1
|
The expression of kappa-opioid receptor promotes the migration of breast cancer cells in vitro. BMC Anesthesiol 2021; 21:210. [PMID: 34461834 PMCID: PMC8404350 DOI: 10.1186/s12871-021-01429-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 08/18/2021] [Indexed: 12/17/2022] Open
Abstract
Background Opioid receptors are implicated in cell proliferation and cancer migration. However, the effects and underlying mechanisms of opioid receptor κ (OPRK1) in breast cancer remain unknown. Methods Small interfering RNA (siRNAs) was used to knockdown the expression of OPRK1. Western blot was used to determine the protein expression and reverse transcription-quantitative PCR (RT-qPCR) determined the genes transcription. Cell viability was detected by MTT assay and cell death rates were determined by Annexin V/PI and flow cytometry. Cell migration and invasion were detected by wound healing analysis and transwell assay, respectively. Results Our research demonstrated that OPRK1 was overexpressed in breast cancer cells compared with the normal human mammary epithelial cells. OPRK1 knockdown could inhibited cell viability and migration in cancer cells, accompanied with the decreased proteins and genes expression of N-cadherin, Snail, MMP2 and Vimentin, while the E-cadherin expression was increased. Additionally, OPRK1 knockdown also promoted PI3K/AKT signaling inactivation. Activation of AKT reversed the OPRK1 knockdown-induced cell viability inhibition and migration suppression, while inhibition of AKT reduced cell viability and promoted cell death. Conclusions Our findings illustrated the role of OPRK1 played on promoting migration in vitro, and we also provided the therapeutic research of OPRK1 knockdown combined with AKT inhibition.
Collapse
|
2
|
Zeng W, Xing Z, Tan M, Wu Y, Zhang C. Propofol regulates activated macrophages metabolism through inhibition of ROS-mediated GLUT1 expression. Inflamm Res 2021; 70:473-481. [PMID: 33751130 DOI: 10.1007/s00011-021-01449-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 02/02/2021] [Accepted: 03/04/2021] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE Activated macrophages undergo a metabolic shift from oxidative phosphorylation (OXPHOS) to aerobic glycolysis, which plays a critical role in inflammation. Increasing evidence suggests the important role of propofol in the regulation of inflammatory response and metabolism, but the effect of propofol on the metabolic shift in macrophage, and the mechanisms involved remain unclear. METHODS The effect of propofol on the metabolic switch was analyzed by extracellular acidification rate and oxygen consumption rate assays. The effect of propofol on glycolysis was analyzed by lactate and glucose uptake assay. The mRNA, protein, cell surface levels of glucose transporter 1 (GLUT1) and the silencing of GLUT1 were employed to understand the effects of GLUT1-mediated metabolism by propofol. Finally, to understand the antioxidation of propofol on the regulation of metabolism, the reactive oxygen species (ROS) production and NADPH activity were performed. RESULTS We show that propofol can change the metabolic pathway switch from aerobic glycolysis to OXPHOS in LPS-activated macrophages. Moreover, propofol suppresses aerobic glycolysis via inhibited GLUT1-mediated glucose uptake. Furthermore, we show that propofol reduces ROS overproduction, which in turn inhibits GLUT1 expression. Finally, we find that propofol reduces ROS production via inhibits NADPH activity. CONCLUSION These findings shed light on the function and mechanism of propofol in the metabolic switch and highlight the importance of targeting metabolism by propofol in the clinical medication of inflammatory diseases.
Collapse
Affiliation(s)
- Wei Zeng
- Department of Anesthesiology, Affiliated Boai Hospital of Zhongshan, Southern Medical University, Guangdong, 528400, China
| | - Zeting Xing
- Department of Anesthesiology, Affiliated Boai Hospital of Zhongshan, Southern Medical University, Guangdong, 528400, China
| | - Meiyun Tan
- Department of Anesthesiology, Affiliated Boai Hospital of Zhongshan, Southern Medical University, Guangdong, 528400, China
| | - Yanwen Wu
- Department of Anesthesiology, Affiliated Boai Hospital of Zhongshan, Southern Medical University, Guangdong, 528400, China
| | - Chunyuan Zhang
- Department of Anesthesiology, Affiliated Boai Hospital of Zhongshan, Southern Medical University, Guangdong, 528400, China.
| |
Collapse
|
3
|
Wang Z, Zhou S. Effect of Compound Propofol Nanoemulsion on Immune Function in Patients with Pancreatic Cancer. JOURNAL OF NANOSCIENCE AND NANOTECHNOLOGY 2021; 21:1390-1396. [PMID: 33183489 DOI: 10.1166/jnn.2021.18661] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Pancreatic cancer is a common malignant tumor of the digestive system, and its incidence is increasing worldwide. The treatment of pancreatic cancer is mainly surgery, but the development of traditional surgery, chemoradiotherapy has entered the plateau stage, and immunotherapy has received more and more attention. Different anesthesia methods, anaesthetic drugs and their preparations have different effects on the perioperative immune function of patients with pancreatic cancer, and even affect the long-term prognosis of patients. Suppression of immune function caused by surgery and anesthesia may promote tumor recurrence and metastasis, thereby affecting the longterm prognosis of patients. The use of propofol intravenous anesthesia during surgery can reduce pro-inflammatory factors and increase anti-inflammatory factors, thereby effectively improving the immune function of perioperative tumor patients. At the same time, using nanoemulsion as a carrier in a drug delivery system can improve the solubility and loading capacity of poorly soluble drugs, and increase the absorption rate and bioavailability of fat-soluble drugs in the body. Therefore, in this study, a combination of propofol nano-injection and remifentanil injection liquid combined with propofol nanoemulsion preparation and propofol injection alone was used in the operation of pancreatic cancer patients. By comparing the two groups of patients Basic indicators during and after surgery, hemorheology levels, lymphocyte subsets, and proinflammatory cytokines, to study the use of the compound propofol nanoemulsion preparation in patients with pancreatic cancer during surgery Effects of immune function. The results showed that compound propofol nanoemulsion can improve the immune function of patients with pancreatic cancer during surgery, reduce the level of inflammatory factors, and has a good prognosis and high safety.
Collapse
Affiliation(s)
- Zhihua Wang
- Department of Anesthesia, Linyi Central Hospital, Linyi City, 276400, Shandong Province, China
| | - Shengcai Zhou
- Department of Anesthesia, Linyi Central Hospital, Linyi City, 276400, Shandong Province, China
| |
Collapse
|
4
|
Teng WN, Wu HL, Tai YH, Lei HJ, Tsou MY, Chang KY. Group-based trajectory analysis of postoperative pain and outcomes after liver cancer surgery. J Chin Med Assoc 2021; 84:95-100. [PMID: 33177401 DOI: 10.1097/jcma.0000000000000446] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Although previous studies have shown connections between pain and worse cancer outcomes, few clinical studies have evaluated their direct association, and the current study aimed to investigate the potential association between acute pain trajectories and postoperative outcomes after liver cancer surgery. METHODS This retrospective study was conducted in a single medical center and included patients who received liver cancer surgery between January 2010 and December 2016. Maximal pain intensity was recorded daily using a numerical rating scale during the first postoperative week. Group-based trajectory analysis was performed to classify the variations in pain scores over time. Cox and linear regression analyses were used to assess the effect of pain trajectories on recurrence-free survival, overall survival, and length of hospital stay (LOS) after surgery and to explore predictors of these outcomes. RESULTS A total of 804 patients with 5396 pain score observations were analyzed within the present study. Group-based trajectory analysis categorized the changes in postoperative pain into three groups: group 1 had constantly mild pain (76.6%), group 2 had moderate/severe pain dropping to mild (10.1%), and group 3 had mild pain rebounding to moderate (13.3%). Multivariable analysis demonstrated that on average, group 3 had a 7% increase in LOS compared with the group 1 (p = 0.02) and no significant difference in the LOS was noted between pain trajectory groups 2 and 1 (p = 0.93). Pain trajectories were not associated with recurrence-free survival or overall survival after liver cancer surgery. CONCLUSION Acute pain trajectories were associated with LOS but not cancer recurrence and survival after liver cancer surgery. Group-based trajectory analysis provided a promising approach for investigating the complex relationships between variations in postoperative pain over time and clinical outcomes.
Collapse
Affiliation(s)
- Wei-Nung Teng
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Hsiang-Ling Wu
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Ying-Hsuan Tai
- Department of Anesthesiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan, ROC
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Hao-Jan Lei
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Mei-Yung Tsou
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Kuang-Yi Chang
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| |
Collapse
|
5
|
A randomized pilot study to investigate the effect of opioids on immunomarkers using gene expression profiling during surgery. Pain 2020; 160:2691-2698. [PMID: 31433352 DOI: 10.1097/j.pain.0000000000001677] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Endogenous opioid peptides and exogenous opioids modulate immune function, and animal and human studies have shown that some have a depressant immunomodulatory effect. This is potentially of high clinical significance, eg, in cancer patients and surgery. The primary objective of this pilot study was to evaluate the effect of morphine and oxycodone on immune pathways associated with immunosuppression in gynecological laparotomy patients. Gene expression was analyzed in CD4, CD8, and natural killer (NK) cells using the 3' Affymetrix microarray. Patients were randomized to receive morphine, oxycodone, or nonopioid "control" analgesia during and after surgery. Genes demonstrating differential expression were those with a ≥±2-fold difference and P-value ≤0.05 after analysis of variance. Cytometric bead array and NK cell degranulation assay were used to investigate changes in serum cytokine concentration and in NK cell cytotoxicity, respectively. Forty patients had satisfactory RNA which was hybridized to gene chips. Genes were identified (Partek Genomics Suite 6.6) at baseline, 2, 6, and 24 hours and were either ≥2-fold upregulated or downregulated from baseline. At 2 hours, a large number of genes were downregulated with morphine but not with control analgesia or oxycodone. Statistically significant increases in IL-6 concentrations were induced by morphine only; NK cell activity was suppressed with morphine, but maintained with oxycodone and epidural analgesia. Gene expression profiles suggest that at 2 hours, post incision morphine appeared to be immunosuppressive as compared to oxycodone and nonopioid control analgesia.
Collapse
|
6
|
Wang D, Yang T, Liu J, Liu Y, Xing N, He J, Yang J, Ai Y. Propofol Inhibits the Migration and Invasion of Glioma Cells by Blocking the PI3K/AKT Pathway Through miR-206/ROCK1 Axis. Onco Targets Ther 2020; 13:361-370. [PMID: 32021281 PMCID: PMC6969687 DOI: 10.2147/ott.s232601] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/19/2019] [Indexed: 12/12/2022] Open
Abstract
Background Propofol has been identified to perform anti-tumor functions in glioma. However, the molecular mechanisms underlying propofol-induced prevention on migration and invasion of glioma cells remain unclear. Methods Cell proliferation, invasion and migration were measured by 3-(4,5)-dimethylthiahiazo(−z-y1)-3,5-di-phenytetrazoliumromide assay and transwell assay, respectively. The expression of microRNA (miR)-206 and Rho-associated coiled coil-containing protein kinase 1 (ROCK1) was detected by quantitative real-time polymerase chain reaction. Western blot was used to measure the activation of the PI3K/AKT pathway. The interaction between miR-206 and ROCK1 was analyzed using the dual-luciferase reporter assay, RNA immunoprecipitation assay, and pull-down assay. Results Propofol treatment inhibited the migration, invasion, and PI3K/AKT pathway activation in glioma cells. MiR-206 was decreased in glioma tissues and cells, while propofol exposure induced the upregulation of miR-206 in glioma cells. Besides that, we also found overexpressed miR-206 enhanced propofol-mediated inhibition on the migration, invasion, and PI3K/AKT pathway activation of glioma cells. Subsequently, ROCK1 was confirmed to be a target of miR-206. ROCK1 was elevated in glioma tissues and cells, but was reduced by propofol exposure in glioma cells. The rescue assay indicated that the miR-206/ROCK1 axis was involved in propofol-induced inhibition on the migration, invasion, and PI3K/AKT pathway activation in glioma cells. Conclusion Propofol inhibited the migration and invasion of glioma cells by blocking the PI3K/AKT pathway through the miR-206/ROCK1 axis, suggesting an effective clinical implication for the anesthetic to prevent the metastasis of glioma.
Collapse
Affiliation(s)
- Dongmei Wang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Tao Yang
- Department of Anesthesiology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Junqi Liu
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Yafei Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Juan He
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Jianjun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Yanqiu Ai
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| |
Collapse
|
7
|
Neuroepithelial Cell Transforming Gene 1 Acts as an Oncogene and Is Mediated by miR-22 in Human Non-Small-Cell Lung Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1648419. [PMID: 32420320 PMCID: PMC7201585 DOI: 10.1155/2020/1648419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/13/2019] [Accepted: 12/18/2019] [Indexed: 02/08/2023]
Abstract
Abnormal expression of neuroepithelial cell transforming gene 1 (NET1) has been authenticated in many human cancers, including lung cancer. We have previously reported that NET1 functioned as an oncogene and promoted human non-small-cell lung cancer (NSCLC) growth and migration. However, the correlation between NET1 and its upstream miRNAs needed further illustration. Our present work demonstrated that miR-22 had a relatively low expression, and NET1 had a relatively high expression in both NSCLC samples and lung adenocarcinoma cell lines compared with corresponding normal controls. Moreover, miR-22 directly regulated NET1 and was verified to weaken cancer cell proliferation and migration, as well as enhance cell apoptosis by suppressing NET1. Furthermore, the inhibitory effect of miR-22 can be reversed via overexpressing NET1 using an ectopic expression vector in NSCLC cells. Our findings showed that miR-22/NET-1 axis may contribute to the inhibition of NSCLC growth and migration and represents a promising therapeutic target for NSCLC.
Collapse
|
8
|
Li G, Zhou Q, Song J, Xiao L, Zhu Y, Hou B. Propofol inhibits proliferation and invasion of endometriotic cells by miR-9-5p/TGFBI axis. ALL LIFE 2020. [DOI: 10.1080/26895293.2020.1789761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- Gang Li
- Department of Anesthesiology, Maternal and Child Health Hospital of Jiangxi Province, Nanchang, People’s Republic of China
| | - Qun Zhou
- Department of Anesthesiology, Maternal and Child Health Hospital of Jiangxi Province, Nanchang, People’s Republic of China
| | - Jie Song
- Department of Anesthesiology, Maternal and Child Health Hospital of Jiangxi Province, Nanchang, People’s Republic of China
| | - Lihua Xiao
- Department of Neurology, the Fourth Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Yirong Zhu
- Department of Gynaecology, Maternal and Child Health Hospital of Jiangxi Province, Nanchang, People’s Republic of China
| | - Benchao Hou
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| |
Collapse
|
9
|
Hamy AS, Derosa L, Valdelièvre C, Yonekura S, Opolon P, Priour M, Guerin J, Pierga JY, Asselain B, De Croze D, Pinheiro A, Lae M, Talagrand LS, Laas E, Darrigues L, Grandal B, Marangoni E, Montaudon E, Kroemer G, Zitvogel L, Reyal F. Comedications influence immune infiltration and pathological response to neoadjuvant chemotherapy in breast cancer. Oncoimmunology 2019; 9:1677427. [PMID: 32002287 PMCID: PMC6959439 DOI: 10.1080/2162402x.2019.1677427] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 09/18/2019] [Accepted: 10/04/2019] [Indexed: 12/22/2022] Open
Abstract
Immunosurveillance plays an important role in breast cancer (BC) prognosis and progression, and can be geared by immunogenic chemotherapy. In a cohort of 1023 BC patients treated with neoadjuvant chemotherapy (NAC), 40% of the individuals took comedications mostly linked to aging and comorbidities. We systematically analyzed the off-target effects of 1178 concurrent comedications (classified according to the Anatomical Therapeutic Chemical (ATC) Classification System) on the density of tumor-infiltrating lymphocytes (TILs) and pathological complete responses (pCR). At level 1 of the ATC system, the main anatomical classes of drugs were those targeting the nervous system (class N, 39.1%), cardiovascular disorders (class C, 26.6%), alimentary and metabolism (class A, 16.9%), or hormonal preparations (class H, 6.5%). At level 2, the most frequent therapeutic classes were psycholeptics (N05), analgesics (N02), and psychoanaleptics (N06). Pre-NAC TIL density in triple-negative BC (TNBC) was influenced by medications from class H, N, and A, while TIL density in HER2+ BC was associated with the use of class C. Psycholeptics (N05) and agents acting on the renin-angiotensin system (C09) were independently associated with pCR in the whole population of BC or TNBC, and in HER2-positive BC, respectively. Importantly, level 3 hypnotics (N05C) alone were able to reduce tumor growth in BC bearing mice and increased the anti-cancer activity of cyclophosphamide in a T cell-dependent manner. These findings prompt for further exploration of drugs interactions in cancer, and for prospective drug-repositioning strategies to improve the efficacy of NAC in BC.
Collapse
Affiliation(s)
- Anne-Sophie Hamy
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Translational Research Department, U932, Immunity and Cancer, Institut Curie, PSL Research University, Paris, France.,Department of Medical Oncology, Institut Curie, Saint-Cloud, France
| | - Lisa Derosa
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France.,Institut National de la Santé Et de la Recherche Medicale (INSERM), Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France.,Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | | | - Satoru Yonekura
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France.,Institut National de la Santé Et de la Recherche Medicale (INSERM), Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France.,Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Paule Opolon
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France.,Institut National de la Santé Et de la Recherche Medicale (INSERM), Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France.,Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Maël Priour
- Informatics Department, Institut Curie, Paris, France
| | - Julien Guerin
- Informatics Department, Institut Curie, Paris, France
| | - Jean-Yves Pierga
- Department of Medical Oncology, Institut Curie, Saint-Cloud, France
| | | | | | - Alice Pinheiro
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Translational Research Department, U932, Immunity and Cancer, Institut Curie, PSL Research University, Paris, France
| | - Marick Lae
- Tumor Biology, Institut Curie, Paris, France
| | | | - Enora Laas
- Department of Surgery, Institut Curie, Paris, France
| | | | | | - Elisabetta Marangoni
- Preclinical investigation laboratory, Translational Research Department, Institut Curie, PSL Research University, Paris
| | - Elodie Montaudon
- Preclinical investigation laboratory, Translational Research Department, Institut Curie, PSL Research University, Paris
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Cell Biology and Metabolomics platforms, Villejuif, France.,INSERM, Paris, France.,Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France.,Institut National de la Santé Et de la Recherche Medicale (INSERM), Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France.,Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Gustave Roussy and Institut Curie, France
| | - Fabien Reyal
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Translational Research Department, U932, Immunity and Cancer, Institut Curie, PSL Research University, Paris, France.,Department of Surgery, Institut Curie, Paris, France.,Sorbonne Paris Cité, Université Paris Descartes, Paris, France
| |
Collapse
|
10
|
Zhang Z, Zang M, Wang S, Wang C. Effects of propofol on human cholangiocarcinoma and the associated mechanisms. Exp Ther Med 2018; 17:472-478. [PMID: 30651824 DOI: 10.3892/etm.2018.6908] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/26/2018] [Indexed: 12/17/2022] Open
Abstract
Cholangiocarcinoma (CCA) is the most common type of biliary duct malignancy. Propofol is a fast-acting intravenous anesthetic, which also exerts an anti-cancer effect. The aim of the current study was to explore the effects of propofol on human CCA and the associated mechanisms in vitro. The results indicated that as concentration (0, 1, 5 and 10 µg/ml) of propofol and treatment time (24, 48 and 72 h) increased, the cell inhibition rate of human CCA QBC939 cells increased. Furthermore, treatment with various concentrations of propofol for 48 h resulted in a decrease in migration and invasion capacity in QBC939 cells. Propofol also induced the apoptosis of QBC939 cells and cell cycle arrest in G1 phase. Propofol treatment increased the expression level of Bax and decreased that of Bcl-2. In addition, the effects of propofol on gene expression were evaluated, including Wnt3α, β-catenin, Snail1 and c-myc in the Wnt/β-catenin signaling pathway. It was identified that as the concentration of propofol increased, the expression of these genes decreased. In conclusion, the current results indicate that propofol is a promising therapeutic agent for the treatment of CCA.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Hepatobiliary and Pancreas Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Mingcui Zang
- Department of Hepatobiliary and Pancreas Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Shuang Wang
- Department of Hepatobiliary and Pancreas Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Chunli Wang
- Department of Hepatobiliary and Pancreas Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
11
|
Li X, Li L, Liang F, Liu G, Zhao G. Anesthetic drug propofol inhibits the expression of interleukin-6, interleukin-8 and cyclooxygenase-2, a potential mechanism for propofol in suppressing tumor development and metastasis. Oncol Lett 2018; 15:9523-9528. [PMID: 29805673 DOI: 10.3892/ol.2018.8515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 05/11/2017] [Indexed: 11/06/2022] Open
Abstract
Previous studies have indicated that anesthesia-associated drugs may directly inhibit cellular immunity and humoral immunity, which may be associated with tumor recurrence. The present study demonstrated that propofol may suppress the proliferation of MCF-7 cells and inhibit the expression of interleukin (IL)-6 and IL-8. Subsequent to treatment with propofol, MCF-7 cells demonstrated downregulated cyclooxygenase-2 (COX-2) protein expression and decreased levels of vascular endothelial growth factor and prostaglandin E2 in the supernatant. Therefore, the mechanism of propofol in suppressing tumor development and metastasis may be associated with the inhibition of IL-6, IL-8 and COX-2.
Collapse
Affiliation(s)
- Xuefeng Li
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Longyun Li
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Feng Liang
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Guifeng Liu
- Department of Radiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Guoqing Zhao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
12
|
Xu W, Zheng J, Bie S, Kang L, Mao Q, Liu W, Guo J, Lu J, Xia R. Propofol inhibits Wnt signaling and exerts anticancer activity in glioma cells. Oncol Lett 2018; 16:402-408. [PMID: 29928428 DOI: 10.3892/ol.2018.8606] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 12/11/2017] [Indexed: 11/06/2022] Open
Abstract
Aberrant activation of Wnt signaling is implicated in gliomagenesis. Propofol, the most commonly used intravenous anesthetic agent in clinics, exhibits potent antitumor activity in a variety of cancer cells through different mechanisms. However, the role of propofol on Wnt signaling and glioma cell growth remains to be fully elucidated. In the present study, propofol was identified as a potent inhibitor of Wnt signaling. In 293T cells transfected with Wnt1 or Wnt3 expression plasmids or treated with Wnt3A-conditioned medium, propofol significantly inhibited the transcriptional activity of the SuperTopFlash reporter and the expression of Wnt target genes. The inhibitory effect of propofol on Wnt signaling was also observed in glioma cells. Further experiments demonstrated that propofol suppressed glioma cell growth by decreasing cell proliferation and enhancing cell apoptosis. Finally, the potential antitumor efficiency of propofol was confirmed using xenograft experiments in vivo. Taken together, the results indicated a novel mechanism for the anticancer activity of propofol and provide supporting evidence for its use as a prospective anticancer drug to treat glioma in patients with deregulated Wnt signaling.
Collapse
Affiliation(s)
- Wei Xu
- Department of Anesthesiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Jiwei Zheng
- Department of Anesthesiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Shijie Bie
- Department of Anesthesiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Liuyu Kang
- Department of Anesthesiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Qingjun Mao
- Department of Anesthesiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Weiwei Liu
- Department of Anesthesiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Jinxin Guo
- Department of Anesthesiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Juan Lu
- Operating Room, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Rui Xia
- Department of Anesthesiology, The First People's Hospital of Jingzhou, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|
13
|
Deng F, Ouyang M, Wang X, Yao X, Chen Y, Tao T, Sun X, Xu L, Tang J, Zhao L. Differential role of intravenous anesthetics in colorectal cancer progression: implications for clinical application. Oncotarget 2018; 7:77087-77095. [PMID: 27780923 PMCID: PMC5363570 DOI: 10.18632/oncotarget.12800] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 10/17/2016] [Indexed: 01/05/2023] Open
Abstract
Anesthetics are unavoidable to colorectal cancer (CRC) patients who underwent surgical treatment. Thus, the molecular mechanisms underlying the role of the intravenous anesthetics in CRC metastasis are still unclear. In this study, the effects of intravenous anesthetics, such as propofol, etomidate and dexmedetomidine, on cell migration were determined. The migration of CRC cells was inhibited by propofol in vitro, but not in vivo. Etomidate, however, promoted the migration of CRC cells both in vitro and in vivo. Epithelial-mesenchymal transition (EMT) mediated the promotive effect of propofol and etomidate on the migration of CRC cells through PI3K/AKT signaling pathway. Dexmedetomidine alone or in combination with propofol or etomidate had minor effect on the migration of CRC cells. These findings indicate that propofol inhibites CRC cell migration in vitro. Etomidate playes a role for prompting CRC metastasis progression by activating (PI3K)/AKT signaling and inducing EMT. It provides an important hint for the clinical application of these anesthetics.
Collapse
Affiliation(s)
- Fengliu Deng
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingwen Ouyang
- Department of anesthesia, Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaofei Wang
- Department of Anesthesia, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xueqing Yao
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou, Guangdong, China
| | - Yeming Chen
- Department of Anesthesia, Huarui Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Tao Tao
- Department of Anesthesia, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xuegang Sun
- The Key Laboratory of Molecular Biology, State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Lijun Xu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Tang
- Department of Anesthesia, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Liang Zhao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Zhou CL, Li JJ, Ji P. Propofol Suppresses Esophageal Squamous Cell Carcinoma Cell Migration and Invasion by Down-Regulation of Sex-Determining Region Y-box 4 (SOX4). Med Sci Monit 2017; 23:419-427. [PMID: 28118321 PMCID: PMC5286919 DOI: 10.12659/msm.899732] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background This study was done to verify whether propofol could inhibit esophageal squamous cell carcinoma (ESCC) cell line EC9706 cell migration and invasion by targeting SOX4. Material/Methods Different concentrations of propofol were co-incubated with EC9706 cells. The pcDNA-SOX4 or SOX4 siRNA plasmid was transfected into cells before the treatment with propofol 5 μg/L. The migratory and invasion ability of EC9706 cells were tested by wound-healing assay and Transwell chambers. Western blotting was used to investigate the expressions of MMP-2, MMP-9, TIMP-1, TIMP-2, and SOX4. Gelatin zymography was employed to detect the activity of MMP2 and MMP-9. Results Compared with the control, the migration and invasion activity of EC9706 cells were decreased after incubation with different concentrations of propofol (P<0.01). The expression of MMP-2, MMP-9, and SOX4 was decreased and that of TIMP-1 was increased in the propofol-treated EC9706 cells (P<0.01). Down-regulation of SOX4 by SOX4-siRNA had the same effect as propofol on EC9706 cells, including suppressing cell migration and invasion, inhibiting the expression and activity of MMP-2/9, and increasing the expression TIMP-1. Over-expression of SOX4 could partly abrogated propofol-mediated inhibition of EC9706 cell migration and invasion. Conclusions Propofol inhibits EC9706 cell migration and invasion by down-regulation of SOX4.
Collapse
Affiliation(s)
- Chun-Li Zhou
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China (mainland)
| | - Jing-Jing Li
- Department of Ophthalmology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China (mainland)
| | - Peng Ji
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China (mainland)
| |
Collapse
|
15
|
Chen X, Zhong SL, Lu P, Wang DD, Zhou SY, Yang SJ, Shen HY, Zhang L, Zhang XH, Zhao JH, Tang JH. miR-4443 Participates in the Malignancy of Breast Cancer. PLoS One 2016; 11:e0160780. [PMID: 27504971 PMCID: PMC4978484 DOI: 10.1371/journal.pone.0160780] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/25/2016] [Indexed: 01/25/2023] Open
Abstract
PURPOSE Chemo-resistance is the leading cause of failure in cancer therapy, however, much remains to be understood about the intrinsic mechanisms. In the present study, we discovered the novel miR-4443 that regulated malignancy of breast cancer both in vitro and in vivo. METHODS We examined the expression of miR-4443 in MDA-MB-231/S and MDA-MB-231 Epirubicin-resistant cell lines with 76 breast cancer formalin-fixed paraffin-embedded tissues by real-time PCR. Also, we investigated the loss- and gain-functions of miR-4443 by MTT assay and flow cytometry. Furthermore, we detected miR-4443 mediated tissue inhibitor of metalloproteinase 2 expression in cells by TargetScan, RT-qPCR and western blot. RESULTS We identified the up-regulated expression of miR-4443 in Epi-resistant cell lines versus MDA-MB-231/S cell(Epi versus S) and in post-chemotherapy FFPE tissues, along with statistically differential expressions in PR(partial response) versus SD(stable disease)/PD(progressive disease) patients. Overexpression of miR-4443 increased the IC50 value of Epi for the target cells transfected, while inhibition of miR-4443 could restored sensitivity of the target cells to Epi. Besides, down-regulation of endogenous miR-4443 by miRNA-inhibitors significantly enhanced Epi-induced apoptosis while up-regulation of miR-4443 by miRNA-mimics lead to less Epi-induced apoptotic cells. Consequently, changes in TIMP2 mRNA and protein expression revealed that miR-4443 mimics suppressed expression of TIMP2 and induced migration in breast cancer cells. Furthermore, TIMP2 expression associated with better prognosis(HR = 0.721, 95%CI: 0.529-0.983). CONCLUSIONS We revealed that miR-4443 induced malignancy of breast cancer mainly in chemo-resistance aspect for the very first time, providing a novel biomarker in breast cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Xiu Chen
- The Fourth Clinical School of Nanjing Medical University, Nanjing, China
- Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Shan-liang Zhong
- Center of Clinical Laboratory Science, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Peng Lu
- School of Public Health Nanjing Medical University, Nanjing, China
| | - Dan-dan Wang
- Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Si-ying Zhou
- Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Su-jin Yang
- Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Hong-yu Shen
- Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Lei Zhang
- Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Xiao-hui Zhang
- Center of Clinical Laboratory Science, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Jian-hua Zhao
- Center of Clinical Laboratory Science, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, China
- * E-mail: (JHT); (JHZ)
| | - Jin-hai Tang
- Department of General Surgery, Jiangsu Cancer Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Department of General Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- * E-mail: (JHT); (JHZ)
| |
Collapse
|