng
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (L.C.); (Z.H.); (Q.L.); (J.L.); (M.L.); (J.L.)
| | - Zhenghui Hu
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (L.C.); (Z.H.); (Q.L.); (J.L.); (M.L.); (J.L.)
| | - Jiawei Gu
- Department of Precision Genomics, Intermountain Healthcare, 5121 Cottonwood St., Murray, UT 84107, USA;
| | - Qian Li
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (L.C.); (Z.H.); (Q.L.); (J.L.); (M.L.); (J.L.)
| | - Jiahao Liu
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (L.C.); (Z.H.); (Q.L.); (J.L.); (M.L.); (J.L.)
| | - Meiling Liu
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (L.C.); (Z.H.); (Q.L.); (J.L.); (M.L.); (J.L.)
| | - Jie Li
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (L.C.); (Z.H.); (Q.L.); (J.L.); (M.L.); (J.L.)
| | - Xiaowen Bi
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (L.C.); (Z.H.); (Q.L.); (J.L.); (M.L.); (J.L.)
| |
Collapse
|
2
|
Kamal MV, Rao M, Damerla RR, Pai A, Sharan K, Palod A, Shetty PS, Usman N, Kumar NAN. A Mechanistic Review of Methotrexate and Celecoxib as a Potential Metronomic Chemotherapy for Oral Squamous Cell Carcinoma. Cancer Invest 2023; 41:144-154. [PMID: 36269850 DOI: 10.1080/07357907.2022.2139840] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The combination of low-dose methotrexate and celecoxib as metronomic chemotherapy (MCT) is a novel therapy, believed to act by modulating the immune response, inhibiting angiogenesis and its cytotoxic action, though the exact mechanism of action is unclear. Clinically, MCT was found to be very effective in delaying tumor progression in patients with head and neck squamous cell carcinoma in both curative and palliative settings. This review was aimed to give a brief insight into the mechanism of action and potential molecular alterations of MCT in the treatment of oral cancers taking into consideration the various in vivo and in vitro studies.
Collapse
Affiliation(s)
- Mehta Vedant Kamal
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Mahadev Rao
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Rama Rao Damerla
- Department of Medical Genetics, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Ananth Pai
- Department of Medical Oncology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Krishan Sharan
- Department of Radiotherapy and Oncology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Akhil Palod
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Preethi S Shetty
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Nawaz Usman
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Naveena A N Kumar
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| |
Collapse
|
3
|
Kolawole OR, Kashfi K. NSAIDs and Cancer Resolution: New Paradigms beyond Cyclooxygenase. Int J Mol Sci 2022; 23:1432. [PMID: 35163356 PMCID: PMC8836048 DOI: 10.3390/ijms23031432] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
Acute inflammation or resolved inflammation is an adaptive host defense mechanism and is self-limiting, which returns the body to a state of homeostasis. However, unresolved, uncontrolled, or chronic inflammation may lead to various maladies, including cancer. Important evidence that links inflammation and cancer is that nonsteroidal anti-inflammatory drugs (NSAIDs), such as aspirin, reduce the risk and mortality from many cancers. The fact that NSAIDs inhibit the eicosanoid pathway prompted mechanistic drug developmental work focusing on cyclooxygenase (COX) and its products. The increased prostaglandin E2 levels and the overexpression of COX-2 in the colon and many other cancers provided the rationale for clinical trials with COX-2 inhibitors for cancer prevention or treatment. However, NSAIDs do not require the presence of COX-2 to prevent cancer. In this review, we highlight the effects of NSAIDs and selective COX-2 inhibitors (COXIBs) on targets beyond COX-2 that have shown to be important against many cancers. Finally, we hone in on specialized pro-resolving mediators (SPMs) that are biosynthesized locally and, in a time, -dependent manner to promote the resolution of inflammation and subsequent tissue healing. Different classes of SPMs are reviewed, highlighting aspirin's potential in triggering the production of these resolution-promoting mediators (resolvins, lipoxins, protectins, and maresins), which show promise in inhibiting cancer growth and metastasis.
Collapse
Affiliation(s)
- Oluwafunke R. Kolawole
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA;
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA;
- Graduate Program in Biology, City University of New York Graduate Center, New York, NY 10091, USA
| |
Collapse
|
4
|
Kumar S, Noronha V, Patil V, Joshi A, Menon N, Prabhash K. Advances in pharmacotherapy for head and neck cancer. Expert Opin Pharmacother 2021; 22:2007-2018. [PMID: 34187268 DOI: 10.1080/14656566.2021.1948011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Introduction: Head and neck squamous cell carcinoma (HNSCC) is one of the most common cancers worldwide and is a leading cause for cancer-related mortality. This review attempts to give a comprehensive summary of the recent developments in pharmacotherapeutic options for locally advanced/metastatic HNSCC.Areas covered: In this review, the authors conducted a systematic literature search for published articles on HNSCC in the PubMed database using the keywords 'head and neck squamous cell carcinoma or HNSCC,' 'targeted therapy,' 'immunotherapy.' The search was restricted to meta-analyses, clinical trials, practice guidelines, and abstract presentations at international meetings. The final search encompassed articles published from 2010 to 2021. Articles published in languages other than English were excluded.Expert opinion: Immune checkpoint inhibition has been the most significant advance in the treatment of R/M HNSCC. Oral metronomic therapy has emerged as an important therapeutic option for low to middle-income countries. H-RAS inhibition is one of the most promising areas of research.
Collapse
Affiliation(s)
- Shikhar Kumar
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Vanita Noronha
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Vijay Patil
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Amit Joshi
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Nandini Menon
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| |
Collapse
|
5
|
Zhang P, Song E, Jiang M, Song Y. Celecoxib and Afatinib synergistic enhance radiotherapy sensitivity on human non-small cell lung cancer A549 cells. Int J Radiat Biol 2020; 97:170-178. [PMID: 33164600 DOI: 10.1080/09553002.2021.1846817] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE Radioresistance is highly correlated with radiotherapy failure in clinical cancer treatment. In the current study, we sought to examine the efficacy of Celecoxib and Afatinib co-treatment as radiosensitizers in the management of non-small cell lung cancer (NSCLC) A549 cells. MATERIALS AND METHODS Generally, A549 cells were cultured with the treatment of Celecoxib and/or Afatinib for 24 h. Then, the cells were exposed to irradiation at 2 Gy/min for 1 min. After the end of treatment, cell viability, clonogenic survival, apoptosis and Prostaglandin E2 (PGE2) Elisa assays were performed. Transcriptional levels of Cyclooxygenase-2 (COX-2) affected by Celecoxib and/or Afatinib were measured by RT-qPCR. Posttranscriptional level of epidermal growth factor receptor (EGFR)-related gene was measured by Western blotting analysis. RESULTS Here, we, for the first time, reported that the co-treatment of Celecoxib and Afatinib regulates the resistance of NSCLC A549 cells to radiation. The co-treatment of Celecoxib and Afatinib sensitized radiotherapy through the radiation-induced loss of cell viability and colony formation, as well as apoptosis. Mechanistically, Celecoxib and Afatinib-treated cells showed the inhibition of COX-2 and EGFR expression, which may be responsible for the A549 cells' increased resistance to radiation. CONCLUSION Our results suggested that Celecoxib and Afatinib regulate cell sensitivity to apoptosis, and thus modulate the resistance of NSCLC to radiation.
Collapse
Affiliation(s)
- Pan Zhang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China
| | - Erqun Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China
| | - Mingdong Jiang
- Department of Radiation Oncology, The Ninth People's Hospital of Chongqing, Chongqing, People's Republic of China
| | - Yang Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China
| |
Collapse
|
6
|
Shi CY, He XB, Zhao C, Wang HJ. Luteoloside Exerts Analgesic Effect in a Complete Freund's Adjuvant-Induced Inflammatory Model via Inhibiting Interleukin-1β Expression and Macrophage/Microglia Activation. Front Pharmacol 2020; 11:1158. [PMID: 32848767 PMCID: PMC7412990 DOI: 10.3389/fphar.2020.01158] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 07/16/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Flavonoid monomers are proved to have an anti-inflammatory effect and may also be promising for chronic pain treatment. In the present study, the analgesic effect and the relevant mechanisms of luteoloside, one of the flavonoid monomers, were investigated. METHODS The analgesic effect of luteoloside was first evaluated in complete Freud's adjuvant induced inflammatory model by von Frey test and Hargreaves test in both male and female mice. The interleukin-1β levels in plantar tissue, serum, dorsal root ganglion, and the dorsal horn of the spinal cord were determined by enzyme-linked immunosorbent assay or immunofluorescence. The activation of macrophage/microglia was tested by Iba-1 staining. RESULTS Our data showed that luteoloside exhibited both acute and chronic analgesic phenotypes. Every single dose of luteoloside solution reached the peak transient analgesic effect 2 h after administration and lasted less than 6 h. About 14 consecutive days administration (one dose per day) later, luteoloside showed a sustained analgesic effect which lasted more than 24 h. Celecoxib 20 mg/kg combined with luteoloside 40 mg/kg achieved a similar analgesic effect as celecoxib 40 mg/kg alone. Luteoloside inhibited interleukin-1β expression in plantar tissue, dorsal root ganglion, the dorsal horn of spinal cord, and serum, after 14 days of continuous administration. Furthermore, our results also showed that the activation of macrophage/microglia in dorsal root ganglions were significantly inhibited 2 h after each single dose in daily luteoloside administration and recovered to a higher level 6 h later. These findings might be involved in the mechanisms of the acute analgesic effect of luteoloside. CONCLUSION Luteoloside presents an analgesic effect via anti-inflammatory and other mechanisms such as inhibiting the activation of macrophage/microglia.
Collapse
Affiliation(s)
- Chun-Yan Shi
- Institute of Chinese Medicine, Shanghai University of Chinese Medicine, Shanghai, China
- Laboratory of Neuropsychopharmacology, College of Fundamental Medicine, Shanghai University of Medicine & Health Science, Shanghai, China
| | - Xi-Biao He
- Laboratory of Neuropsychopharmacology, College of Fundamental Medicine, Shanghai University of Medicine & Health Science, Shanghai, China
| | - Chao Zhao
- National Clinical Research Center for Aging and Medicine, Huashan Hospital and MOE/NHC/CAMS Key Lab of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hui-Jing Wang
- Laboratory of Neuropsychopharmacology, College of Fundamental Medicine, Shanghai University of Medicine & Health Science, Shanghai, China
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
7
|
Mishra S, Srivastava S, Divakar A, Mandal P, Dewangan J, Chaturvedi S, Wahajuddin M, Kumar S, Tripathi A, Rath SK. Celecoxib reduces Deoxynivalenol induced proliferation, inflammation and protein kinase C translocation via modulating downstream targets in mouse skin. Chem Biol Interact 2020; 326:109128. [PMID: 32416088 DOI: 10.1016/j.cbi.2020.109128] [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: 09/23/2019] [Revised: 04/13/2020] [Accepted: 05/05/2020] [Indexed: 12/26/2022]
Abstract
Exposure to mycotoxins is mostly by ingestion but also occurs by the dermal and inhalation routes. The present study for the first time demonstrated that mycotoxin Deoxynivalenol (DON), permeates through Swiss albino mice skin, which demands awareness of health risks in people who are dermally exposed to mycotoxins especially agricultural farmers. Despite the widespread contamination of DON in food commodities studies to alleviate DON's toxicity are sparsely reported. Thus effective measures to combat mycotoxins associated toxicity remains an imperative aspect to be considered from the angle of dermal exposure. Topical application of Celecoxib (1-2 mg), followed by DON (100 μg) application on the dorsal side of mice, resulted in substantial decrease in DON-induced (i) edema, hyperplasia, cell proliferation (ii) inhibition of cytokine and prostaglandin-E2 levels (iii) phosphorylation of ERK1/2, JNK, p38, MAPKKs, CREB, P90-RSK (iv) downregulation of c-Jun, c- Fos, phospho-NF-kB and their downstream target proteins cyclin D1 and COX-2. Using Ro-31-8220 (Protein-Kinase-C inhibitor), it was observed PKC was responsible for DON induced upregulation of COX-2 and iNOS proteins. Treatment of Celecoxib decreased DON-induced translocation of Protein Kinase C isozymes (α,ε,γ), demonstrating the role of PKC in DON-mediated biochemical and molecular alterations responsible for its dermal toxicity. The present findings indicate that topical application of celecoxib is effective in the management of inflammatory skin disorders induced by foodborne fungal toxin DON. The skin permeation potential of Celecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor NSAID, was also assessed, and the results indicated that the permeation was relatively lower as compared to the oral mode of administration. Hence topical use of celecoxib may be preferred over oral dosing because of lower systemic absorption and to avoid the unwanted side effects. This study provides a prospect for exploring the clinical efficacy of topically applied COX-2 inhibitors for the management of inflammatory skin disorders induced by foodborne fungal toxins.
Collapse
Affiliation(s)
- Sakshi Mishra
- Genotoxicity Laboratory, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Sonal Srivastava
- Genotoxicity Laboratory, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Aman Divakar
- Genotoxicity Laboratory, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Payal Mandal
- Food Drug and Chemical Toxicology Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, 226 001, Uttar Pradesh, India
| | - Jayant Dewangan
- Genotoxicity Laboratory, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Swati Chaturvedi
- Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Muhammad Wahajuddin
- Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Sadan Kumar
- Immunotoxicology Laboratory, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Anurag Tripathi
- Food Drug and Chemical Toxicology Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, 226 001, Uttar Pradesh, India
| | - Srikanta Kumar Rath
- Genotoxicity Laboratory, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India.
| |
Collapse
|
8
|
Buzharevski A, Paskas S, Sárosi MB, Laube M, Lönnecke P, Neumann W, Mijatovic S, Maksimovic-Ivanic D, Pietzsch J, Hey-Hawkins E. Carboranyl Analogues of Celecoxib with Potent Cytostatic Activity against Human Melanoma and Colon Cancer Cell Lines. ChemMedChem 2019; 14:315-321. [PMID: 30602073 DOI: 10.1002/cmdc.201800685] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/14/2018] [Indexed: 12/13/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are the most common way of treating inflammatory disorders. Their widespread use helped reveal their other modes of action as pharmaceuticals, such as a profound effect on various cancers. Celecoxib has proven to be a very prominent member of this group with cytostatic activities. On the other hand, the highly dynamic field of drug design is constantly searching for new ways of modifying known structures to obtain more powerful and less harmful drugs. A very interesting development is the implementation of carboranes in pharmacologically active structures, mostly as phenyl mimetics. Herein we report the synthesis of three carborane-containing derivatives of the COX-2-selective NSAID celecoxib. The new compounds proved to have promising cytostatic potential against various melanoma and colorectal adenocarcinoma cell lines. Inhibited proliferation accompanied by caspase-independent apoptotic cell death was found to be the main cause of decreased cell viability upon treatment with the most efficient celecoxib analogue, 3 b (4-[5-(1,7-dicarba-closo-dodecaboranyl)-3-trifluoromethyl-1H-pyrazol-1-yl]-1-methylsulfonylbenzene).
Collapse
Affiliation(s)
- Antonio Buzharevski
- Institut für Anorganische Chemie, Universität Leipzig, Johannisallee 29, 04103, Leipzig, Germany
| | - Svetlana Paskas
- Department of Immunology, Institute for Biological Research "Sinisa Stankovic", Belgrade University, Belgrade, Serbia
| | - Menyhárt-Botond Sárosi
- Institut für Anorganische Chemie, Universität Leipzig, Johannisallee 29, 04103, Leipzig, Germany
| | - Markus Laube
- Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Peter Lönnecke
- Institut für Anorganische Chemie, Universität Leipzig, Johannisallee 29, 04103, Leipzig, Germany
| | - Wilma Neumann
- Institut für Anorganische Chemie, Universität Leipzig, Johannisallee 29, 04103, Leipzig, Germany
| | - Sanja Mijatovic
- Department of Immunology, Institute for Biological Research "Sinisa Stankovic", Belgrade University, Belgrade, Serbia
| | - Danijela Maksimovic-Ivanic
- Department of Immunology, Institute for Biological Research "Sinisa Stankovic", Belgrade University, Belgrade, Serbia
| | - Jens Pietzsch
- Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstrasse 4, 01062, Dresden, Germany
| | - Evamarie Hey-Hawkins
- Institut für Anorganische Chemie, Universität Leipzig, Johannisallee 29, 04103, Leipzig, Germany
| |
Collapse
|
9
|
p38 Expression and Modulation of STAT3 Signaling in Oral Cancer. Pathol Oncol Res 2018; 26:183-192. [DOI: 10.1007/s12253-018-0405-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/07/2018] [Indexed: 12/19/2022]
|
10
|
Sung MW, Lee DY, Park SW, Oh SM, Choi JJ, Shin ES, Kwon SK, Ahn SH, Kim YH. Celecoxib enhances the inhibitory effect of 5-FU on human squamous cell carcinoma proliferation by ROS production. Laryngoscope 2016; 127:E117-E123. [PMID: 27666139 DOI: 10.1002/lary.26309] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/23/2016] [Accepted: 08/05/2016] [Indexed: 01/13/2023]
Abstract
OBJECTIVES The role of celecoxib in preventing and treating tumors has attracted broad attention in recent years because of its selective and specific inhibition of COX-2 activity. We investigated the inhibitory effects and mechanisms of celecoxib combined with 5-fluorouracil (5-FU) on proliferation of squamous cell carcinoma cells in vivo and in vitro. STUDY DESIGN Animal study and basic research. METHODS SNU-1041 and SNU-1076 squamous cell lines and an orthotopic tongue cancer mouse model were used to study growth inhibition with 5-FU enhanced by celecoxib. Sensitivity of cells to drug treatment was analyzed by the MTT assay, and generation of reactive oxygen species (ROS) was measured using dichlorofluorescein diacetate. Phosphorylation of AKT was detected by Western blotting. Survival analysis in the mouse model was assessed according to combination treatment with 5-FU and celecoxib. RESULTS Reactive oxygen species production in vitro was highest when celecoxib was administered 48 hours after 5-FU treatment. 5-FU-induced inhibition of cell proliferation was enhanced when combined with celecoxib, which was positively correlated with ROS production. Antioxidant treatment reversed 5-FU-inhibited cell proliferation by up to 60%. Cotreatment with celecoxib and 5-FU partially blocked AKT phosphorylation, although no significant changes in total AKT protein levels were detected. An increased survival time was observed in an orthotopic mouse model treated with a combination of celecoxib and 5-FU compared to treatment with either agent alone. CONCLUSION Celecoxib may have an enhanced anticancer effect in combination with 5-FU. Reactive oxygen species production may be a key mechanism in this combination therapy by inhibiting the AKT pathway. LEVEL OF EVIDENCE N/A. Laryngoscope, 127:E117-E123, 2017.
Collapse
Affiliation(s)
- Myung-Whun Sung
- Department of Otorhinolaryngology Head and Neck Surgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul.,Cancer Research Institute, Seoul.,Sheikh Khalifa Specialty Hospital, Ras Al Khaimah, United Arab Emirates
| | - Doh Young Lee
- Cancer Research Institute, Seoul.,Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Anam Hostpital, Seoul
| | | | | | - Jun-Jae Choi
- Department of Otorhinolaryngology Head and Neck Surgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul
| | - Eun Sil Shin
- Department of Otorhinolaryngology Head and Neck Surgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul
| | - Seong Keun Kwon
- Department of Otorhinolaryngology Head and Neck Surgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul.,Cancer Research Institute, Seoul
| | - Soon-Hyun Ahn
- Department of Otorhinolaryngology Head and Neck Surgery, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Young Ho Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul
| |
Collapse
|
11
|
Park SW, Hah JH, Oh SM, Jeong WJ, Sung MW. 5-lipoxygenase mediates docosahexaenoyl ethanolamide and N-arachidonoyl-L-alanine-induced reactive oxygen species production and inhibition of proliferation of head and neck squamous cell carcinoma cells. BMC Cancer 2016; 16:458. [PMID: 27411387 PMCID: PMC4942960 DOI: 10.1186/s12885-016-2499-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 07/04/2016] [Indexed: 01/01/2023] Open
Abstract
Background Endocannabinoids have recently drawn attention as promising anti-cancer agents. We previously observed that anandamide (AEA), one of the representative endocannabinoids, effectively inhibited the proliferation of head and neck squamous cell carcinoma (HNSCC) cell lines in a receptor-independent manner. In this study, using HNSCC cell lines, we examined the anti-cancer effects and the mechanisms of action of docosahexaenoyl ethanolamide (DHEA) and N-arachidonoyl-L-alanine (NALA), which are polyunsaturated fatty acid (PUFA)-based ethanolamides like AEA. Methods and Results DHEA and NALA were found to effectively inhibit HNSCC cell proliferation. These anti-proliferative effects seemed to be mediated in a cannabinoid receptor-independent manner, since the antagonist of cannabinoid receptor-1 (CB1) and vanilloid receptor-1 (VR1), two endocannabinoid receptors, did not reverse the ability of DHEA and NALA to induce cell death. Instead, we observed an increase in reactive oxygen species (ROS) production and a decrease of phosphorylated Akt as a result of DHEA and NALA treatment. Antioxidants efficiently reversed the inhibition of cell proliferation and the decrease of phosphorylated Akt induced by DHEA and NALA; inhibition of 5-lipoxygenase (5-LO), which is expected to be involved in DHEA- and NALA-degradation pathway, also partially blocked the ability of DHEA and NALA to inhibit cell proliferation and phosphorylated Akt. Interestingly, ROS production as a result of DHEA and NALA treatment was decreased by inhibition of 5-LO. Conclusions From these findings, we suggest that ROS production induced by the 5-LO pathway mediates the anti-cancer effects of DHEA and NALA on HNSCC cells. Finally, our findings suggest the possibility of a new cancer-specific therapeutic strategy, which utilizes 5-LO activity rather than inhibiting it. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2499-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Seok-Woo Park
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - J Hun Hah
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul, South Korea.,Clinical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Sang-Mi Oh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Woo-Jin Jeong
- Department of Otorhinolaryngology, Seoul National University Bundang Hospital, Seoul, South Korea
| | - Myung-Whun Sung
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea. .,Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul, South Korea. .,Clinical Research Institute, Seoul National University Hospital, Seoul, South Korea. .,Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul National University Hospital, Seoul, South Korea.
| |
Collapse
|
12
|
Sobolewski C, Rhim J, Legrand N, Muller F, Cerella C, Mack F, Chateauvieux S, Kim JG, Yoon AY, Kim KW, Dicato M, Diederich M. 2,5-Dimethyl-celecoxib inhibits cell cycle progression and induces apoptosis in human leukemia cells. J Pharmacol Exp Ther 2015; 355:308-28. [PMID: 26330537 DOI: 10.1124/jpet.115.225011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 08/28/2015] [Indexed: 03/08/2025] Open
Abstract
Cyclooxygenase-2 (COX-2) is an essential regulator of cancer promotion and progression. Extensive efforts to target this enzyme have been developed to reduce growth of cancer cells for chemopreventive and therapeutic reasons. In this context, cyclooxygenase-2 inhibitors present interesting antitumor effects. However, inhibition of COX-2 by anti-COX-2 compounds such as celecoxib was recently associated with detrimental cardiovascular side effects limiting their clinical use. As many anticancer effects of celecoxib are COX-2 independent, analogs such as 2,5-dimethyl-celecoxib (DMC), which lacks COX-2-inhibitory activity, represent a promising alternative strategy. In this study, we investigated the effect of this molecule on growth of hematologic cancer cell lines (U937, Jurkat, Hel, Raji, and K562). We found that this molecule is able to reduce the growth and induces apoptosis more efficiently than celecoxib in all the leukemic cell lines tested. Cell death was associated with downregulation of Mcl-1 protein expression. We also found that DMC induces endoplasmic reticulum stress, which is associated with a decreased of GRP78 protein expression and an alteration of cell cycle progression at the G1/S transition in U937 cells. Accordingly, typical downregulation of c-Myc and cyclin D1 and an upregulation of p27 were observed. Interestingly, for shorter time points, an alteration of mitotic progression, associated with the downregulation of survivin protein expression was observed. Altogether, our data provide new evidence about the mode of action of this compound on hematologic malignancies.
Collapse
Affiliation(s)
- Cyril Sobolewski
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, Luxembourg, Luxembourg (C.S., N.L., F.Mu. C.C., F.Ma., S.C., M.Dic.); Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea (J.R., S.C., M.Die.); and SNU-Harvard Neurovascular Protection Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea (J.G.K., A.Y.Y., K.W.K.)
| | - Jiyun Rhim
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, Luxembourg, Luxembourg (C.S., N.L., F.Mu. C.C., F.Ma., S.C., M.Dic.); Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea (J.R., S.C., M.Die.); and SNU-Harvard Neurovascular Protection Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea (J.G.K., A.Y.Y., K.W.K.)
| | - Noémie Legrand
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, Luxembourg, Luxembourg (C.S., N.L., F.Mu. C.C., F.Ma., S.C., M.Dic.); Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea (J.R., S.C., M.Die.); and SNU-Harvard Neurovascular Protection Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea (J.G.K., A.Y.Y., K.W.K.)
| | - Florian Muller
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, Luxembourg, Luxembourg (C.S., N.L., F.Mu. C.C., F.Ma., S.C., M.Dic.); Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea (J.R., S.C., M.Die.); and SNU-Harvard Neurovascular Protection Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea (J.G.K., A.Y.Y., K.W.K.)
| | - Claudia Cerella
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, Luxembourg, Luxembourg (C.S., N.L., F.Mu. C.C., F.Ma., S.C., M.Dic.); Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea (J.R., S.C., M.Die.); and SNU-Harvard Neurovascular Protection Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea (J.G.K., A.Y.Y., K.W.K.)
| | - Fabienne Mack
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, Luxembourg, Luxembourg (C.S., N.L., F.Mu. C.C., F.Ma., S.C., M.Dic.); Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea (J.R., S.C., M.Die.); and SNU-Harvard Neurovascular Protection Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea (J.G.K., A.Y.Y., K.W.K.)
| | - Sébastien Chateauvieux
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, Luxembourg, Luxembourg (C.S., N.L., F.Mu. C.C., F.Ma., S.C., M.Dic.); Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea (J.R., S.C., M.Die.); and SNU-Harvard Neurovascular Protection Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea (J.G.K., A.Y.Y., K.W.K.)
| | - Jeoung-Gyun Kim
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, Luxembourg, Luxembourg (C.S., N.L., F.Mu. C.C., F.Ma., S.C., M.Dic.); Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea (J.R., S.C., M.Die.); and SNU-Harvard Neurovascular Protection Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea (J.G.K., A.Y.Y., K.W.K.)
| | - Ah-Young Yoon
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, Luxembourg, Luxembourg (C.S., N.L., F.Mu. C.C., F.Ma., S.C., M.Dic.); Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea (J.R., S.C., M.Die.); and SNU-Harvard Neurovascular Protection Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea (J.G.K., A.Y.Y., K.W.K.)
| | - Kyu-Won Kim
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, Luxembourg, Luxembourg (C.S., N.L., F.Mu. C.C., F.Ma., S.C., M.Dic.); Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea (J.R., S.C., M.Die.); and SNU-Harvard Neurovascular Protection Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea (J.G.K., A.Y.Y., K.W.K.)
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, Luxembourg, Luxembourg (C.S., N.L., F.Mu. C.C., F.Ma., S.C., M.Dic.); Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea (J.R., S.C., M.Die.); and SNU-Harvard Neurovascular Protection Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea (J.G.K., A.Y.Y., K.W.K.)
| | - Marc Diederich
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, Luxembourg, Luxembourg (C.S., N.L., F.Mu. C.C., F.Ma., S.C., M.Dic.); Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, South Korea (J.R., S.C., M.Die.); and SNU-Harvard Neurovascular Protection Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea (J.G.K., A.Y.Y., K.W.K.)
| |
Collapse
|
13
|
Patil VM, Noronha V, Joshi A, Muddu VK, Dhumal S, Bhosale B, Arya S, Juvekar S, Banavali S, D'Cruz A, Bhattacharjee A, Prabhash K. A prospective randomized phase II study comparing metronomic chemotherapy with chemotherapy (single agent cisplatin), in patients with metastatic, relapsed or inoperable squamous cell carcinoma of head and neck. Oral Oncol 2015; 51:279-86. [PMID: 25578869 DOI: 10.1016/j.oraloncology.2014.12.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/01/2014] [Accepted: 12/03/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND Cetuximab based treatment is the recommended chemotherapy for head and neck squamous cell cancers in the palliative setting. However, due to financial constraints, intravenous (IV) chemotherapy without cetuximab is commonly used in lesser developed countries. We believe that oral metronomic chemotherapy may be safer and more effective in this setting. METHODS We conducted an open label, superiority, parallel design, randomized phase II trial comparing oral MCT [daily celecoxib (200mg twice daily) and weekly methotrexate (15mg/m(2))] to intravenous single agent cisplatin (IP) (75mg/m(2)) given 3 weekly. Eligible patients had head and neck cancers requiring palliative chemotherapy with ECOG PS 0-2 and adequate organ functions who could not afford cetuximab. The primary end point was progression-free survival. RESULTS 110 Patients were recruited between July 2011 to May 2013, 57 randomized to the MCT arm and 53 to the IP arm. Patients in the MCT arm had significantly longer PFS (median 101 days, 95% CI: 58.2-143.7 days) compared to the IP arm (median 66 days, 95% CI; 55.8-76.1 days) (p=0.014). The overall survival (OS) was also increased significantly in the MCT arm (median 249 days, 95% CI: 222.5-275.5 days) compared to the IP arm (median 152 days, 95% CI: 104.2-199.8 days) (p=0.02). There were fewer grade 3/4 adverse effects with MCT, which was not significant. (18.9% vs. 31.4%, P=0.14). CONCLUSION Oral metronomic chemotherapy has significantly better PFS and OS than single agent platinum in the palliative setting.
Collapse
Affiliation(s)
| | - Vanita Noronha
- Department of Medical Oncology, Tata Memorial Centre, Mumbai, India
| | - Amit Joshi
- Department of Medical Oncology, Tata Memorial Centre, Mumbai, India
| | | | - Sachin Dhumal
- Department of Medical Oncology, Tata Memorial Centre, Mumbai, India
| | | | - Supreeta Arya
- Department of Radio Diagnosis, Tata Memorial Centre, Mumbai, India
| | | | - Shripad Banavali
- Department of Medical Oncology, Tata Memorial Centre, Mumbai, India
| | - Anil D'Cruz
- Department of Head and Neck Surgery, Tata Memorial Centre, Mumbai, India
| | - Atanu Bhattacharjee
- Division of Clinical Research and Biostatistics, Malabar Cancer Centre, India
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Centre, Mumbai, India.
| |
Collapse
|
14
|
Park SW, Kim JE, Oh SM, Cha WJ, Hah JH, Sung MW. Anticancer effects of anandamide on head and neck squamous cell carcinoma cells via the production of receptor-independent reactive oxygen species. Head Neck 2014; 37:1187-92. [DOI: 10.1002/hed.23727] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 02/23/2014] [Accepted: 04/28/2014] [Indexed: 11/06/2022] Open
Affiliation(s)
- Seok-Woo Park
- Cancer Research Institute; Seoul National University College of Medicine; Seoul Korea
| | - Ji-Eun Kim
- Cancer Research Institute; Seoul National University College of Medicine; Seoul Korea
| | - Sang-Mi Oh
- Cancer Research Institute; Seoul National University College of Medicine; Seoul Korea
| | - Won-Jae Cha
- Department of Otorhinolaryngology; Seoul National University Hospital; Seoul Korea
| | - Jeong-Hun Hah
- Cancer Research Institute; Seoul National University College of Medicine; Seoul Korea
- Department of Otorhinolaryngology; Seoul National University Hospital; Seoul Korea
- Clinical Research Institute; Seoul National University Hospital; Seoul Korea
| | - Myung-Whun Sung
- Cancer Research Institute; Seoul National University College of Medicine; Seoul Korea
- Department of Otorhinolaryngology; Seoul National University Hospital; Seoul Korea
- Clinical Research Institute; Seoul National University Hospital; Seoul Korea
- Sensory Organ Research Institute; Seoul National University Medical Research Center, Seoul National University Hospital; Seoul Korea
| |
Collapse
|
15
|
Gillette DD, Tridandapani S, Butchar JP. Monocyte/macrophage inflammatory response pathways to combat Francisella infection: possible therapeutic targets? Front Cell Infect Microbiol 2014; 4:18. [PMID: 24600590 PMCID: PMC3930869 DOI: 10.3389/fcimb.2014.00018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 02/02/2014] [Indexed: 01/05/2023] Open
Abstract
Francisella tularensis can bypass and suppress host immune responses, even to the point of manipulating immune cell phenotypes and intercellular inflammatory networks. Strengthening these responses such that immune cells more readily identify and destroy the bacteria is likely to become a viable (and perhaps necessary) strategy for combating infections with Francisella, especially given the likelihood of antibiotic resistance in the foreseeable future. Monocytes and macrophages offer a niche wherein Francisella can invade and replicate, resulting in substantially higher bacterial load that can overcome the host. As such, understanding their responses to Francisella may uncover potential avenues of therapy that could promote a lowering of bacterial burden and clearance of infection. These response pathways include Toll-like Receptor 2 (TLR2), the caspase-1 inflammasome, Interferons, NADPH oxidase, Phosphatidylinositide 3-kinase (PI3K), and the Ras pathway. In this review we summarize the literature pertaining to the roles of these pathways during Francisella infection, with an emphasis on monocyte/macrophage responses. The therapeutic targeting of one or more such pathways may ultimately become a valuable tool for the treatment of tularemia, and several possibilities are discussed.
Collapse
Affiliation(s)
- Devyn D Gillette
- Department of Internal Medicine, Wexner Medical Center, The Ohio State University Columbus, OH, USA
| | - Susheela Tridandapani
- Department of Internal Medicine, Wexner Medical Center, The Ohio State University Columbus, OH, USA
| | - Jonathan P Butchar
- Department of Internal Medicine, Wexner Medical Center, The Ohio State University Columbus, OH, USA
| |
Collapse
|
16
|
Zhang Q, Meng X, Zheng G, Chen G, Pang R, Hua T, Yang S. Antitumor activity of celecoxib, a selective cyclooxygenase-2 inhibitor, in medullary thyroid carcinoma. Mol Med Rep 2014; 9:768-772. [PMID: 24346019 DOI: 10.3892/mmr.2013.1869] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 12/04/2013] [Indexed: 11/06/2022] Open
Abstract
The purpose of this study was to investigate the mechanisms of the antitumor effect of celecoxib (CXB) in the treatment of human medullary thyroid carcinoma (MTC). Human MTC TT cells were cultured with different concentrations (0, 20, 40, 60 µmol/l) of CXB following 0-72 h in vitro. An MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) assay was used to determine the growth inhibition of MTC in vitro. Flow cytometry was performed to analyze the cell cycle of TT cells. Levels of prostaglandin E2 (PGE2) were measured by enzyme-linked immunosorbent assay (ELISA) method. The expression profile of cyclooxygenase-2 (COX-2) and vascular endothelial growth factor (VEGF) was measured by western blot analysis. In the present study, it was identified that CXB inhibited TT cell proliferation and induced apoptosis in a dose- and time-dependent manner. The cell cycle was arrested at G0/G1 and the percentage of cells in S phase was markedly decreased. The expression levels of PGE2 were inhibited by CXB. CXB effectively downregulated the expression of COX-2 and VEGF in a dose- and time-dependent manner. These data demonstrated that CXB inhibited the proliferation of MTC TT cells in vitro and thus may be effective as an antitumor therapy for human MTC.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Thyroid Surgery, First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xianying Meng
- Department of Thyroid Surgery, First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guibin Zheng
- Department of Thyroid Surgery, First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guang Chen
- Department of Thyroid Surgery, First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Renzhu Pang
- Department of Thyroid Surgery, First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Tebo Hua
- Department of Thyroid Surgery, First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Shuai Yang
- Department of Thyroid Surgery, First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
17
|
Wang YJ, Niu XP, Yang L, Han Z, Ma YJ. Effects of celecoxib on cycle kinetics of gastric cancer cells and protein expression of cytochrome C and caspase-9. Asian Pac J Cancer Prev 2013; 14:2343-7. [PMID: 23725138 DOI: 10.7314/apjcp.2013.14.4.2343] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE This investigation aimed to determine effects of celecoxib on the cell cycle kinetics of the gastric cancer cell line MGC803 and the mechanisms involved by assessing expression of cytochrome C and caspase-9 at the protein level. METHODS Cell proliferation of MGC803 was determined by MTT assay after treatment with celecoxib. Apoptosis was assessed using fluorescence staining and cell cycle kinetics by flow cytometry. Western blotting was used to detect the expression of caspase-9 protein and of cytochrome C protein in cell cytosol and mitochondria. RESULTS Celecoxib was able to restrain proliferation and induce apoptosis in a dose- and time- dependent manner, inducing G0/G1 cell cycle arrest, release of cytochrome C into the cytosol, and cleavage of pro-caspase-9 into its active form. CONCLUSION Celecoxib can induce apoptosis in MGC803 cells through a mechanism involving cell cycle arrest, mitochondrial cytochrome C release and caspase activation.
Collapse
Affiliation(s)
- Yu-Jie Wang
- Department of Gastroenterology, Zhengzhou People's Hospital Affiliated to Southern Medical University, Zhengzhou, Henan, China
| | | | | | | | | |
Collapse
|
18
|
Farooq F, Abadía-Molina F, MacKenzie D, Hadwen J, Shamim F, O'Reilly S, Holcik M, MacKenzie A. Celecoxib increases SMN and survival in a severe spinal muscular atrophy mouse model via p38 pathway activation. Hum Mol Genet 2013; 22:3415-24. [PMID: 23656793 DOI: 10.1093/hmg/ddt191] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The loss of functional Survival Motor Neuron (SMN) protein due to mutations or deletion in the SMN1 gene causes autosomal recessive neurodegenerative spinal muscle atrophy (SMA). A potential treatment strategy for SMA is to upregulate the amount of SMN protein originating from the highly homologous SMN2 gene, compensating in part for the absence of the functional SMN1 gene. We have previously shown that in vitro activation of the p38 pathway stabilizes and increases SMN mRNA levels leading to increased SMN protein levels. In this report, we explore the impact of the p38 activating, FDA-approved, blood brain barrier permeating compound celecoxib on SMN levels in vitro and in a mouse model of SMA. We demonstrate a significant induction of SMN protein levels in human and mouse neuronal cells upon treatment with celecoxib. We show that activation of the p38 pathway by low doses celecoxib increases SMN protein in a HuR protein-dependent manner. Furthermore, celecoxib treatment induces SMN expression in brain and spinal cord samples of wild-type mice in vivo. Critically, celecoxib treatment increased SMN levels, improved motor function and enhanced survival in a severe SMA mouse model. Our results identify low dose celecoxib as a potential new member of the SMA therapeutic armamentarium.
Collapse
|
19
|
Wang ZL, Fan ZQ, Jiang HD, Qu JM. Selective Cox-2 inhibitor celecoxib induces epithelial-mesenchymal transition in human lung cancer cells via activating MEK-ERK signaling. Carcinogenesis 2012; 34:638-46. [DOI: 10.1093/carcin/bgs367] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
20
|
Celecoxib inhibits growth of human autosomal dominant polycystic kidney cyst-lining epithelial cells through the VEGF/Raf/MAPK/ERK signaling pathway. Mol Biol Rep 2012; 39:7743-53. [PMID: 22415852 PMCID: PMC3358558 DOI: 10.1007/s11033-012-1611-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 01/24/2012] [Indexed: 12/13/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a progressive chronic kidney disease. To date there are no effective medicines to halt development and growth of cysts. In the present study, we explored novel effects of celecoxib (CXB), a COX-2 specific inhibitor, on primary cultures of human ADPKD cyst-lining epithelial cells. Primary cultures of ADPKD cyst-lining epithelial cells were obtained from five patients. Effects of CXB were measured by various assays to detect BrdU incorporation, apoptosis and proliferation in vitro. Additionally, effects of CXB on kidney weight, the cyst index, the fibrosis index, blood urea nitrogen (BUN), serum creatinine (SCr), serum 6-keto-PGF-1α, serum thromboxane-2 (TXB2) and renal PCNA expression were assessed in Han:SPRD rat, a well-characterized rodent model of PKD. CXB inhibited proliferation of ADPKD cyst-lining epithelial cells, blocked the release of VEGF from the cells and induced extensive apoptosis in a time- and dose-dependent manner. Moreover, CXB up-regulated the cell cycle negative regulator p21CIP/WAF1 and the cell cycle positive regulator Cyclin A, blocked ERK1/2 phosphorylation, induced apoptotic factors (Bax and caspase-3) and reduced Bcl-2. Furthermore, CXB inhibited the expression of VEGFR-2 and Raf-1 in ADPKD cyst-lining epithelial cells. CXB markedly reduced the cyst index, the fibrosis index, leukocyte infiltration, BUN, SCr, serum 6-keto-PGF-1α, TXB2 and renal PCNA expression in Han:SPRD rat. We demonstrated for the first time that CXB could suppress renal cyst-lining growth both in vitro and in vivo in Han:SPRD rat. CXB can inhibit proliferation, suppress cell cycle progression, and induce apoptosis in ADPKD cyst-lining epithelial cells through the inhibition of the VEGF/VEGFR-2/Raf-1/MAPK/ERK signaling pathway.
Collapse
|
21
|
Current World Literature. Curr Opin Oncol 2011; 23:303-10. [DOI: 10.1097/cco.0b013e328346cbfa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|