1
|
Bukhari S, Leth MF, Laursen CCW, Larsen ME, Tornøe AS, Eriksen VR, Hovmand AEK, Jakobsen JC, Maagaard M, Mathiesen O. Risks of serious adverse events with non-steroidal anti-inflammatory drugs in gastrointestinal surgery: A systematic review with meta-analysis and trial sequential analysis. Acta Anaesthesiol Scand 2024; 68:871-887. [PMID: 38629348 DOI: 10.1111/aas.14425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Non-steroidal anti-inflammatory drugs (NSAIDs) are commonly recommended for perioperative opioid-sparing multimodal analgesic treatments. Concerns regarding the potential for serious adverse events (SAEs) associated with perioperative NSAID treatment are especially relevant following gastrointestinal surgery. We assessed the risks of SAEs with perioperative NSAID treatment in patients undergoing gastrointestinal surgery. METHODS We conducted a systematic review of randomised clinical trials assessing the harmful effects of NSAIDs versus placebo, usual care or no intervention in patients undergoing gastrointestinal surgery. The primary outcome was an incidence of SAEs. We systematically searched for eligible trials in five major databases up to January 2024. We performed risk of bias assessments to account for systematic errors, trial sequential analysis (TSA) to account for the risks of random errors, performed meta-analyses using R and used the Grading of Recommendations Assessment, Development and Evaluation framework to describe the certainty of evidence. RESULTS We included 22 trials enrolling 1622 patients for our primary analyses. Most trials were at high risk of bias. Meta-analyses (risk ratio 0.78; 95% confidence interval [CI] 0.51-1.19; I2 = 4%; p = .24; very low certainty of evidence) and TSA indicated a lack of information on the effects of NSAIDs compared to placebo on the risks of SAEs. Post-hoc beta-binomial regression sensitivity analyses including trials with zero events showed a reduction in SAEs with NSAIDs versus placebo (odds ratio 0.73; CI 0.54-0.99; p = .042). CONCLUSION In adult patients undergoing gastrointestinal surgery, there was insufficient information to draw firm conclusions on the effects of NSAIDs on SAEs. The certainty of the evidence was very low.
Collapse
Affiliation(s)
- Shaheer Bukhari
- Centre for Anaesthesiological Research, Department of Anesthesiology, Zealand University Hospital, Køge, Denmark
| | - Morten F Leth
- Centre for Anaesthesiological Research, Department of Anesthesiology, Zealand University Hospital, Køge, Denmark
| | - Christina C W Laursen
- Centre for Anaesthesiological Research, Department of Anesthesiology, Zealand University Hospital, Køge, Denmark
| | - Mia E Larsen
- Department of Anesthesiology, Nykøbing Falster Hospital, Nykøbing Falster, Denmark
| | - Anders S Tornøe
- Department of Anesthesiology, Nordland Hospital Trust, Bodø, Norway
| | - Vibeke R Eriksen
- Centre for Anaesthesiological Research, Department of Anesthesiology, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, Copenhagen University, Copenhagen, Denmark
| | - Alfred E K Hovmand
- Department of Anesthesiology, University Hospital Northern Norway, Tromsø, Norway
| | - Janus C Jakobsen
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Regional Health Research, The Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Mathias Maagaard
- Centre for Anaesthesiological Research, Department of Anesthesiology, Zealand University Hospital, Køge, Denmark
| | - Ole Mathiesen
- Centre for Anaesthesiological Research, Department of Anesthesiology, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
2
|
Tołoczko-Iwaniuk N, Dziemiańczyk-Pakieła D, Nowaszewska BK, Celińska-Janowicz K, Miltyk W. Celecoxib in Cancer Therapy and Prevention - Review. Curr Drug Targets 2020; 20:302-315. [PMID: 30073924 DOI: 10.2174/1389450119666180803121737] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/04/2018] [Accepted: 08/02/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVES It is generally accepted that inflammatory cells found in the tumor microenvironment are involved in the neoplastic process, promoting cell proliferation, survival, and migration. Therefore, administering anti-inflammatory medication in cancer therapy seems to be justified. A potential pathway associated with the aforementioned issue is cyclooxygenase-2 inhibition, particularly as the overexpression of this enzyme has been proven to occur in cancer tissues and is also associated with a poor prognosis in several types of human malignancies. Celecoxib, a COX-2 selective inhibitor, has been utilized for over 20 years, particularly as an anti-inflammatory, analgesic and antipyretic medication. However, to date, its antineoplastic properties have not been sufficiently investigated. In recent years, the number of research studies on the antineoplastic effects of celecoxib has increased considerably. The vast majority of publications refers to preclinical studies attempting to elucidate its mechanisms of action. Clinical trials concerning celecoxib have focused primarily on the treatment of cancers of the colon, breast, lung, prostate, stomach, head and neck, as well as premalignant lesions such as familial adenoma polyposis. In this review article authors attempt to summarise the latest research which has elucidated celecoxib use in the treatment and prevention of cancer. CONCLUSION Both preclinical and clinical studies have demonstrated promising results of the role of celecoxib in the treatment and prevention of cancer - the best outcome was observed in colon, breast, prostate and head and neck cancers. However, more clinical trials providing real evidence-based clinical advances of celecoxib use are needed.
Collapse
Affiliation(s)
- Natalia Tołoczko-Iwaniuk
- Department of Pharmaceutical Analysis, Medical University of Bialystok, Mickiewicza 2D Street, 15-222 Bialystok, Poland
| | - Dorota Dziemiańczyk-Pakieła
- Department of Maxillofacial and Plastic Surgery, Medical University of Bialystok, Skłodowskiej-Curie 24A, 15-404 Bialystok, Poland
| | - Beata Klaudia Nowaszewska
- Department of Maxillofacial and Plastic Surgery, Medical University of Bialystok, Skłodowskiej-Curie 24A, 15-404 Bialystok, Poland
| | - Katarzyna Celińska-Janowicz
- Department of Pharmaceutical Analysis, Medical University of Bialystok, Mickiewicza 2D Street, 15-222 Bialystok, Poland
| | - Wojciech Miltyk
- Department of Pharmaceutical Analysis, Medical University of Bialystok, Mickiewicza 2D Street, 15-222 Bialystok, Poland
| |
Collapse
|
3
|
Lu H, Wang B. SIRT1 exerts neuroprotective effects by attenuating cerebral ischemia/reperfusion-induced injury via targeting p53/microRNA-22. Int J Mol Med 2016; 39:208-216. [PMID: 27878231 DOI: 10.3892/ijmm.2016.2806] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 10/04/2016] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to investigate whether the SIRT1 exerts neuroprotective effects by attenuating cerebral ischemia/reperfusion-induced injury (CIRI) via targeting p53/microRNA-22. We found that the overexpression of sirtuin 1 (SIRT1) decreased the infarct volume, suppressed p53 protein expression and activated microRNA-22 expression following CIRI. An injection of lipopolysaccharide (LPS, 1 mg/ml; Sigma, St. Louis, MO USA) into the corpus callosum was used to induce CIRI in rats. The infarct volume and neurological deficit score were used to examine the effects of SIRT1 on CIRI. Furthermore, the overexpression of SIRT1 was found to suppress caspase-3 activity, inhibit the activation of the Bax signaling pathway, reduce tumor necrosis factor-α (TNF-α) and interleukin (IL)-6) activity, decrease cyclooxygenase (COX)‑2 and inducible nitric oxide synthase (iNOS) protein expression, and increase IL-10 activity following CIRI. Following the downregulation of SIRT1, p53 protein expression was significantly increased, microRNA-22 expression was inhibited, caspase-3 activity was increased and the Bax signaling pathway was activated. In addition, the activity of TNF-α and IL-6 was was enhanced, COX-2 and iNOS protein expression was increased, and IL-10 activity was reduced following CIRI. Thus, the data from our study suggest that SIRT1 attenuates CIRI by targeting the p53/microRNA-22 axix, while suppressing apoptosis, inflammation, COX-2 and iNOS expression.
Collapse
Affiliation(s)
- Hui Lu
- Department of Neurology, Cangzhou Central Hospital, Hebei 060000, P.R. China
| | - Bincheng Wang
- Department of Neurology, Xuan Wu Hospital, Beijing 100010, P.R. China
| |
Collapse
|
4
|
Jawanjal P, Salhan S, Dhawan I, Das N, Aggarwal R, Tripathi R, Rath G. Augmented Activity of Cyclooxygenase-2 in Tissue and Serum of Patients With Cervical Cancer. J Clin Lab Anal 2016; 30:1198-1207. [PMID: 27292107 DOI: 10.1002/jcla.22003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 04/21/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cyclooxygenase-2 (Cox-2) is frequently overexpressed in cervical carcinoma, but little is known about its altered serum concentration. Hence, this study evaluates clinical utility of cellular and serum level of Cox-2 enzyme in cervical cancer. METHODS The expression of Cox-2 was evaluated in cervical tissues and serum samples collected from normal controls (n = 100; n = 68), cervical intraepithelial neoplasia patients (CIN, n = 67; n = 12), and invasive squamous cell carcinoma patients (SCCs; n = 153; n = 127) by immunohistochemical and enzyme-linked immunosorbent assay (ELISA) analyses. RESULTS The significant cytoplasmic overexpression of Cox-2 was noted in 50.7% of CIN and 69.9% of SCCs as compared with normal (P = 0.0001). Serum level of Cox-2 was also found to be elevated both in CIN (median 4.35 ng/ml) and in SCCs (median 19.39 ng/ml) with respect to normal (median 0.44 ng/ml; P = 0.0001), respectively. The ROC analysis revealed the potential of serum Cox-2 over its cellular expression to distinguish CIN and SCCs from normal. CONCLUSION Augmented Cox-2 activity is implicated in the pathogenesis of cervical cancer, and its serum level could serve a potential to distinguish this malignancy. Therefore, it is suggested that serum Cox-2 may be useful in monitoring the diagnosis and treatment outcome of patients.
Collapse
Affiliation(s)
- Poonam Jawanjal
- Department of Anatomy, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Sudha Salhan
- Department of Obstetrics and Gynecology, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Indrani Dhawan
- Department of Histopathology, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Nirmalendu Das
- Department of Radiotherapy, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Ruby Aggarwal
- Department of Anatomy, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Richa Tripathi
- Division of Molecular Genetics & Biochemistry, Institute of Cytology and Preventive Oncology (ICPO), ICMR, Noida, India
| | - Gayatri Rath
- Department of Anatomy, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India. ,
| |
Collapse
|
5
|
Park J, Shim MK, Jin M, Rhyu MR, Lee Y. Methyl syringate, a TRPA1 agonist represses hypoxia-induced cyclooxygenase-2 in lung cancer cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:324-329. [PMID: 26969386 DOI: 10.1016/j.phymed.2016.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 12/21/2015] [Accepted: 01/14/2016] [Indexed: 06/05/2023]
Abstract
BACKGROUND We have previously found that methyl syringate is a specific and selective agonist of the human transient receptor potential channel ankyrin 1 (TRPA1) and suppresses food intake and gastric emptying in imprinting control region mice. Because TRPA1 has been implicated in inflammatory responses, and inflammation and tumorigenesis are stimulated by the cyclooxygenase-2 (COX-2)/prostaglandin E2 pathway in hypoxic cancer cells. PURPOSE This study examined the effects of methyl syringate on hypoxia-induced COX-2 in human distal lung epithelial A549 cells. STUDY DESIGN The effect of the methyl syringate on suppression of hypoxia-induced COX-2 in A549 cells were determined by Western blot and/or quantitative real-time polymerase chain reaction. The anti-invasive effect of methyl syringate was evaluated on A549 cells using matrigel invasion assay. RESULTS Methyl syringate suppressed hypoxia-induced COX-2 protein and mRNA expression and promoter activity and reduced hypoxia-induced cell migration and invasion and secretion of vascular endothelial growth factor. These effects were antagonized by a TRPA1 antagonist, implying their mediation by the TRPA1 pathway. CONCLUSION Together, these results indicate that methyl syringate inhibits the hypoxic induction of COX-2 expression and cell invasion through TRPA1 activation. These findings suggest that methyl syringate could be effective to suppress hypoxia-induced inflammation and indicate an additional functional effect of methyl syringate.
Collapse
Affiliation(s)
- Joonwoo Park
- Department of Bioscience and Biotechnology, College of Life Science, Sejong University, Kwangjingu, Kunjadong, Seoul 143-747, Republic of Korea
| | - Myeong Kuk Shim
- Department of Bioscience and Biotechnology, College of Life Science, Sejong University, Kwangjingu, Kunjadong, Seoul 143-747, Republic of Korea
| | - Mirim Jin
- Laboratory of Pharmacology, College of Korean Medicine, Daejeon University, Daejeon 301-724, Republic of Korea
| | - Mee-Ra Rhyu
- Division of Metabolism and Functionality Research, Korea Food Research Institute, Sungnam 463-746, Republic of Korea.
| | - YoungJoo Lee
- Department of Bioscience and Biotechnology, College of Life Science, Sejong University, Kwangjingu, Kunjadong, Seoul 143-747, Republic of Korea.
| |
Collapse
|
6
|
Jiang SP, Zhang ZD, Kang LM, Wang QH, Zhang L, Chen HP. Celecoxib reverts oxaliplatin-induced neuropathic pain through inhibiting PI3K/Akt2 pathway in the mouse dorsal root ganglion. Exp Neurol 2016; 275 Pt 1:11-6. [DOI: 10.1016/j.expneurol.2015.11.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 10/29/2015] [Accepted: 11/02/2015] [Indexed: 02/07/2023]
|
7
|
Casey SC, Amedei A, Aquilano K, Azmi AS, Benencia F, Bhakta D, Bilsland AE, Boosani CS, Chen S, Ciriolo MR, Crawford S, Fujii H, Georgakilas AG, Guha G, Halicka D, Helferich WG, Heneberg P, Honoki K, Keith WN, Kerkar SP, Mohammed SI, Niccolai E, Nowsheen S, Vasantha Rupasinghe HP, Samadi A, Singh N, Talib WH, Venkateswaran V, Whelan RL, Yang X, Felsher DW. Cancer prevention and therapy through the modulation of the tumor microenvironment. Semin Cancer Biol 2015; 35 Suppl:S199-S223. [PMID: 25865775 PMCID: PMC4930000 DOI: 10.1016/j.semcancer.2015.02.007] [Citation(s) in RCA: 251] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 02/06/2023]
Abstract
Cancer arises in the context of an in vivo tumor microenvironment. This microenvironment is both a cause and consequence of tumorigenesis. Tumor and host cells co-evolve dynamically through indirect and direct cellular interactions, eliciting multiscale effects on many biological programs, including cellular proliferation, growth, and metabolism, as well as angiogenesis and hypoxia and innate and adaptive immunity. Here we highlight specific biological processes that could be exploited as targets for the prevention and therapy of cancer. Specifically, we describe how inhibition of targets such as cholesterol synthesis and metabolites, reactive oxygen species and hypoxia, macrophage activation and conversion, indoleamine 2,3-dioxygenase regulation of dendritic cells, vascular endothelial growth factor regulation of angiogenesis, fibrosis inhibition, endoglin, and Janus kinase signaling emerge as examples of important potential nexuses in the regulation of tumorigenesis and the tumor microenvironment that can be targeted. We have also identified therapeutic agents as approaches, in particular natural products such as berberine, resveratrol, onionin A, epigallocatechin gallate, genistein, curcumin, naringenin, desoxyrhapontigenin, piperine, and zerumbone, that may warrant further investigation to target the tumor microenvironment for the treatment and/or prevention of cancer.
Collapse
Affiliation(s)
- Stephanie C Casey
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Asfar S Azmi
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Fabian Benencia
- Department of Biomedical Sciences, Ohio University, Athens, OH, United States
| | - Dipita Bhakta
- School of Chemical and Biotechnology, SASTRA University, Thanjavur 613401, Tamil Nadu, India
| | - Alan E Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Chandra S Boosani
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| | - Sophie Chen
- Ovarian and Prostate Cancer Research Laboratory, Guildford, Surrey, United Kingdom
| | | | - Sarah Crawford
- Department of Biology, Southern Connecticut State University, New Haven, CT, United States
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Gunjan Guha
- School of Chemical and Biotechnology, SASTRA University, Thanjavur 613401, Tamil Nadu, India
| | | | - William G Helferich
- University of Illinois at Urbana-Champaign, Champaign-Urbana, IL, United States
| | - Petr Heneberg
- Charles University in Prague, Third Faculty of Medicine, Prague, Czech Republic
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sid P Kerkar
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | | | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Graduate School, Mayo Medical School, Mayo Clinic, Rochester, MN, United States
| | - H P Vasantha Rupasinghe
- Department of Environmental Sciences, Faculty of Agriculture, Dalhousie University, Nova Scotia, Canada
| | | | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advanced Research), King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Wamidh H Talib
- Department of Clinical Pharmacy and Therapeutics, Applied Science University, Amman, Jordan
| | | | - Richard L Whelan
- Mount Sinai Roosevelt Hospital, Icahn Mount Sinai School of Medicine, New York City, NY, United States
| | - Xujuan Yang
- University of Illinois at Urbana-Champaign, Champaign-Urbana, IL, United States
| | - Dean W Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
8
|
Zhang C, Hu X, Liu XY, Liang P, Zhang J, Cao L, Wang ZL, Liu HR, Yin XG, Dong CY, Wang LM. Effect of tumor-associated macrophages on gastric cancer stem cell in omental milky spots and lymph node micrometastasis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:13795-13805. [PMID: 26823693 PMCID: PMC4713479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 10/24/2015] [Indexed: 06/05/2023]
Abstract
We observed whether the effect of tumor-associated macrophages on gastric cancer stem cell in omental milky spots and lymph nodes micrometastasis and research its possible mechanism. Macrophage THP-1 cells and Human gastric adenocarcinoma SGC-7901 cells were collectively cultivated in vivo. We found macrophage could suppress the proliferation and accelerated cell death of MFC cell. Meanwhile, these effects may be concerned with many signaling pathways, and we detected MCP-1 and COX-2 miRNA expressions, PGE-2 release levels, IL-4 and IL-10 activities, and TGF-β, IFN-γ, VEGF, MMP-2 and MMP-9 protein expressions in collectively cultivated cell. We found that MCP-1 and COX-2 miRNA expressions, and PGE-2 release levels were suppressed, IL-4 activity was inhibited and IL-10 activity was activated in collectively cultivated cell. Meanwhile, TGF-β, MMP-2 and MMP-9 protein expressions were inhibited and IFN-γ and VEGF protein expressions were activated in collectively cultivated cell. Taken together, these results suggest that the effect of tumor-associated macrophages on gastric cancer stem cell in omental milky spots and lymph nodes micrometastasis via COX-2/PGE-2/TGF-β/VEGF signal pathways.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Dalian Medical Universiey Dalian, China
| | - Xiang Hu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Dalian Medical Universiey Dalian, China
| | - Xiao-Yu Liu
- Liaoning Laboratory of Cancer Genetics and Epigenetics, Department of Cell Biology, Dalian Medical University Dalian, China
| | - Pin Liang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Dalian Medical Universiey Dalian, China
| | - Jian Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Dalian Medical Universiey Dalian, China
| | - Liang Cao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Dalian Medical Universiey Dalian, China
| | - Zheng-Lin Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Dalian Medical Universiey Dalian, China
| | - Huan-Ran Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Dalian Medical Universiey Dalian, China
| | - Xun-Guo Yin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Dalian Medical Universiey Dalian, China
| | - Cheng-Yong Dong
- Department of Hepatobiliary Pancreas Surgery, The Second Affiliated Hospital of Dalian Medical University Dalian, China
| | - Li-Ming Wang
- Department of Hepatobiliary Pancreas Surgery, The Second Affiliated Hospital of Dalian Medical University Dalian, China
| |
Collapse
|
9
|
Lim W, Shim MK, Kim S, Lee Y. Red ginseng represses hypoxia-induced cyclooxygenase-2 through sirtuin1 activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:597-604. [PMID: 26055124 DOI: 10.1016/j.phymed.2015.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 02/17/2015] [Accepted: 03/05/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND Korean red ginseng (KRG) is a traditional herbal medicine made by steaming and drying the fresh ginseng, leading to chemical transformation of some components by heat. It ameliorates various inflammatory diseases and strengthens the endocrine, immune, and central nervous systems. The cyclooxygenase-2 (COX-2)/prostaglandin E2 pathway in hypoxic cancer cells has important implications for stimulation of inflammation and tumorigenesis. PURPOSE In this study we examined the effects and the mechanism underlying Korean red ginseng water extract (KRG-WE) inhibition of hypoxia-induced COX-2 in human distal lung epithelial A549 cells. STUDY DESIGN The effect of the KRG on suppression of hypoxia-induced COX-2 in A549 cells were determined by Western blot and/or qRT-PCR. The anti-invasive effect of KRG-WE was evaluated on A549 cells using matrigel invasion assay. The activation of glucocorticoid receptor (GR) and sirtuin1 (Sirt1) was examined by using specific inhibitors. RESULTS We first observed that hypoxia induced COX-2 protein and mRNA levels and promoter activity were suppressed by KRG-WE. Second, we observed that hypoxia-induced cell migration is dramatically reduced by KRG-WE. Third, we found that the effect of KRG-WE was not antagonized by the GR antagonist RU486 implying that the effect is mediated other than GR pathway. Finally, we demonstrated that inhibition of Sirt1 abolished the effect of KRG-WE on hypoxia-induced COX-2 suppression and cell-invasion indicating that the suppression is mediated by Sirt1. CONCLUSION Taken together, KRG-WE inhibits the hypoxic induction of COX-2 expression and cell invasion through Sirt1 activation. Our results imply that KRG-WE could be effective for suppression of inflammation under hypoxia.
Collapse
Affiliation(s)
- Wonchung Lim
- Department of Bioscience and Biotechnology, College of Life Science, Sejong University, Kwangjingu, Kunjadong, Seoul 143-747, Korea; Department of Sports Medicine, College of Health Science, Cheongju University, Cheongju 363-764, Korea
| | - Myeong Kuk Shim
- Department of Bioscience and Biotechnology, College of Life Science, Sejong University, Kwangjingu, Kunjadong, Seoul 143-747, Korea
| | - Sikwan Kim
- Department of Biomedical Chemistry, Konkuk University, Chungju 380-701, Korea
| | - YoungJoo Lee
- Department of Bioscience and Biotechnology, College of Life Science, Sejong University, Kwangjingu, Kunjadong, Seoul 143-747, Korea.
| |
Collapse
|
10
|
Liu N, Chen L, Cai N. Celecoxib attenuates retinal angiogenesis in a mouse model of oxygen-induced retinopathy. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:4990-4998. [PMID: 26191192 PMCID: PMC4503064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 04/10/2015] [Indexed: 06/04/2023]
Abstract
This study aimed to investigate the anti-angiogenic effects of Celecoxib on the expression of vascular endothelial growth factor (VEGF) and hypoxia-inducible transcription factor 1α (HIF-1α) in a mouse model for oxygen-induced retinopathy (OIR). The OIR mice were exposed to 75% oxygen from postnatal day 7 (P7) to P12, after which the mice were randomly assigned to two groups (Celecoxib and vehicle) and were brought to room air for additional five days. Celecoxib or vehicle was administered from P12 to P17. Age-matched mice maintained in room air from birth to P17 were administered vehicle from P12 to P17 (RA group). Blood vessel profiles in the retina were used to count by histologic methods. Retina protein and mRNA of VEGF and HIF-1α were assessed by immunohistochemistry, western-blot and RT-PCR. Compared with the RA group, the OIR mice exhibited over-expression in VEGF and HIF-1α mRNA and protein. In addition, they had a positive and spatial correlation. Celecoxib- treated OIR mice reduced the retinal neovascular tufts and the levels of VEGF and HIF-1α. These data suggest that Celecoxib inhibits retinal pathogenic angiogenesis through down-regulating HIF-1α expression which suppressing VEGF transcription. Celecoxib could potentially serve as a portent pharmaceutical agent to inhibit retinal angiogenesis.
Collapse
Affiliation(s)
- Ningning Liu
- Department of Ophthalmology, The First Affiliated Hospital of China Medical University Shenyang, Liaoning Province, China
| | - Lei Chen
- Department of Ophthalmology, The First Affiliated Hospital of China Medical University Shenyang, Liaoning Province, China
| | - Na Cai
- Department of Ophthalmology, The First Affiliated Hospital of China Medical University Shenyang, Liaoning Province, China
| |
Collapse
|
11
|
Protective effect of boswellic acids versus pioglitazone in a rat model of diet-induced non-alcoholic fatty liver disease: influence on insulin resistance and energy expenditure. Naunyn Schmiedebergs Arch Pharmacol 2015; 388:587-600. [PMID: 25708949 DOI: 10.1007/s00210-015-1102-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Accepted: 02/03/2015] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is closely linked to insulin resistance, oxidative stress, and cytokine imbalance. Boswellic acids, a series of pentacyclic triterpene molecules that are produced by plants in the genus Boswellia, has been traditionally used for the treatment of a variety of diseases. This study aimed at evaluating the protective effect of boswellic acids in a model of diet-induced NAFLD in rats in comparison to the standard insulin sensitizer, pioglitazone. Rats were fed with a high-fat diet (HFD) for 12 weeks to induce NAFLD. Starting from week 5, rats received boswellic acids (125 or 250 mg/kg) or pioglitazone parallel to the HFD. Feeding with HFD induced hepatic steatosis and inflammation in rats. In addition, liver index, insulin resistance index, activities of liver enzymes, and serum lipids deviated from normal. Further, serum tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and cyclooxygenase 2 were elevated; this was associated with an increase in hepatic expression of inducible nitric oxide synthase (iNOS) and formation of 4-hydroxy-2-nonenal (HNE). Rats treated with boswellic acids (125 or 250 mg/kg) or pioglitazone showed improved insulin sensitivity and a reduction in liver index, activities of liver enzymes, serum TNF-α and IL-6 as well as hepatic iNOS expression and HNE formation compared to HFD group. Furthermore, at the cellular level, boswellic acids (250 mg/kg) ameliorated the expression of thermogenesis-related mitochondrial uncoupling protein-1 and carnitine palmitoyl transferase-1 in white adipose tissues. Data from this study indicated that boswellic acids might be a promising therapy in the clinical management of NAFLD if appropriate safety and efficacy data are available.
Collapse
|
12
|
Pantziarka P, Bouche G, Meheus L, Sukhatme V, Sukhatme VP. Repurposing drugs in oncology (ReDO)-cimetidine as an anti-cancer agent. Ecancermedicalscience 2014; 8:485. [PMID: 25525463 PMCID: PMC4268104 DOI: 10.3332/ecancer.2014.485] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Indexed: 12/19/2022] Open
Abstract
Cimetidine, the first H2 receptor antagonist in widespread clinical use, has anti-cancer properties that have been elucidated in a broad range of pre-clinical and clinical studies for a number of different cancer types. These data are summarised and discussed in relation to a number of distinct mechanisms of action. Based on the evidence presented, it is proposed that cimetidine would synergise with a range of other drugs, including existing chemotherapeutics, and that further exploration of the potential of cimetidine as an anti-cancer therapeutic is warranted. Furthermore, there is compelling evidence that cimetidine administration during the peri-operative period may provide a survival benefit in some cancers. A number of possible combinations with other drugs are discussed in the supplementary material accompanying this paper.
Collapse
Affiliation(s)
- Pan Pantziarka
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium ; The George Pantziarka TP53 Trust, London KT1 2JP, UK
| | | | - Lydie Meheus
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium
| | | | - Vikas P Sukhatme
- GlobalCures, Inc; Newton MA 02459, USA ; Beth Israel Deaconess Medical Centre and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
13
|
Sobolesky PM, Halushka PV, Garrett-Mayer E, Smith MT, Moussa O. Regulation of the tumor suppressor FOXO3 by the thromboxane-A2 receptors in urothelial cancer. PLoS One 2014; 9:e107530. [PMID: 25202904 PMCID: PMC4159332 DOI: 10.1371/journal.pone.0107530] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 08/19/2014] [Indexed: 01/27/2023] Open
Abstract
The transcription factor FOXO3 is a well-established tumor suppressor whose activity, stability, and localization are regulated by phosphorylation and acetylation. Previous data by our laboratory demonstrated amplified thromboxane-A2 signaling was associated with poor prognoses in bladder cancer patients and overexpression of the thromboxane-A2 isoform-β receptor (TPβ), but not TPα, induced malignant transformation of immortalized bladder cells in vivo. Here, we describe a mechanism of TP mediated modulation of FOXO3 activity and localization by phosphorylation and deacetylation in a bladder cancer cell model. In vitro gain and loss of function studies performed in non-transformed cell lines, UROsta and SV-HUC, revealed knockdown of FOXO3 expression by shRNA increased cell migration and invasion, while exogenously overexpressing TPβ raised basal phosphorylated (p)FOXO3-S294 levels. Conversely, overexpression of ERK-resistant, mutant FOXO3 reduced increases in UMUC3 cell migration and invasion, including that mediated by TP agonist (U46619). Additionally, stimulation of UMUC3 cells with U46619 increased pFOXO3-S294 expression, which could be attenuated by treatment with a TP antagonist (PTXA2) or ERK inhibitor (U0126). Initially U46619 caused nuclear accumulation of pFOXO3-S294; however, prolonged stimulation increased FOXO3 cytoplasmic localization. U46619 stimulation decreased overall FOXO3 transcriptional activity, but was associated with increased expression of its pro-survival target, manganese superoxide dismutase. The data also shows that TP stimulation increased the expression of the histone deacetylase, SIRT1, and corresponded with decreased acetylated-FOXO3. Collectively, the data suggest a role for TP signaling in the regulation of FOXO3 activity, mediated in part through phosphorylation and deacetylation.
Collapse
Affiliation(s)
- Philip M Sobolesky
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America; Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Perry V Halushka
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, United States of America; Departments of Pharmacology and Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Elizabeth Garrett-Mayer
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, United States of America; Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Michael T Smith
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Omar Moussa
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America; Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| |
Collapse
|