1
|
Alkhathami AG, Sahib AS, Al Fayi MS, Fadhil AA, Jawad MA, Shafik SA, Sultan SJ, Almulla AF, Shen M. Glycolysis in human cancers: Emphasis circRNA/glycolysis axis and nanoparticles in glycolysis regulation in cancer therapy. ENVIRONMENTAL RESEARCH 2023; 234:116007. [PMID: 37119844 DOI: 10.1016/j.envres.2023.116007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 06/19/2023]
Abstract
The metabolism of cancer has been an interesting hallmark and metabolic reprogramming, especially the change from oxidative phosphorylation in mitochondria to glucose metabolism known as glycolysis occurs in cancer. The molecular profile of glycolysis, related molecular pathways and enzymes involved in this mechanism such as hexokinase have been fully understood. The glycolysis inhibition can significantly decrease tumorigenesis. On the other hand, circRNAs are new emerging non-coding RNA (ncRNA) molecules with potential biological functions and aberrant expression in cancer cells which have received high attention in recent years. CircRNAs have a unique covalently closed loop structure which makes them highly stable and reliable biomarkers in cancer. CircRNAs are regulators of molecular mechanisms including glycolysis. The enzymes involved in the glycolysis mechanism such as hexokinase are regulated by circRNAs to modulate tumor progression. Induction of glycolysis by circRNAs can significantly increase proliferation rate of cancer cells given access to energy and enhance metastasis. CircRNAs regulating glycolysis can influence drug resistance in cancers because of theirimpact on malignancy of tumor cells upon glycolysis induction. TRIM44, CDCA3, SKA2 and ROCK1 are among the downstream targets of circRNAs in regulating glycolysis in cancer. Additionally, microRNAs are key regulators of glycolysis mechanism in cancer cells and can affect related molecular pathways and enzymes. CircRNAs sponge miRNAs to regulate glycolysis as a main upstream mediator. Moreover, nanoparticles have been emerged as new tools in tumorigenesis suppression and in addition to drug and gene delivery, then mediate cancer immunotherapy and can be used for vaccine development. The nanoparticles can delivery circRNAs in cancer therapy and they are promising candidates in regulation of glycolysis, its suppression and inhibition of related pathways such as HIF-1α. The stimuli-responsive nanoparticles and ligand-functionalized ones have been developed for selective targeting of glycolysis and cancer cells, and mediating carcinogenesis inhibition.
Collapse
Affiliation(s)
- Ali G Alkhathami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Ameer S Sahib
- Department of Pharmacy, Al- Mustaqbal University College, 51001 Hilla, Iraq
| | - Majed Saad Al Fayi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | - Mohammed Abed Jawad
- Department of Medical Laboratories Technology, Al-Nisour University College, Iraq
| | - Sahar Ahmad Shafik
- Professor of Community Health Nursing, Faculty of Nursing, Fayum University, Egypt; College of Nursing, National University of Science and Technology, Iraq
| | | | - Abbas F Almulla
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Min Shen
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, China.
| |
Collapse
|
2
|
Frank M, Nabb AT, Gilbert SP, Bentley M. Propofol attenuates kinesin-mediated axonal vesicle transport and fusion. Mol Biol Cell 2022; 33:ar119. [PMID: 36103253 PMCID: PMC9634964 DOI: 10.1091/mbc.e22-07-0276] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Propofol is a widely used general anesthetic, yet the understanding of its cellular effects is fragmentary. General anesthetics are not as innocuous as once believed and have a wide range of molecular targets that include kinesin motors. Propofol, ketamine, and etomidate reduce the distances that Kinesin-1 KIF5 and Kinesin-2 KIF3 travel along microtubules in vitro. These transport kinesins are highly expressed in the CNS, and their dysfunction leads to a range of human pathologies including neurodevelopmental and neurodegenerative diseases. While in vitro data suggest that general anesthetics may disrupt kinesin transport in neurons, this hypothesis remains untested. Here we find that propofol treatment of hippocampal neurons decreased vesicle transport mediated by Kinesin-1 KIF5 and Kinesin-3 KIF1A ∼25-60%. Propofol treatment delayed delivery of the KIF5 cargo NgCAM to the distal axon. Because KIF1A participates in axonal transport of presynaptic vesicles, we tested whether prolonged propofol treatment affects synaptic vesicle fusion mediated by VAMP2. The data show that propofol-induced transport delay causes a significant decrease in vesicle fusion in distal axons. These results are the first to link a propofol-induced delay in neuronal trafficking to a decrease in axonal vesicle fusion, which may alter physiological function during and after anesthesia.
Collapse
Affiliation(s)
- Madeline Frank
- Department of Biological Sciences and Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Alec T. Nabb
- Department of Biological Sciences and Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Susan P. Gilbert
- Department of Biological Sciences and Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Marvin Bentley
- Department of Biological Sciences and Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180,*Address correspondence to: Marvin Bentley ()
| |
Collapse
|
3
|
Zeng X, Li J, Yang F, Xia R. The effect of narcotics on ferroptosis-related molecular mechanisms and signalling pathways. Front Pharmacol 2022; 13:1020447. [PMID: 36313359 PMCID: PMC9606818 DOI: 10.3389/fphar.2022.1020447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/30/2022] [Indexed: 11/26/2022] Open
Abstract
Ferroptosis is a novel programmed cell death form characterized by iron-mediated reactive oxygen species-induced lipid peroxidation and subsequent cell damage that is distinct from apoptosis, necroptosis, pyroptosis, and autophagy. Most studies on ferroptosis are based on its function and mechanism, but there have been relatively few studies on the effects of drugs, especially anaesthetics, on ferroptosis. Therefore, we summarized the recent literature on the effects of anaesthetics on ferroptosis to understand the underlying mechanism. In particular, we focused on the targets of various anaesthetics in different mechanisms of ferroptosis and the effects of ferroptosis induction or inhibition by narcotics on various diseases. The aims of this review are to provide a relatively reasonable drug regimen for clinicians, to explore potential ferroptosis protection drugs and targets, to reduce perioperative complications and to improve the postoperative performance of patients, especially those who are critically ill.
Collapse
Affiliation(s)
- Xiaoqin Zeng
- Department of Anaesthesiology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Jingda Li
- College of Life Sciences, Yangtze University, Jingzhou, Hubei, China
| | - Fuyuan Yang
- School of Basic Medicine, Yangtze University Health Science Center, Jingzhou, Hubei, China
- *Correspondence: Fuyuan Yang, ; Rui Xia,
| | - Rui Xia
- Department of Anaesthesiology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
- *Correspondence: Fuyuan Yang, ; Rui Xia,
| |
Collapse
|
4
|
Yang KS, Che PC, Hsieh MJ, Lee IN, Wu YP, Chen MS, Chen JC. Propofol induces apoptosis and ameliorates 5‑fluorouracil resistance in OSCC cells by reducing the expression and secretion of amphiregulin. Mol Med Rep 2021; 25:36. [PMID: 34859260 PMCID: PMC8669682 DOI: 10.3892/mmr.2021.12552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/29/2021] [Indexed: 12/20/2022] Open
Abstract
Among the different types of oral cancer, >90% of cases are oral squamous cell carcinoma (OSCC). 5-fluorouracil (5-FU) is a commonly used treatment for OSCC, but cells typically display resistance to the drug. Propofol, an intravenous anesthetic agent, exhibits certain anticancer effects, including the inhibition of cancer cell proliferation, migration and invasion. Secreted proteins, such as growth factors and cytokines are involved in cancer development and progression, but the effect of propofol on secreted proteins in OSCC is not completely understood. An MTT assay, flow cytometry and western blotting were performed to determine the anticancer effects of propofol. The secretion profile of OSCC was determined using an antibody array, and clinical importance was assessed using the Gene Expression Profiling Interactive Analysis database. The results were verified by performing reverse transcription-quantitative PCR (RT-qPCR) and western blotting. 5-FU-resistant cells were established to determine the role of the gene of interest in drug resistance. The results demonstrated that propofol decreased cell viability and promoted cell apoptosis. The antibody array results showed that propofol attenuated the secretion of multiple growth factors. The bioinformatics results indicated that amphiregulin (AREG) was expressed at significantly higher levels in cancer tissues, which was also related to poor prognosis. The results of RT-qPCR and western blotting revealed that propofol decreased AREG expression. Pretreatment with exogenous recombinant AREG increased EGFR activation and conferred propofol resistance. Moreover, the results indicated that the expression and activation of AREG was also related to 5-FU resistance, but propofol ameliorated 5-FU drug resistance. Therefore, the present study suggested that propofol combination therapy may serve as an effective treatment strategy for OSCC.
Collapse
Affiliation(s)
- Kung-Ssu Yang
- Department of Anesthesiology, Ditmanson Medical Foundation Chia‑Yi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
| | - Pi-Cheng Che
- Department of Anesthesiology, Ditmanson Medical Foundation Chia‑Yi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
| | - Ming-Ju Hsieh
- Cancer Research Center, Changhua Christian Hospital, Changhua 500209, Taiwan, R.O.C
| | - I-Neng Lee
- Department of Medical Research, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan, R.O.C
| | - Yu-Ping Wu
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 60004, Taiwan, R.O.C
| | - Ming-Shan Chen
- Department of Anesthesiology, Ditmanson Medical Foundation Chia‑Yi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
| | - Jui-Chieh Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 60004, Taiwan, R.O.C
| |
Collapse
|
5
|
Zhao X, Chen F. Propofol induces the ferroptosis of colorectal cancer cells by downregulating STAT3 expression. Oncol Lett 2021; 22:767. [PMID: 34589146 PMCID: PMC8442167 DOI: 10.3892/ol.2021.13028] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/22/2021] [Indexed: 12/24/2022] Open
Abstract
Propofol is a commonly used intravenous anesthetic agent that can also suppress the proliferation of various human cancer types, including colorectal cancer (CRC). The present study aimed to investigate whether propofol could induce the ferroptosis of CRC cells by regulating signal transducer and activator of transcription 3 (STAT3). STAT3 expression in normal and CRC tissues was measured. Human normal colonic epithelial NCM460 cells and human CRC SW480 cells were exposed to different concentrations of propofol and then cell viability was detected. SW480 cells were transfected with a vector overexpressing STAT3 and treated with propofol, and the cell viability, colony formation, cell proliferation, iron level, ROS production and ferroptosis of these cells and control cells were evaluated. Overall, the results showed that STAT3 was highly expressed in CRC tissues. Propofol exerted no marked effect on NCM460 cell viability, but inhibited SW480 cell viability in a concentration-dependent manner. Meanwhile, STAT3 was downregulated by propofol in a concentration-dependent manner. Propofol also inhibited CRC cell proliferation and colony formation, and enhanced cellular iron and ROS levels. Additionally, the expression of proteins involved in ferroptosis was also altered by propofol, including the upregulation of CHAC1 and PTGS2 expression in CRC cells, and the inhibition of GPX4 expression. However, STAT3 overexpression blocked the effect of propofol on CRC cells. In conclusion, propofol may trigger the ferroptosis of CRC cells by downregulating STAT3 expression.
Collapse
Affiliation(s)
- Xining Zhao
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Fei Chen
- Department of Anesthesiology, Mindong Hospital Affiliated to Fujian Medical University, Fu'an, Fujian 355000, P.R. China
| |
Collapse
|
6
|
Bredthauer A, Geiger A, Gruber M, Pfaehler SM, Petermichl W, Bitzinger D, Metterlein T, Seyfried T. Propofol Ameliorates Exaggerated Human Neutrophil Activation in a LPS Sepsis Model. J Inflamm Res 2021; 14:3849-3862. [PMID: 34408467 PMCID: PMC8366786 DOI: 10.2147/jir.s314192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 06/17/2021] [Indexed: 12/29/2022] Open
Abstract
Background Sepsis is a leading cause of morbidity and mortality worldwide. Many patients suffering from sepsis are treated on intensive care units and many of them require mechanical ventilation under sedation or general anesthesia. Propofol, a drug used for these purposes, is known to interact with polymorphonuclear granulocytes (PMNs). Therefore, the aim of this study was to investigate the influence of propofol on PMN functions after experimental Gram-negative induced sepsis using lipopolysaccharide (LPS) stimulation. Methods A total of 34 granulocyte-enriched samples were collected from healthy subjects. PMNs were isolated by density gradient centrifugation and incubated simultaneously with either 6 µg/mL or 60 µg/mL propofol, or none (control). Additionally, the experimental sepsis samples were incubated with either 40 pg/mL or 400 pg/mL LPS. Live cell imaging was conducted in order to observe granulocyte chemotactic migration, ROS production, and NETosis. Flow cytometry was used to analyze viability and antigen expression. Results Propofol led to significantly reduced PMN track length (p < 0.001) and track speed (p < 0.014) after LPS-induced sepsis in a dose-dependent manner. NETosis (p = 0.018) and ROS production (p = 0.039) were accelerated by propofol without LPS incubation, indicating improved immune function. Propofol also ameliorated LPS-induced increased NETosis and ROS-production. Antigen expression for CD11b, CD62l and CD66b was unaffected by propofol. Conclusion Propofol improves LPS-induced exaggerated PMN activation in an ex vivo model. Beneficial effects due to restored immune function in septic patients might be possible, but needs further investigation.
Collapse
Affiliation(s)
- Andre Bredthauer
- Department of Anesthesiology, University Medical Center Regensburg, Regensburg, Germany.,Department of Neurology at the University of Regensburg - Center for Vascular Neurology and Intensive Care Medicine, Regensburg, Germany
| | - Angela Geiger
- Department of Anesthesiology, University Medical Center Regensburg, Regensburg, Germany
| | - Michael Gruber
- Department of Anesthesiology, University Medical Center Regensburg, Regensburg, Germany
| | - Sophie-Marie Pfaehler
- Department of Anesthesiology, University Medical Center Regensburg, Regensburg, Germany
| | - Walter Petermichl
- Department of Anesthesiology, University Medical Center Regensburg, Regensburg, Germany
| | - Diane Bitzinger
- Department of Anesthesiology, University Medical Center Regensburg, Regensburg, Germany
| | - Thomas Metterlein
- Department of Anesthesiology, University Medical Center Regensburg, Regensburg, Germany.,Department of Anesthesiology, Ansbach Hospital, Ansbach, Germany
| | - Timo Seyfried
- Department of Anesthesiology, University Medical Center Regensburg, Regensburg, Germany.,Department of Anesthesiology, Ernst von Bergmann Hospital, Potsdam, Germany
| |
Collapse
|
7
|
Lee KM, Han SM, Lee S, Jeong TY, Kim HJ, Min H, Kim KH, Cha S, Oh WK, Lee J. Fluoride-assisted liquid chromatography-tandem mass spectrometry method for simultaneous analysis of propofol and its metabolites without derivatization in urine. J Chromatogr A 2021; 1652:462360. [PMID: 34246057 DOI: 10.1016/j.chroma.2021.462360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/09/2021] [Accepted: 06/18/2021] [Indexed: 11/18/2022]
Abstract
The misuse of propofol for recreational purposes has become a serious social issue. Accordingly, practical and sensitive analytical methods to investigate the chronic abuse and toxicity of propofol are required. However, current propofol determination methods using liquid chromatography-mass spectrometry (LC-MS/MS) suffer from problems associated with loss in sample preparation due to its volatility and its poor ionization efficiency and collision-induced dissociation in mass spectrometry. Herein, we have developed a sensitive and accurate fluoride-assisted LC-MS/MS method combined with direct-injection for propofol determination. Ionization via fluoride-ion attachment/induced deprotonation, effected by ammonium fluoride in the mobile phase, was found to dramatically improve the sensitivity of propofol without derivatization. Furthermore, direct injection without derivatization enables the simultaneous analysis of propofol and its phase II metabolites without analyte loss. The optimal concentration of ammonium fluoride in the mobile phase was found to be 1 mM under methanol conditions. The linearity is good (R2 ≥ 0.999) and the intra- and inter-day precisions for propofol determination are between 1.9 and 8.7%. The accuracies range from 87.5% to 105.4% and the limits of detection and quantitation for propofol in urine are 0.15 and 0.44 ng mL-1, respectively. The present method was successfully applied to human urine and showed a sufficient sensitivity to determine propofol and five phase II metabolites over 48 h in human urine after administration. Consequently, the fluoride-assisted LC-MS/MS method was demonstrated to be sensitive, accurate, and practical for the determination of propofol and its metabolites.
Collapse
Affiliation(s)
- Kang Mi Lee
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Doping Control Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Sang Moon Han
- Doping Control Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Seunghwa Lee
- Doping Control Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Tae Young Jeong
- Doping Control Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Ho Jun Kim
- Doping Control Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Hophil Min
- Doping Control Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Ki Hun Kim
- Doping Control Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Sangwon Cha
- Department of Chemistry, Dongguk University, Seoul 04620, Republic of Korea
| | - Won Keun Oh
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| | - Jaeick Lee
- Doping Control Center, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea.
| |
Collapse
|
8
|
Mechanistic basis of propofol-induced disruption of kinesin processivity. Proc Natl Acad Sci U S A 2021; 118:2023659118. [PMID: 33495322 DOI: 10.1073/pnas.2023659118] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Propofol is a widely used general anesthetic to induce and maintain anesthesia, and its effects are thought to occur through impact on the ligand-gated channels including the GABAA receptor. Propofol also interacts with a large number of proteins including molecular motors and inhibits kinesin processivity, resulting in significant decrease in the run length for conventional kinesin-1 and kinesin-2. However, the molecular mechanism by which propofol achieves this outcome is not known. The structural transition in the kinesin neck-linker region is crucial for its processivity. In this study, we analyzed the effect of propofol and its fluorine derivative (fropofol) on the transition in the neck-linker region of kinesin. Propofol binds at two crucial surfaces in the leading head: one at the microtubule-binding interface and the other in the neck-linker region. We observed in both the cases the order-disorder transition of the neck-linker was disrupted and kinesin lost its signal for forward movement. In contrast, there was not an effect on the neck-linker transition with propofol binding at the trailing head. Free-energy calculations show that propofol at the microtubule-binding surface significantly reduces the microtubule-binding affinity of the kinesin head. While propofol makes pi-pi stacking and H-bond interactions with the propofol binding cavity, fropofol is unable to make a suitable interaction at this binding surface. Therefore, the binding affinity of fropofol is much lower compared to propofol. Hence, this study provides a mechanism by which propofol disrupts kinesin processivity and identifies transitions in the ATPase stepping cycle likely affected.
Collapse
|
9
|
Gao J, Ding C, Zhou J, Wu G, Han Z, Li J, Hei F. Propofol suppresses lung cancer tumorigenesis by modulating the circ-ERBB2/miR-7-5p/FOXM1 axis. Thorac Cancer 2021; 12:824-834. [PMID: 33506582 PMCID: PMC7952809 DOI: 10.1111/1759-7714.13856] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Propofol is a commonly used anesthetic for cancer surgery. Previous studies have shown that propofol has an anticancer role in various cancers, including lung cancer. This study aimed to investigate the role of propofol in lung cancer and its underlying mechanism. METHODS Cell proliferation was determined by cell counting kit-8 (CCK-8) and colony formation assays. Flow cytometry and transwell assays were used to detect cell apoptosis and invasion, respectively. Glycolysis was evaluated by detecting glucose consumption, lactate production and ATP/ADP ratios. The levels of circular RNA erb-b2 receptor tyrosine kinase 2 (circ-ERBB2), microRNA-7-5p (miR-7-5p) and forkhead box M1 (FOXM1) were tested by quantitative real-time PCR and Western blot. The binding relationship between miR-7-5p and circ-ERBB2/FOXM1 was verified by dual-luciferase reporter assay. Moreover, in vivo experiments were performed by establishing a mouse xenograft model. RESULTS Propofol suppressed cell proliferation, invasion and glycolysis and expedited apoptosis in lung cancer cells. Circ-ERBB2 and FOXM1 were upregulated, while miR-7-5p was decreased in lung cancer tissues and cells. Propofol suppressed lung cancer cell progression by regulating circ-ERBB2. Additionally, miR-7-5p directly interacted with circ-ERBB2 and FOXM1. Also, propofol played an antitumor role in lung cancer via modulating miR-7-5p or FOXM1. Moreover, circ-ERBB2 knockdown enhanced the suppressive effect of propofol on tumor growth in vivo. CONCLUSIONS Propofol inhibited lung cancer progression via mediating circ-ERBB2/miR-7-5p/FOXM1 axis, which might provide an effective therapeutic target for lung cancer therapy.
Collapse
Affiliation(s)
- Jie Gao
- Department of Anesthesiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Chengzhi Ding
- Department of Thoracic Surgery, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Junhui Zhou
- Department of Anesthesiology, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Gang Wu
- Department of Cardiovascular Surgery, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Zongmao Han
- Department of Cardiology, People's Hospital of Zhengzhou University, People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Jianchao Li
- Department of Extracorporeal Circulation, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Feilong Hei
- Department of Extracorporeal Circulation, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| |
Collapse
|
10
|
Drexler B, Grenz J, Grasshoff C, Antkowiak B. Allopregnanolone Enhances GABAergic Inhibition in Spinal Motor Networks. Int J Mol Sci 2020; 21:ijms21197399. [PMID: 33036451 PMCID: PMC7582554 DOI: 10.3390/ijms21197399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 11/16/2022] Open
Abstract
The neurosteroid allopregnanolone (ALLO) causes unconsciousness by allosteric modulation of γ-aminobutyric acid type A (GABAA) receptors, but its actions on the spinal motor networks are unknown. We are therefore testing the hypothesis that ALLO attenuates the action potential firing of spinal interneurons and motoneurons predominantly via enhancing tonic, but not synaptic GABAergic inhibition. We used video microscopy to assess motoneuron-evoked muscle activity in organotypic slice cultures prepared from the spinal cord and muscle tissue. Furthermore, we monitored GABAA receptor-mediated currents by performing whole-cell voltage-clamp recordings. We found that ALLO (100 nM) reduced the action potential firing of spinal interneurons by 27% and that of α-motoneurons by 33%. The inhibitory effects of the combination of propofol (1 µM) and ALLO on motoneuron-induced muscle contractions were additive. Moreover, ALLO evoked a tonic, GABAA receptor-mediated current (amplitude: 41 pA), without increasing phasic GABAergic transmission. Since we previously showed that at a clinically relevant concentration of 1 µM propofol enhanced phasic, but not tonic GABAergic inhibition, we conclude that ALLO and propofol target distinct subpopulations of GABAA receptors. These findings provide first evidence that the combined application of ALLO and propofol may help to reduce intraoperative movements and undesired side effects that are frequently observed under total intravenous anesthesia.
Collapse
|
11
|
Drexler B, Seeger T, Worek F, Thiermann H, Antkowiak B, Grasshoff C. Impact of soman and acetylcholine on the effects of propofol in cultured cortical networks. Toxicol Lett 2020; 322:98-103. [PMID: 31954869 DOI: 10.1016/j.toxlet.2020.01.012] [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: 09/30/2019] [Revised: 12/20/2019] [Accepted: 01/15/2020] [Indexed: 11/29/2022]
Abstract
Patients intoxicated with organophosphorous compounds may need general anaesthesia to enable mechanical ventilation or for control of epileptiform seizures. It is well known that cholinergic overstimulation attenuates the efficacy of general anaesthetics to reduce spontaneous network activity in the cortex. However, it is not clear how propofol, the most frequently used intravenous anaesthetic today, is affected. Here, we investigated the effects of cholinergic overstimulation induced by soman and acetylcholine on the ability of propofol to depress spontaneous action potential activity in organotypic cortical slices measured by extracellular voltage recordings. Cholinergic overstimulation by co-application of soman and acetylcholine (10 μM each) did not reduce the relative inhibition of propofol (1.0 μM; mean normalized action potential firing rate 0.49 ± 0.06 of control condition, p < 0.001, Wilcoxon signed rank test) but clearly reduced its efficacy. Co-application of atropine (10 nM) did not improve the efficacy. Propofol preserved its relative inhibitory potential but did not produce a degree of neuronal depression which can be expected to assure hypnosis in humans. Since a combination with atropine did not improve its efficacy, an increase in dosage will probably be necessary when propofol is used in victims suffering from organophosphorous intoxication.
Collapse
Affiliation(s)
- Berthold Drexler
- Experimental Anesthesiology Section, Department of Anesthesiology and Intensive Care Medicine, Eberhard-Karls-University, Waldhoernlestrasse 22, 72072, Tuebingen, Germany.
| | - Thomas Seeger
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937, Munich, Germany.
| | - Franz Worek
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937, Munich, Germany.
| | - Horst Thiermann
- Bundeswehr Institute of Pharmacology and Toxicology, Neuherbergstrasse 11, 80937, Munich, Germany.
| | - Bernd Antkowiak
- Experimental Anesthesiology Section, Department of Anesthesiology and Intensive Care Medicine, Eberhard-Karls-University, Waldhoernlestrasse 22, 72072, Tuebingen, Germany; Werner Reichardt Center for Integrative Neuroscience, Eberhard-Karls-University, Tuebingen, Germany.
| | - Christian Grasshoff
- Experimental Anesthesiology Section, Department of Anesthesiology and Intensive Care Medicine, Eberhard-Karls-University, Waldhoernlestrasse 22, 72072, Tuebingen, Germany.
| |
Collapse
|
12
|
Abstract
In a series of articles dealing with hypnotics for induction of anesthesia, this article describes the development and current value of propofol. Its significance far exceeds that of a pure induction hypnotic (sedation in diagnostic and therapeutic procedures and on the intensive care unit). Propofol is also used for sedation in diagnostic and therapeutic procedures and on the intensive care unit. In the field of induction of anesthesia, the alternatives are barely used. Some contraindications are still controversial whereas others are no longer sufficiently anchored in the users' awareness (widespread off-label use). Adverse effects, such as injection pain, infection risk and propofol-related infusion syndrome (PRIS) could be significantly reduced by pharmacovigilance. With appropriate caution nearly the whole spectrum of anesthesiology patients can be treated using propofol. The hemodynamic side effects and the rare but potentially fatal PRIS are limitations. Further developments address the water solubility and the solubilizing agents of propofol.
Collapse
Affiliation(s)
- D Bolkenius
- Klinik für Anästhesiologie und Operative Intensivmedizin, Klinikum Augsburg, Stenglinstr. 2, 86156, Augsburg, Deutschland.
| | - C Dumps
- Klinik für Anästhesiologie und Operative Intensivmedizin, Klinikum Augsburg, Stenglinstr. 2, 86156, Augsburg, Deutschland
| | - E Halbeck
- Klinik für Anästhesiologie und Operative Intensivmedizin, Klinikum Augsburg, Stenglinstr. 2, 86156, Augsburg, Deutschland
| |
Collapse
|
13
|
Dankoski E. The 2018 Lasker~DeBakey Clinical Medical Research Award recognizes John Baird Glen for the discovery of propofol. J Clin Invest 2018; 128:4198-4200. [PMID: 30199850 PMCID: PMC6159986 DOI: 10.1172/jci124375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
14
|
Hulsman N, Hollmann M, Preckel B. Newer propofol, ketamine, and etomidate derivatives and delivery systems relevant to anesthesia practice. Best Pract Res Clin Anaesthesiol 2018; 32:213-221. [DOI: 10.1016/j.bpa.2018.08.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 08/01/2018] [Indexed: 12/20/2022]
|
15
|
Common general anesthetic propofol impairs kinesin processivity. Proc Natl Acad Sci U S A 2017; 114:E4281-E4287. [PMID: 28484025 DOI: 10.1073/pnas.1701482114] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Propofol is the most widely used i.v. general anesthetic to induce and maintain anesthesia. It is now recognized that this small molecule influences ligand-gated channels, including the GABAA receptor and others. Specific propofol binding sites have been mapped using photoaffinity ligands and mutagenesis; however, their precise target interaction profiles fail to provide complete mechanistic underpinnings for the anesthetic state. These results suggest that propofol and other common anesthetics, such as etomidate and ketamine, may target additional protein networks of the CNS to contribute to the desired and undesired anesthesia end points. Some evidence for anesthetic interactions with the cytoskeleton exists, but the molecular motors have received no attention as anesthetic targets. We have recently discovered that propofol inhibits conventional kinesin-1 KIF5B and kinesin-2 KIF3AB and KIF3AC, causing a significant reduction in the distances that these processive kinesins can travel. These microtubule-based motors are highly expressed in the CNS and the major anterograde transporters of cargos, such as mitochondria, synaptic vesicle precursors, neurotransmitter receptors, cell signaling and adhesion molecules, and ciliary intraflagellar transport particles. The single-molecule results presented show that the kinesin processive stepping distance decreases 40-60% with EC50 values <100 nM propofol without an effect on velocity. The lack of a velocity effect suggests that propofol is not binding at the ATP site or allosteric sites that modulate microtubule-activated ATP turnover. Rather, we propose that a transient propofol allosteric site forms when the motor head binds to the microtubule during stepping.
Collapse
|
16
|
Anti-VEGF therapy in ophthalmology: a qualitative analysis of transformative drug development. Drug Discov Today 2016; 21:1019-26. [DOI: 10.1016/j.drudis.2016.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 03/25/2016] [Accepted: 05/03/2016] [Indexed: 11/20/2022]
|
17
|
Wang W, Zhou L, Wu LX, Wang T, Zhang CB, Sun L. 5-HT3 Receptor Antagonists for Propofol Injection Pain: A Meta-Analysis of Randomized Controlled Trials. Clin Drug Investig 2016; 36:243-53. [DOI: 10.1007/s40261-016-0375-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|