1
|
Sue SH, Tseng WC, Wu ZS, Huang SM, Chen JL, Wu ZF, Lai HC. The synergistic mechanisms of propofol with cisplatin or doxorubicin in human ovarian cancer cells. J Ovarian Res 2024; 17:187. [PMID: 39272193 PMCID: PMC11401282 DOI: 10.1186/s13048-024-01509-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Most ovarian cancer cases are diagnosed at an advanced stage, leading to poor outcomes and a relatively low 5-year survival rate. While tumor resection in the early stages can be highly effective, recurrence following primary treatment remains a significant cause of mortality. Propofol is a commonly used intravenous anesthetic agent in cancer resection surgery. Previous research has shown that propofol anesthesia was associated with improved survival in patients undergoing elective surgery for epithelial ovarian cancer. However, the underlying antitumor mechanisms are not yet fully understood. METHODS This study aimed to uncover the antitumor properties of propofol alone and combined with cisplatin or doxorubicin, in human SKOV3 and OVCAR3 ovarian cancer cells. We applied flowcytometry analysis for mitochondrial membrane potential, apoptosis, and autophagy, colony formation, migration, and western blotting analysis. RESULTS Given that chemotherapy is a primary clinical approach for managing advanced and recurrent ovarian cancer, it is essential to address the limitations of current chemotherapy, particularly in the use of cisplatin and doxorubicin, which are often constrained by their side effects and the development of resistance. First of all, propofol acted synergistically with cisplatin and doxorubicin in SKOV3 cells. Moreover, our data further showed that propofol suppressed colony formation, disrupted mitochondrial membrane potential, and induced apoptosis and autophagy in SKOV3 and OVCAR3 cells. Finally, the effects of combined propofol with cisplatin or doxorubicin on mitochondrial membrane potential, apoptosis, autophagy, and epithelial-mesenchymal transition were different in SKOV3 and OVCAR3 cells, depending on the p53 status. CONCLUSION In summary, repurposing propofol could provide novel insights into the existing chemotherapy strategies for ovarian cancer. It holds promise for overcoming resistance to cisplatin or doxorubicin and may potentially reduce the required chemotherapy dosages and associated side effects, thus improving treatment outcomes.
Collapse
Affiliation(s)
- Sung-How Sue
- Department of Surgery, Taipei City Hospital Renai Branch, Taipei City, 106, Taiwan, Republic of China
| | - Wei-Cheng Tseng
- Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114, Taiwan, Republic of China
| | - Zih-Syuan Wu
- Institute of Life Sciences, National Defense Medical Center, Taipei City, 114, Taiwan, Republic of China
| | - Shih-Ming Huang
- Institute of Life Sciences, National Defense Medical Center, Taipei City, 114, Taiwan, Republic of China
- Department of Biochemistry, National Defense Medical Center, Taipei City, 114, Taiwan, Republic of China
| | - Jia-Lin Chen
- Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114, Taiwan, Republic of China.
| | - Zhi-Fu Wu
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City 807, Taiwan, Republic of China.
- Department of Anesthesiology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City 807, Taiwan, Republic of China.
- Center for Regional Anesthesia and Pain Medicine, Wan Fang Hospital, Taipei Medical University, Taipei City 116, Taiwan, Taiwan, Republic of China.
| | - Hou-Chuan Lai
- Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114, Taiwan, Republic of China.
| |
Collapse
|
2
|
Di Fiore R, Drago-Ferrante R, Suleiman S, Calleja N, Calleja-Agius J. The role of microRNA-9 in ovarian and cervical cancers: An updated overview. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024:108546. [PMID: 39030109 DOI: 10.1016/j.ejso.2024.108546] [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: 04/02/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024]
Abstract
Ovarian and cervical cancers are the two most frequent kind of gynaecological cancers (GCs). In spite of advances in prevention, screening and treatment, cervical cancer still leads to an increased morbidity and mortality worldwide. Ovarian cancer is often detected at a late stage, which significantly reduces the effectiveness of available treatments. Therefore, novel methods are desperately needed to improve the clinical care of GC patients. MicroRNAs, also known as short noncoding RNAs (miRNAs/miRs), are a diverse group of RNAs with a length of 22 nucleotides. These typically cause translational repression and mRNA degradation by interacting with target mRNAs' 3' untranslated region (3'-UTR), together with other regions and gene promoters. Under certain conditions, they are also able to activate translation or regulate transcription. It has been demonstrated that miRNAs are crucial to several biological processes leading to tumorigenesis, including GCs. Recent research has shown that miR-9 affects carcinogenesis. In this review, we will provide an overview of current research on the potential utility of miR-9 in the diagnosis, prognosis, and therapy of ovarian and cervical malignancies.
Collapse
Affiliation(s)
- Riccardo Di Fiore
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSD, 2080, Msida, Malta; Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA.
| | - Rosa Drago-Ferrante
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSD, 2080, Msida, Malta; BioDNA Laboratories, Malta Life Sciences Park, SGN, 3000, San Gwann, Malta.
| | - Sherif Suleiman
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSD, 2080, Msida, Malta.
| | - Neville Calleja
- Department of Public Health, Faculty of Medicine and Surgery, University of Malta, MSD, 2080, Msida, Malta.
| | - Jean Calleja-Agius
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSD, 2080, Msida, Malta.
| |
Collapse
|
3
|
Yao L, Zhai W, Jiang Z, He R, Xie W, Li Y, Hu Y. The Inhibitory Effects of Propofol on Colorectal Cancer Progression through the NF-κB/HIF-1α Signaling Pathway. Anticancer Agents Med Chem 2024; 24:878-888. [PMID: 38571352 DOI: 10.2174/0118715206283884240326170501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND AND OBJECTIVE Colorectal cancer (CRC) is a neoplastic disease that gradually develops due to genetic variations and epigenetic changes. Surgical excision is the first-line treatment for CRC. Accumulating evidence has shown that total intravenous anesthesia has beneficial effects for CRC patients as it decreases the probability of tumor recurrence and metastasis. Propofol is one of the most frequently used intravenous anesthetics in clinical practice. However, it remains unknown whether it can reduce recurrence and metastasis after surgery in cancer patients. METHODS CRC cell lines (HCT116 and SW480) were cultured in vitro, and different concentrations of propofol were added to the cell culture medium. The proliferation effect of propofol on CRC cell lines was evaluated by CCK-8 assay. The effect of propofol on the migration and invasion of CRC cells was evaluated by scratch healing and Transwell experiments. The inhibitory effects of propofol on NF-κB and HIF-1α expressions in CRC cell lines were determined by Western blotting and immunofluorescence assays to further clarify the regulatory effects of propofol on NF-κB and HIF-1α. RESULTS Compared to the control, propofol significantly inhibited the proliferation, migration, and invasion abilities of CRC cells (HCT116 and SW480) (p < 0.0001). The expression levels of NF-κB and HIF-1α gradually decreased with increasing propofol concentration in both cell lines. After activation and inhibition of NF-κB, the expression of HIF-1α changed. Further studies showed that propofol inhibited LPS-activated NF-κB-induced expression of HIF-1α, similar to the NF-κB inhibitor Bay17083 (p < 0.0001). CONCLUSION In vitro, propofol inhibited the proliferation, migration, and invasion of CRC cells (HCT116 and SW480) in a dose-dependent manner, possibly by participating in the regulation of the NF-κB/HIF-1α signaling pathway.
Collapse
Affiliation(s)
- Liuxu Yao
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wen Zhai
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zongming Jiang
- Department of Anesthesiology, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, China
| | - Rui He
- Department of Anesthesiology, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, China
| | - Weiying Xie
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuhong Li
- Department of Anesthesiology, Shulan (Hangzhou) Hospital, Shulan International Medical College, Zhejiang Shuren College, Hangzhou, Zhejiang, China
| | - Yiyang Hu
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
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: 4] [Impact Index Per Article: 2.0] [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
|
5
|
Debel W, Ramadhan A, Vanpeteghem C, Forsyth RG. Does the Choice of Anaesthesia Affect Cancer? A Molecular Crosstalk between Theory and Practice. Cancers (Basel) 2022; 15:cancers15010209. [PMID: 36612205 PMCID: PMC9818147 DOI: 10.3390/cancers15010209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/01/2023] Open
Abstract
In recent years, there has been an increasing scientific interest in the interaction between anaesthesia and cancer development. Retrospective studies show that the choice of anaesthetics may influence cancer outcome and cancer recurrence; however, these studies show contradictory results. Recently, some large randomized clinical trials have been completed, yet they show no significant effect of anaesthetics on cancer outcomes. In this scoping review, we compiled a body of in vivo and in vitro studies with the goal of evaluating the biological effects of anaesthetics on cancer cells in comparison to clinical effects as described in recent studies. It was found that sevoflurane, propofol, opioids and lidocaine are likely to display direct biological effects on cancer cells; however, significant effects are only found in studies with exposure to high concentrations of anaesthetics and/or during longer exposure times. When compared to clinical data, these differences in exposure and dose-effect relation, as well as tissue selectivity, population selection and unclear anaesthetic dosing protocols might explain the lack of outcome.
Collapse
Affiliation(s)
- Wiebrecht Debel
- Department of Anesthesiology, University Hospital Ghent, 9000 Ghent, Belgium
| | - Ali Ramadhan
- Department of Pathological Anatomy, Universitair Ziekenhuis Brussel, 1090 Brussels, Belgium
| | | | - Ramses G. Forsyth
- Department of Pathological Anatomy, Universitair Ziekenhuis Brussel, 1090 Brussels, Belgium
- Laboratorium for Experimental Pathology (EXPA), Vrije Universiteit Brussel, 1090 Brussels, Belgium
- Correspondence: ; Tel.: +32-(2)-4775084
| |
Collapse
|
6
|
Bonvini JM. General Anesthetics in Cancer Surgery: Can Anesthesiologists Help the Patient with More than a Safe Sleep. Medicina (B Aires) 2022; 58:medicina58091156. [PMID: 36143832 PMCID: PMC9503073 DOI: 10.3390/medicina58091156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/16/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Most patients suffering from neoplastic diseases will at some point during their illness be approached surgically. Surgery itself may be unfortunately responsible for tumor proliferation and metastatic spread. With the perioperative period increasingly becoming a focus of research in anesthesia, anesthesiologists have looked at the chance to influence cancer progression based on their choice of anesthesia regimen and strategy. Many anesthetic agents have been investigated for their potential impact on the course of cancer disease. There is an abundance of retrospective studies and very few prospective ones that tackled this issue. The aim of this article is to review the current state of the evidence on general anesthesia involving volatile and intravenous agents as substrates, focusing on halogenated inhalational agents and propofol, to guide clinical decision making in assessments of the best practice for perioperative management of cancer surgery.
Collapse
Affiliation(s)
- John Michael Bonvini
- Department of Anesthesiology, Ars Medica Clinic, Via Grumo, 16, 6929 Gravesano, Switzerland;
- University of Zurich, Rämistrasse, 71, 8006 Zurich, Switzerland
| |
Collapse
|
7
|
Buddeberg BS, Seeberger MD. Anesthesia and Oncology: Friend or Foe? Front Oncol 2022; 12:802210. [PMID: 35359377 PMCID: PMC8963958 DOI: 10.3389/fonc.2022.802210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/17/2022] [Indexed: 12/24/2022] Open
Abstract
Cancer is a leading cause of death, and surgery is an important treatment modality. Laboratory research and retrospective studies have raised the suspicion that the choice of anesthetics for cancer surgery might affect the course of cancerous disease. The aim of this review is to provide a critical overview of the current state of knowledge. Inhalational anesthesia with volatiles or total intravenous anesthesia (TIVA) with propofol are the two most commonly used anesthetic techniques. Most data comparing volatile anesthetics with TIVA is from either in vitro or retrospective studies. Although conflicting, data shows a trend towards favoring propofol. Opioids are commonly used in anesthesia. Data on potential effects of opioids on growth and recurrence of cancer are scarce and conflicting. Preclinical studies have shown that opioids stimulate cancer growth through the µ-opioid receptor. Opioids also act as immunosuppressants and, therefore, have the potential to facilitate metastatic spread. However, the finding of an adverse effect of opioids on tumor growth and cancer recurrence by some retrospective studies has not been confirmed by prospective studies. Regional anesthesia has not been found to have a beneficial effect on the outcome of surgically treated cancer patients, but prospective studies are scarce. Local anesthetics might have a beneficial effect, as observed in animal and in vitro studies. However, prospective clinical studies strongly question such an effect. Blood products, which may be needed during extensive cancer surgery suppress the immune system, and data strongly suggest a negative impact on cancer recurrence. The potential effects of other commonly used anesthetic agents on the outcome of cancer patients have not been sufficiently studied for drawing valid conclusions. In conclusion, laboratory data and most retrospective studies suggest a potential advantage of TIVA over inhalational anesthesia on the outcome of surgical cancer patients, but prospective, randomized studies are missing. Given the state of weak scientific evidence, TIVA may be used as the preferred type of anesthesia unless there is an individual contraindication against it. Studies on the effects of other drugs frequently used in anesthesia are limited in number and quality, and have found conflicting results.
Collapse
Affiliation(s)
- Bigna S. Buddeberg
- Clinic for Anesthesia, Intermediate Care, Prehospital Emergency Medicine and Pain Therapy, University Hospital Basel, Basel, Switzerland
- Medical School, University of Basel, Basel, Switzerland
| | - Manfred D. Seeberger
- Medical School, University of Basel, Basel, Switzerland
- *Correspondence: Manfred D. Seeberger,
| |
Collapse
|
8
|
Pandit A, Begum Y, Saha P, Srivastava AK, Swarnakar S. Approaches Toward Targeting Matrix Metalloproteases for Prognosis and Therapies in Gynecological Cancer: MicroRNAs as a Molecular Driver. Front Oncol 2022; 11:720622. [PMID: 35145899 PMCID: PMC8821656 DOI: 10.3389/fonc.2021.720622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 12/30/2021] [Indexed: 12/24/2022] Open
Abstract
Gene expression can be regulated by small non-coding RNA molecules like microRNAs (miRNAs) which act as cellular mediators necessary for growth, differentiation, proliferation, apoptosis, and metabolism. miRNA deregulation is often observed in many human malignancies, acting both as tumor-promoting and suppressing, and their abnormal expression is linked to unrestrained cellular proliferation, metastasis, and perturbation in DNA damage as well as cell cycle. Matrix Metalloproteases (MMPs) have crucial roles in both growth, and tissue remodeling in normal conditions, as well as in promoting cancer development and metastasis. Herein, we outline an integrated interactive study involving various MMPs and miRNAs and also feature a way in which these communications impact malignant growth, movement, and metastasis. The present review emphasizes on important miRNAs that might impact gynecological cancer progression directly or indirectly via regulating MMPs. Additionally, we address the likely use of miRNA-mediated MMP regulation and their downstream signaling pathways towards the development of a potential treatment of gynecological cancers.
Collapse
Affiliation(s)
- Anuradha Pandit
- Infectious Diseases & Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Yasmin Begum
- Infectious Diseases & Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Priyanka Saha
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Amit Kumar Srivastava
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Snehasikta Swarnakar
- Infectious Diseases & Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- *Correspondence: Snehasikta Swarnakar,
| |
Collapse
|
9
|
Zhu X, Peng C, Peng Z, Chang R, Guo Q. Sevoflurane Inhibits Metastasis in Hepatocellular Carcinoma by Inhibiting MiR-665-Induced Activation of the ERK/MMP Pathway. Cell Transplant 2022; 31:9636897221104447. [PMID: 35699095 PMCID: PMC9201366 DOI: 10.1177/09636897221104447] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Recent evidence has indicated that inhalational anesthetics may affect the growth
and malignant potential of tumor cells and ultimately influence tumor recurrence
after surgery. Sevoflurane, a volatile anesthetic, is used extensively in
hepatectomy. However, the effect of sevoflurane on the growth of hepatocellular
carcinoma (HCC) cells remains unknown. The aim of this study was to explore the
effects of sevoflurane on HCC metastasis and its potential mechanisms in the
human HCC cell lines, HepG2 and SMMC7721. HepG2 and SMMC7721 cells were treated
with 1.7%, 3.4%, and 5.1 % sevoflurane for 6 h. Cell migration was analyzed
using invasion, migration, and scratch assays. Based on previous literature,
several microRNAs (miRNAs) were screened to determine regulatory miRNA targets
of sevoflurane in HepG2 and SMMC7721 cells; miR-665 was detected as a potential
target and overexpressed or inhibited in HepG2 and SMMC7721 cells by a
lentiviral system. The p-ERK/MMP pathway was also measured by western blotting.
Sevoflurane inhibited the migration and invasion of HCC cells in a
dose-dependent manner. It also inhibited miR-665 expression in HCC cells. We
further observed that sevoflurane inhibited HCC metastasis via miR-665.
Sevoflurane-induced downregulation of miRNA-665 led to phosphorylation of ERK and
matrix metalloproteinase (MMP-9) via suppression of SPRED1. These results
demonstrated that sevoflurane may inhibit invasion and migration via the
p-ERK/MMP-9 signaling pathway in HCC cells.
Collapse
Affiliation(s)
- Xiaoyan Zhu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chuchu Peng
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhiyong Peng
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Guangdong, China
| | - Ruimin Chang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
10
|
Saha P, Das A, Chatterjee N, Chakrabarti D, Sinha D. Impact of anesthetics on oncogenic signaling network: a review on propofol and isoflurane. Fundam Clin Pharmacol 2021; 36:49-71. [PMID: 34655261 DOI: 10.1111/fcp.12732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/05/2021] [Accepted: 10/14/2021] [Indexed: 12/26/2022]
Abstract
Propofol as an intravenous anesthetic and isoflurane as an inhalational/volatile anesthetic continue to be an important part of surgical anesthetic interventions worldwide. The impact of these anesthetics on tumor progression, immune modulation, and survival rates of cancer patients has been widely investigated. Although most of the preclinical studies have provided a beneficial effect of propofol over isoflurane or other volatile anesthetics, several investigations have shown contradictory results, which warrant more preclinical and clinical studies. Propofol mostly exhibits antitumor properties, whereas isoflurane being a cost-effective anesthetic is frequently used. However, isoflurane has been also reported with protumorigenic activity. This review provides an overall perspective on the network of signaling pathways that may modulate several steps of tumor progression from inflammation, immunomodulation, epithelial-mesenchymal transition (EMT) to invasion, metastasis, angiogenesis, and cancer stemness and extracellular vesicles along with chemotherapeutic applications and clinical status of these anesthetics. A clear understanding of the mechanistic viewpoints of these anesthetics may pave the way for more prospective clinical trials with the ultimate goal of obtaining a safe and optimal anesthetic intervention that would prevent cancer recurrence and may influence better postoperative survival.
Collapse
Affiliation(s)
- Priyanka Saha
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata, India
| | - Ananya Das
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata, India
| | - Nabanita Chatterjee
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata, India
| | - Deepa Chakrabarti
- Department of Anesthesiology, Chittaranjan National Cancer Institute, Kolkata, India
| | - Dona Sinha
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata, India
| |
Collapse
|
11
|
Tseng WC, Lee MS, Lin YC, Lai HC, Yu MH, Wu KL, Wu ZF. Propofol-Based Total Intravenous Anesthesia is Associated with Better Survival than Desflurane Anesthesia in Epithelial Ovarian Cancer Surgery: A Retrospective Cohort Study. Front Pharmacol 2021; 12:685265. [PMID: 34630078 PMCID: PMC8497698 DOI: 10.3389/fphar.2021.685265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 09/10/2021] [Indexed: 02/01/2023] Open
Abstract
Background: Previous studies have shown that anesthetic techniques can affect outcomes of cancer surgery. We investigated the association between anesthetic techniques and patient outcomes after elective epithelial ovarian cancer surgery. Methods: This was a retrospective cohort study of patients who received elective open surgery for epithelial ovarian cancer between January 2009 and December 2014. Patients were grouped according to the administration of propofol or desflurane anesthesia. Kaplan–Meier analysis was performed, and survival curves were constructed from the date of surgery to death. Univariate and multivariate Cox regression models were used to compare hazard ratios for death after propensity matching. Subgroup analyses were performed for age, body mass index, preoperative carbohydrate antigen-125 level, International Federation of Gynecology and Obstetrics staging, and operation and anesthesia time. Results: In total, 165 patients (76 deaths, 46.1%) who received desflurane anesthesia and 119 (30 deaths, 25.2%) who received propofol anesthesia were eligible for analysis. After propensity matching, 104 patients were included in each group. In the matched analysis, patients who received propofol anesthesia had better survival with a hazard ratio of 0.52 (95% confidence interval, 0.33–0.81; p = 0.005). Subgroup analyses also showed significantly better survival with old age, high body mass index, elevated carbohydrate antigen-125 level, advanced International Federation of Gynecology and Obstetrics stage, and prolonged operation and anesthesia time in the matched propofol group. In addition, patients administered with propofol anesthesia had less postoperative recurrence and metastasis than those administered with desflurane anesthesia in the matched analysis. Conclusion: Propofol anesthesia was associated with better survival in patients who underwent elective epithelial ovarian cancer open surgery. Prospective studies are warranted to evaluate the effects of propofol anesthesia on oncological outcomes in patients with epithelial ovarian cancer.
Collapse
Affiliation(s)
- Wei-Cheng Tseng
- Department of Anesthesiology, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Meei-Shyuan Lee
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Ying-Chih Lin
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Hou-Chuan Lai
- Department of Anesthesiology, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Mu-Hsien Yu
- Department of Obstetrics and Gynecology, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Ke-Li Wu
- Department of General Medicine, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Zhi-Fu Wu
- Department of Anesthesiology, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan.,Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Anesthesiology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
12
|
Bimonte S, Cascella M, Forte CA, Esposito G, Del Prato F, Raiano N, Del Prete P, Cuomo A. Effects of the Hypnotic Alkylphenol Derivative Propofol on Breast Cancer Progression. A Focus on Preclinical and Clinical Studies. In Vivo 2021; 35:2513-2519. [PMID: 34410937 DOI: 10.21873/invivo.12532] [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: 05/05/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 11/10/2022]
Abstract
Propofol is a hypnotic alkylphenol derivative with many biological activities. It is predominantly used in anesthesia and is the most used parenteral anesthetic agent in the United States. Accumulating preclinical studies have shown that this compound may inhibit cancer recurrence and metastasis. Nevertheless, other investigations provided evidence that this compound may promote breast cancer cell progression by modulating different molecular pathways. Clinical data on this topic are scarce and derive from retrospective analyses. For this reason, we reviewed and evaluated the available data to reveal insight into this controversial issue. More preclinical and clinical investigations are necessary to determine the potential role of propofol in the proliferation of breast cancer cells.
Collapse
Affiliation(s)
- Sabrina Bimonte
- Division of Anesthesia and Pain Medicine, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Naples, Italy;
| | - Marco Cascella
- Division of Anesthesia and Pain Medicine, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Naples, Italy
| | - Cira Antonietta Forte
- Division of Anesthesia and Pain Medicine, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Naples, Italy
| | - Gennaro Esposito
- Division of Anesthesia and Pain Medicine, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Naples, Italy
| | - Francesco Del Prato
- Division of Anesthesia and Pain Medicine, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Naples, Italy
| | - Nicola Raiano
- Radiology Division, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Via Mariano Semmola, Naples, Italy
| | - Paola Del Prete
- Direzione Scientifica, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Naples, Italy
| | - Arturo Cuomo
- Division of Anesthesia and Pain Medicine, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Naples, Italy
| |
Collapse
|
13
|
Ashrafizadeh M, Ahmadi Z, Farkhondeh T, Samarghandian S. Anti-tumor Activity of Propofol: A Focus on MicroRNAs. Curr Cancer Drug Targets 2021; 20:104-114. [PMID: 31657687 DOI: 10.2174/1568009619666191023100046] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/02/2019] [Accepted: 09/23/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND MicroRNAs are endogenous, short, non-coding RNAs with the length as low as 20 to 25 nucleotides. These RNAs are able to negatively affect the gene expression at the post-transcriptional level. It has been demonstrated that microRNAs play a significant role in cell proliferation, cell migration, cell death, cell differentiation, infection, immune response, and metabolism. Besides, the dysfunction of microRNAs has been observed in a variety of cancers. So, modulation of microRNAs is of interest in the treatment of disorders. OBJECTIVE The aim of the current review is to investigate the modulatory effect of propofol on microRNAs in cancer therapy. METHODS This review was performed at PubMed, SCOPUS and Web of Science data-bases using keywords "propofol', "microRNA", "cancer therapy", "propofol + microRNA" and "propofol + miR". RESULTS It was found that propofol dually down-regulates/upregulates microRNAs to exert its antitumor activity. In terms of oncogenesis microRNAs, propofol exert an inhibitory effect, while propofol significantly enhances the expression of oncosuppressor microRNAs. CONCLUSION It seems that propofol is a potential modulator of microRNAs and this capability can be used in the treatment of various cancers.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Zahra Ahmadi
- Department of Basic Science, Veterinary Medicine Faculty, Shushtar University, Khuzestan, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
14
|
Regulation of Nuclear Factor-KappaB (NF-κB) signaling pathway by non-coding RNAs in cancer: Inhibiting or promoting carcinogenesis? Cancer Lett 2021; 509:63-80. [PMID: 33838282 DOI: 10.1016/j.canlet.2021.03.025] [Citation(s) in RCA: 176] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/18/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022]
Abstract
The nuclear factor-kappaB (NF-κB) signaling pathway is considered as a potential therapeutic target in cancer therapy. It has been well established that transcription factor NF-κB is involved in regulating physiological and pathological events including inflammation, immune response and differentiation. Increasing evidences suggest that deregulated NF-κB signaling can enhance cancer cell proliferation, metastasis and also mediate radio-as well as chemo-resistance. On the contrary, non-coding RNAs (ncRNAs) have been found to modulate NF-κB signaling pathway under different settings. MicroRNAs (miRNAs) can dually inhibit/induce NF-κB signaling thereby affecting the growth and migration of cancer cells. Furthermore, the response of cancer cells to radiotherapy and chemotherapy may also be regulated by miRNAs. Regulation of NF-κB by miRNAs may be mediated via binding to 3/-UTR region. Interestingly, anti-tumor compounds can increase the expression of tumor-suppressor miRNAs in inhibiting NF-κB activation and the progression of cancers. Long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) can also effectively modulate NF-κB signaling thus affecting tumorigenesis. It is noteworthy that several studies have demonstrated that lncRNAs and circRNAs can affect miRNAs in targeting NF-κB activation. They can act as competing endogenous RNA (ceRNA) thereby reducing miRNA expression to induce NF-κB activation that can in turn promote cancer progression and malignancy.
Collapse
|
15
|
Propofol inhibits proliferation, migration, invasion and promotes apoptosis by regulating HOST2/JAK2/STAT3 signaling pathway in ovarian cancer cells. Cytotechnology 2021; 73:243-252. [PMID: 33927479 DOI: 10.1007/s10616-021-00462-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/23/2021] [Indexed: 12/24/2022] Open
Abstract
We aimed to investigate the effect of propofol on the growth of human ovarian cancer cells ES2 and OVCAR-3 in vitro by regulating long non-coding RNA HOST2 (human ovarian cancer-specific transcript 2) and the inhibition of JAK2/STAT3 signaling pathway. In the present study, ES-2 and OVCAR-3 cells were firstly treated with different concentrations of propofol (0, 1, 5 and 10 μg/ml). The expression of HOST2 in ES-2 and OVCAR-3 cells were detected by quantitative reverse transcription-PCR (qRT-PCR). Then, the expression of HOST2 was changed by transfection of HOST2 overexpression plasmid into ES-2 and OVCAR-3 cells. Cell proliferation, migration, invasion and apoptosis were performed using CCK-8, wound-healing, Transwell assays and Flow Cytometry. Western blot analysis was performed to detect the expressions of apoptosis-associated proteins and JAK2/STAT3 pathway-related proteins. Results showed that cell viability and intracellular HOST2 expression in ES-2 and OVCAR-3 cells were decreased gradually with the increase of propofol concentration in a dose-dependent manner. Compared with the propofol group, overexpression of HOST2 significantly promoted the cell proliferation, migration, invasion and inhibited apoptosis, and ameliorated the inhibitory effect of propofol on the growth of tumor cells. Western blot analysis showed that compared with propofol group, the expression of Bcl-2 was significantly increased whereas Bax and the ratio of Cleaved caspase3/caspase3 were significantly decreased in pcDNA-HOST2 group. In addition, overexpression of HOST2 significantly enhanced the phosphorylation level of JAK2 and STAT3, and reduced the suppressive effect of propofol on JAK2/STAT3 signaling. Our results illustrated that propofol could significantly inhibit the proliferation, migration, invasion and induce apoptosis of ES-2 and OVCAR-3 cells by downregulating HOST2. The regulation mechanism may be achieved by inhibiting the activation of JAK2/STAT3 signaling pathway.
Collapse
|
16
|
Lu H, Zheng G, Gao X, Chen C, Zhou M, Zhang L. Propofol suppresses cell viability, cell cycle progression and motility and induces cell apoptosis of ovarian cancer cells through suppressing MEK/ERK signaling via targeting circVPS13C/miR-145 axis. J Ovarian Res 2021; 14:30. [PMID: 33563314 PMCID: PMC7874627 DOI: 10.1186/s13048-021-00775-3] [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: 11/09/2020] [Accepted: 01/25/2021] [Indexed: 12/11/2022] Open
Abstract
Background Propofol is a kind of common intravenous anaesthetic agent that plays an anti-tumor role in a variety of cancers, including ovarian cancer. However, the working mechanism of Propofol in ovarian cancer needs further exploration. Methods The viability and metastasis of ovarian cancer cells were assessed by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and transwell assays. Flow cytometry was used to evaluate the cell cycle and apoptosis. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to examine the abundance of circular RNA vacuolar protein sorting 13 homolog C (circVPS13C) and microRNA-145 (miR-145). The target relationship between miR-145 and circVPS13C was predicted by circinteractome database and verified by dual-luciferase reporter assay, RNA-binding protein immunoprecipitation (RIP) assay and RNA-pull down assay. Western blot assay was used to detect the levels of phosphorylated extracellular regulated MAP kinase (p-ERK), ERK, p-MAP kinse-ERK kinase (p-MEK) and MEK, in ovarian cancer cells. Results Propofol treatment suppressed the viability, cell cycle and motility and elevated the apoptosis rate of ovarian cancer cells. Propofol up-regulated miR-145 in a dose-dependent manner. Propofol exerted an anti-tumor role partly through up-regulating miR-145. MiR-145 was a direct target of circVPS13C. Propofol suppressed the progression of ovarian cancer through up-regulating miR-145 via suppressing circVPS13C. Propofol functioned through circVPS13C/miR-145/MEK/ERK signaling in ovarian cancer cells. Conclusion Propofol suppressed the proliferation, cell cycle, migration and invasion and induced the apoptosis of ovarian cancer cells through circVPS13C/miR-145/MEK/ERK signaling in vitro.
Collapse
Affiliation(s)
- Huan Lu
- Department of Anesthesiology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, No.18 daoshan Road, Fuzhou City, 350001, Fujian Province, China.
| | - Guanlin Zheng
- Department of Anesthesiology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, No.18 daoshan Road, Fuzhou City, 350001, Fujian Province, China
| | - Xiang Gao
- Department of Anesthesiology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, No.18 daoshan Road, Fuzhou City, 350001, Fujian Province, China
| | - Chanjuan Chen
- Department of Anesthesiology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, No.18 daoshan Road, Fuzhou City, 350001, Fujian Province, China
| | - Min Zhou
- Department of Anesthesiology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, No.18 daoshan Road, Fuzhou City, 350001, Fujian Province, China
| | - Longxin Zhang
- Department of Anesthesiology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, No.18 daoshan Road, Fuzhou City, 350001, Fujian Province, China
| |
Collapse
|
17
|
Zhou HY, Jiang F, Cao Z, Shen QY, Feng YJ, Hou ZH. Propofol protects PC12 cells from cobalt chloride-induced injury by mediating miR-134. Histol Histopathol 2021; 36:425-435. [PMID: 33410125 DOI: 10.14670/hh-18-298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Propofol (PRO) was reported to exert a neuroprotective effect by decreasing microRNA-134 (miR-134), a brain-specific miRNA, thus, the role of PRO against cobalt chloride (CoCl₂)-induced injury in rat pheochromocytoma cells (PC12) via mediating miR-134 was explored. METHODS CoCl₂-induced PC12 cells treated with PRO were transfected with or without miR-134 negative control (NC)/ inhibitor/mimic, and the following detections were then performed using cell counting kit-8 (CCK-8), Annexin V-fluorescein isothiocyanate/propidium iodide (Annexin V-FITC/PI) and Hoechst 33258 staining. Autophagy was observed by transmission electron microscope (TEM). Mitochondrial membrane potential (MMP) was detected by Rhodamine-123 (Rh123) staining, and reactive oxygen species (ROS) by dichloro-dihydro-fluorescein diacetate (DCFH-DA) staining. Protein and gene expressions were measured by Western blotting and quantitative reverse transcriptase polymerase chain reaction (qRT-PCR), respectively. RESULTS PRO reversed the CoCl₂-induced decrease in the PC12 cell viability and increased miR-134 in a dose-dependent manner. CoCl₂ increased LC3II/I ratio and Beclin-1 expression, but decreased p62 expression, which was abolished by PRO. In addition, an increased cell apoptosis rates triggered by CoCl₂ were reduced by PRO with the down-regulations of Bax and Caspase-3 and the up-regulation of Bcl-2. Furthermore, PRO decreased methylenedioxyamphetamine (MDA), nitric oxide (NO) and ROS in CoCl₂-induced PC12 cells accompanying the increase in glutathione peroxidase (GSH-Px) and MMP. The effects of PRO on autophagy, apoptosis and oxidative stress in CoCl₂-induced PC12 cell were reversed by miR-134 mimic. CONCLUSION PRO may mitigate CoCl₂-induced autophagy in PC12 cells with decreased apoptosis and improved oxidative stress via mediating miR-134.
Collapse
Affiliation(s)
- Hong-Yi Zhou
- Department of Anesthesiology, Tongzhou Maternal and Child Health Hospital of Beijing, Beijing, China.
| | - Fan Jiang
- Department of General Medicine, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Zhong Cao
- Department of Anesthesiology, Tongzhou Maternal and Child Health Hospital of Beijing, Beijing, China
| | - Qi-Yun Shen
- Department of Anesthesiology, Tongzhou Maternal and Child Health Hospital of Beijing, Beijing, China
| | - Yu-Jing Feng
- Department of Anesthesiology, Tongzhou Maternal and Child Health Hospital of Beijing, Beijing, China
| | - Zhen-Huan Hou
- Department of Anesthesiology, Tongzhou Maternal and Child Health Hospital of Beijing, Beijing, China
| |
Collapse
|
18
|
Song F, Liu J, Feng Y, Jin Y. Propofol‑induced HOXA11‑AS promotes proliferation, migration and invasion, but inhibits apoptosis in hepatocellular carcinoma cells by targeting miR‑4458. Int J Mol Med 2020; 46:1135-1145. [PMID: 32705160 PMCID: PMC7387087 DOI: 10.3892/ijmm.2020.4667] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 06/03/2020] [Indexed: 12/12/2022] Open
Abstract
Propofol is a commonly used drug for the induction and maintenance of anesthesia. Previous studies have reported that propofol is involved in the progression of numerous human cancer types, including hepatocellular carcinoma (HCC). However, the underlying molecular mechanisms in HCC are yet to be elucidated. The present study aimed to investigate the potential mechanism of propofol in HCC development. MTT assay, flow cytometry analysis and Transwell assays were conducted to examine cell proliferation, apoptosis, migration and invasion, respectively. Western blotting was also performed to determine the protein expression levels of Bcl‑2 and cleaved‑caspase 3. An in vivo experiment was performed to assess the effect of propofol on tumor growth. Moreover, reverse transcription‑quantitative PCR was conducted to measure the mRNA expression levels of HOMEOBOX A11 (HOXA11) antisense RNA (HOXA11‑AS) and microRNA (miR)‑4458. Dual‑luciferase reporter and RNA pull‑down assays were performed to evaluate the target relationship between HOXA11‑AS and miR‑4458. It was demonstrated that propofol inhibited HCC cell proliferation, migration and invasion, and promoted cell apoptosis in vitro. Furthermore, propofol could suppress tumor growth in vivo. Propofol suppressed the expression of HOXA11‑AS in HCC cells, while HOXA11‑AS overexpression reversed the inhibitory effect of propofol treatment on cell progression in HCC. In addition, miR‑4458 was identified as a target of HOXA11‑AS, and miR‑4458 inhibition reversed the effect of HOXA11‑AS knockdown on HCC cell progression. The results also indicated that propofol promoted the expression of miR‑4458, while HOXA11‑AS restored this effect in HCC. Thus, it was suggested that propofol suppressed cell progression by modulating the HOXA11‑AS/miR‑4458 axis in HCC.
Collapse
Affiliation(s)
- Furong Song
- Department of Anesthesiology
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Jun Liu
- Department of Anesthesiology
| | | | - Yi Jin
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
19
|
Zeng J, Li YK, Quan FF, Zeng X, Chen CY, Zeng T, Zou J, Tong WJ. Propofol‑induced miR‑125a‑5p inhibits the proliferation and metastasis of ovarian cancer by suppressing LIN28B. Mol Med Rep 2020; 22:1507-1517. [PMID: 32627014 PMCID: PMC7346589 DOI: 10.3892/mmr.2020.11223] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 05/01/2020] [Indexed: 12/13/2022] Open
Abstract
Propofol, a commonly used intravenous anesthetic agent during surgery, has relatively widespread pharmacological actions. Previous studies have reported that propofol may act as an antitumor drug in several cancer types, such as pancreatic cancer, lung cancer and gastric cancer. However, the underlying mechanism in ovarian cancer remain unknown. Therefore, the present study investigated the pharmacological effect of propofol on microRNAs (miRNAs) in ovarian cancer treatment. Propofol (1, 5 or 10 µg/ml) was used to treat A2780 and SKOV3 ovarian cancer cells for 1, 2, 3, 4 or 5 days. The MTT assay was used to detect cell viability, while wound healing and Transwell assays were utilized to assess the invasive and migratory abilities. The bioinformatics prediction approach identified differentially expressed miRNAs (miRs) that were used in Gene Ontology, Gene Set Enrichment Analysis and Kyoto Encyclopedia of Genes and Genomes analyses. The expression levels of miR‑125a‑5p and lin‑28 homolog B (LIN28B) were evaluated by reverse transcription‑quantitative PCR (RT‑qPCR). A luciferase assay was performed to identify the relationship between miR‑125a‑5p and LIN28B. Western blotting was conducted to measure the protein expression of LIN28B. It was demonstrated that propofol significantly upregulated miR‑125a‑5p to exert its antitumor activity. RT‑qPCR results suggested that propofol could upregulate miR‑125a‑5p and LIN28B expression levels in ovarian cancer cell lines. Western blot analysis also indicated that propofol could enhance the expression of LIN28B in ovarian cancer cell lines. The luciferase assay identified that miR‑125a‑5p could directly inhibit the expression of LIN28B to suppress proliferation and metastasis in ovarian cancer. In conclusion, these results suggested that propofol inhibited ovarian cancer proliferation and metastasis by enhancing miR‑125a‑5p, which targets LIN28B.
Collapse
Affiliation(s)
- Juan Zeng
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yu-Kun Li
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Fei-Fei Quan
- Department of Gynecology, Foshan First People's Hospital, Foshan, Guangdong 528000, P.R. China
- Department of Gynecology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xin Zeng
- Department of Histology and Embryology, Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Chang-Ye Chen
- Department of Gynecology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Tian Zeng
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Juan Zou
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, Hunan 421001, P.R. China
- Correspondence to: Dr Juan Zou, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, University of South China, 28 West Changsheng Road, Hengyang, Hunan 421001, P.R. China, E-mail:
| | - Wen-Juan Tong
- Department of Obstetrics, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
- Dr Wen-Juan Tong, Department of Obstetrics, The First Affiliated Hospital of University of South China, 69 Chuanshan Road, Hengyang, Hunan 421001, P.R. China, E-mail:
| |
Collapse
|
20
|
Xu Y, Pan S, Jiang W, Xue F, Zhu X. Effects of propofol on the development of cancer in humans. Cell Prolif 2020; 53:e12867. [PMID: 32596964 PMCID: PMC7445405 DOI: 10.1111/cpr.12867] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer is one of most the significant threats to human health worldwide, and the primary method of treating solid tumours is surgery. Propofol, one of the most widely used intravenous anaesthetics in surgery, was found to be involved in many cancer‐related pathophysiology processes, mainly including anti‐tumour and minor cancer‐promoting effects in various types of cancer. An increasing number of studies have identified that propofol plays a role in cancer by regulating the expression of multiple signalling pathways, downstream molecules, microRNAs and long non‐coding RNAs. Emerging evidence has indicated that propofol can enhance the anti‐tumour effect of chemotherapeutic drugs or some small molecular compounds. Additionally, in vivo animal models have shown that propofol inhibits tumour growth and metastasis. Furthermore, most clinical trials indicate that propofol is associated with better survival outcomes in cancer patients after surgery. Propofol use is encouraged in cancers that appear to have a better prognosis after its use during surgery. We hope that future large and prospective multicenter studies will provide more precise answers to guide the choice of anaesthetics during cancer surgery.
Collapse
Affiliation(s)
- Yichi Xu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shuya Pan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenxiao Jiang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fang Xue
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
21
|
Wu X, Li X, Xu G. Propofol suppresses the progression of non‑small cell lung cancer via downregulation of the miR‑21‑5p/MAPK10 axis. Oncol Rep 2020; 44:487-498. [PMID: 32468043 PMCID: PMC7336515 DOI: 10.3892/or.2020.7619] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/16/2020] [Indexed: 12/16/2022] Open
Abstract
Non‑small cell lung cancer (NSCLC) accounts for >80% of lung cancer cases and is the leading cause of cancer‑associated mortality worldwide. Propofol is an anesthetic drug frequently used during tumor resection. It is also known to exert inhibitory effects on cancer. Although the role of propofol in NSCLC has been reported, its underlying mechanisms remain unknown. The present study aimed therefore to investigate the mechanisms of propofol action on NSCLC. Starbase V3.0 project was used to analyze the expression levels of microRNA‑21‑5p (miR‑21‑5p) and mitogen‑activated protein kinase 10 (MAPK10) in NSCLC and adjacent normal tissues from patients with NSCLC and the association between miR‑21‑5p and MAPK10 expression level in NSCLC tissues. The correlation between MAPK10 expression and disease‑free survival (DFS) in patients with NSCLC was analyzed using GEPIA software version 1.0. miR‑21‑5p and MAPK10 expression in tumor and adjacent normal tissues from patients with NSCLC was evaluated by reverse transcription‑quantitative (RT‑q) PCR and western blotting. Cell viability and apoptosis were assessed by using Cell Counting Kit‑8 assay and flow cytometry, respectively. The interaction between miR‑21‑5p and MAPK10 was predicted by TargetScan/miRanda and verified by dual luciferase assay. The regulatory effect of propofol on miR‑21‑5p and MAPK10 expression in NSCLC cell lines was examined by RT‑qPCR and western blotting. Starbase V3.0 project and the results of the present study indicated that tumor tissues presented a significantly lower MAPK10 level and a higher miR‑21‑5p level compared with the normal samples, and that miR‑21‑5p expression was negatively correlated with MAPK10 expression in the tumor tissues of patients with NSCLC. Furthermore, miR‑21‑5p targeted the 3'‑untranslated region of MAPK10. In addition, compared with BEAS‑2B cells, a higher miR‑21‑5p and a lower MAPK10 expression was observed in the NSCLC cell lines A549 and H1299, which was reversed by propofol. The overexpression of miR‑21‑5p abrogated the effects of propofol on A549 and H1299 cell viability and apoptosis by targeting MAPK10. Taken together, these findings demonstrated that propofol inhibited the viability and promoted the apoptosis of NSCLC cells by downregulating the miR‑21‑5p/MAPK10 axis.
Collapse
Affiliation(s)
- Xinhua Wu
- Department of Anesthesiology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang UygurAutonomous Region, P.R.China
| | - Xuebin Li
- Department of Anesthesiology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang UygurAutonomous Region, P.R.China
| | - Guiping Xu
- Department of Anesthesiology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang UygurAutonomous Region, P.R.China
| |
Collapse
|
22
|
Wang H, Zhao L, Wu J, Hong J, Wang S. Propofol induces ROS-mediated intrinsic apoptosis and migration in triple-negative breast cancer cells. Oncol Lett 2020; 20:810-816. [PMID: 32566008 PMCID: PMC7285815 DOI: 10.3892/ol.2020.11608] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 04/03/2020] [Indexed: 11/05/2022] Open
Abstract
Propofol is widely applied in general anesthesia owing to its short effect and rapid recovery. Apart from its anesthetic advantages, propofol has also been observed to inhibit the growth of several types of cancer cells. Breast cancer is the most diagnosed cancer in females worldwide and triple negative breast cancer (TNBC) constitutes 15-20% of all breast cancer cases. TNBC is characterized by a high recurrence rate, which is associated with its high mortality rate. The present study aimed to evaluate apoptosis in MDA-MB-468 cells treated with propofol. The Cell Counting Kit-8 assay was used to assess proliferation in cells treated with different concentrations of propofol. In addition, Annexin V-FITC was used to detect apoptosis. Furthermore, the generation of reactive oxygen species (ROS) was examined. The relative expression of proteins in the intrinsic apoptosis pathway, such as Bak, Bax, Bcl-2, Cytochrome c, apoptotic peptidase-activating factor 1 (Apaf-1), Caspase 3 and Caspase 9, were calculated relative to GAPDH with western blot analysis. A wound healing assay was performed to examine the effect of propofol on MDA-MB-468 cell migration. The present study revealed that propofol inhibited the proliferation and increased the level of ROS in MDA-MB-468 cells. The expression levels of Cytochrome c, Apaf-1, Bax, Bak and cleaved Caspase 3/9 were upregulated compared with GAPDH. The level of Bcl-2 protein was upregulated by propofol at a concentration of 5 µM and downregulated at concentrations of 10 and 20 µM. In the wound-healing assay, propofol reduced the scratch distance and area. Taken together, the results of the present study suggested that propofol may induce ROS-mediated intrinsic apoptosis and promote migration in TNBC cells.
Collapse
Affiliation(s)
- Hao Wang
- Department of Traditional Chinese Medicine, The General Hospital Affiliated to Shanghai Jiaotong University, Shanghai 200080, P.R. China.,Department of Pneumology, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, Jiangsu 224003, P.R. China
| | - Lidong Zhao
- Department of Internal and Emergency Medicine, The General Hospital Affiliated to Shanghai Jiaotong University, Shanghai 200080, P.R. China
| | - Jing Wu
- School of Pharmacy, Shanghai Jiaotong University, Shanghai 200080, P.R. China
| | - Jiang Hong
- Department of Internal and Emergency Medicine, The General Hospital Affiliated to Shanghai Jiaotong University, Shanghai 200080, P.R. China
| | - Songpo Wang
- Department of Traditional Chinese Medicine, The General Hospital Affiliated to Shanghai Jiaotong University, Shanghai 200080, P.R. China
| |
Collapse
|
23
|
Gao X, Mi Y, Guo N, Luan J, Xu H, Hu Z, Wang N, Zhang D, Gou X, Xu L. The mechanism of propofol in cancer development: An updated review. Asia Pac J Clin Oncol 2020; 16:e3-e11. [DOI: 10.1111/ajco.13301] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Xingchun Gao
- Institute of Basic Medical Sciences & Shaanxi Key Laboratory of Brain DisordersShaanxi Key Laboratory of Ischemic Cardiovascular DiseaseXi'an Medical University Xi'an China
- State Key Laboratory of Military StomatologyDepartment of AnesthesiologySchool of StomatologyThe Fourth Military Medical University Xi'an China
| | - Yajing Mi
- Institute of Basic Medical Sciences & Shaanxi Key Laboratory of Brain DisordersShaanxi Key Laboratory of Ischemic Cardiovascular DiseaseXi'an Medical University Xi'an China
- State Key Laboratory of Military StomatologyDepartment of AnesthesiologySchool of StomatologyThe Fourth Military Medical University Xi'an China
| | - Na Guo
- Institute of Basic Medical Sciences & Shaanxi Key Laboratory of Brain DisordersShaanxi Key Laboratory of Ischemic Cardiovascular DiseaseXi'an Medical University Xi'an China
| | - Jing Luan
- Institute of Basic Medical Sciences & Shaanxi Key Laboratory of Brain DisordersShaanxi Key Laboratory of Ischemic Cardiovascular DiseaseXi'an Medical University Xi'an China
- State Key Laboratory of Military StomatologyDepartment of AnesthesiologySchool of StomatologyThe Fourth Military Medical University Xi'an China
| | - Hao Xu
- Institute of Basic Medical Sciences & Shaanxi Key Laboratory of Brain DisordersShaanxi Key Laboratory of Ischemic Cardiovascular DiseaseXi'an Medical University Xi'an China
- State Key Laboratory of Military StomatologyDepartment of AnesthesiologySchool of StomatologyThe Fourth Military Medical University Xi'an China
| | - Zhifang Hu
- Institute of Basic Medical Sciences & Shaanxi Key Laboratory of Brain DisordersShaanxi Key Laboratory of Ischemic Cardiovascular DiseaseXi'an Medical University Xi'an China
| | - Ning Wang
- Institute of Basic Medical Sciences & Shaanxi Key Laboratory of Brain DisordersShaanxi Key Laboratory of Ischemic Cardiovascular DiseaseXi'an Medical University Xi'an China
| | - Dian Zhang
- Institute of Basic Medical Sciences & Shaanxi Key Laboratory of Brain DisordersShaanxi Key Laboratory of Ischemic Cardiovascular DiseaseXi'an Medical University Xi'an China
| | - Xingchun Gou
- Institute of Basic Medical Sciences & Shaanxi Key Laboratory of Brain DisordersShaanxi Key Laboratory of Ischemic Cardiovascular DiseaseXi'an Medical University Xi'an China
| | - Lixian Xu
- Institute of Basic Medical Sciences & Shaanxi Key Laboratory of Brain DisordersShaanxi Key Laboratory of Ischemic Cardiovascular DiseaseXi'an Medical University Xi'an China
- State Key Laboratory of Military StomatologyDepartment of AnesthesiologySchool of StomatologyThe Fourth Military Medical University Xi'an China
| |
Collapse
|
24
|
Huang Y, Lei L, Liu Y. Propofol Improves Sensitivity of Lung Cancer Cells to Cisplatin and Its Mechanism. Med Sci Monit 2020; 26:e919786. [PMID: 32225124 PMCID: PMC7142322 DOI: 10.12659/msm.919786] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cisplatin (cis-diamminedichloroplatinum, DDP) resistance is identified as the primary obstacle during lung cancer treatment, while DDP resistance is exist extensively. This report was to investigate the roles of propofol in lung cancer cells tolerance to DDP and the potential mechanisms. MATERIAL AND METHODS A549 and A549/DDP cells were treated with DDP for 48 hours, and cell proliferation suppression rate was detected by MTT (thiazolyl blue tetrazolium bromide) assay and half maximal inhibitory concentration (IC₅₀) of DDP to lung cancer cells was calculated. Besides, cell proliferation and apoptosis were determined by MTT assay and flow cytometry assay respectively in propofol-treated A549/DDP and A549 cells. Furthermore, we performed MTT assay to determine the influence of propofol on the sensitivity of lung cancer cells to DDP. RESULTS The results demonstrated that the IC₅₀ of DDP to A549 cells was lower than that in A549/DDP cells. Propofol dramatically inhibited cell proliferation and promoted cell apoptosis of A549/DDP and A549 cells. In addition, propofol significantly improved the anti-proliferative impact of DDP in A549/DDP and A549 cells, and the value of IC₅₀ for DDP in the A549/DDP and A549 cells were decreased after propofol treatment compare to the control group. Moreover, propofol inhibited the Wnt/ß-catenin pathway in a dose-dependent manner in both A549/DDP and A549 cells. CONCLUSIONS Our report indicated that propofol could control lung cancer cell proliferation and apoptosis, and stimulated the suppression function of DDP on lung cancer cell multiplication via the Wnt/ß-catenin signaling pathway, and also provided a new treatment for DDP tolerance to cure lung cancer in clinical.
Collapse
Affiliation(s)
- Yunfeng Huang
- Department of Anesthesia, Hubei Cancer Hospital, Wuhan, Hubei, China (mainland)
| | - Lirong Lei
- Department of Anesthesia, Hubei Cancer Hospital, Wuhan, Hubei, China (mainland)
| | - Yishu Liu
- Department of Anesthesia, Hubei Cancer Hospital, Wuhan, Hubei, China (mainland)
| |
Collapse
|
25
|
Sun Y, Peng YB, Ye LL, Ma LX, Zou MY, Cheng ZG. Propofol inhibits proliferation and cisplatin resistance in ovarian cancer cells through regulating the microRNA‑374a/forkhead box O1 signaling axis. Mol Med Rep 2020; 21:1471-1480. [PMID: 32016462 PMCID: PMC7003056 DOI: 10.3892/mmr.2020.10943] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 11/18/2019] [Indexed: 12/15/2022] Open
Abstract
Ovarian cancer is a prominent disease that demonstrates high incidence rates in women and often presents multidrug resistance. Propofol has been demonstrated to suppress the malignancy of various types of human cancer; however, the underlying molecular mechanisms of propofol in ovarian cancer remain largely unknown. The present study aimed to investigate whether and how propofol inhibits proliferation and cisplatin (DDP) resistance in ovarian cancer cells. Ovarian cancer cell viability was assessed by the Cell Counting kit-8 assay; apoptosis and cell cycle progression were determined by flow cytometry; the relative expression levels of microRNA (miR)-374a and forkhead box O1 (FOXO1) were analyzed using reverse transcription-quantitative PCR; the binding ability of miR-374a to FOXO1 was assessed by the dual-luciferase reporter assay; cellular sensitivity to DDP was detected using the MTT assay; and finally, the protein expression levels of FOXO1, p27, and Bcl-2-like-protein 11 (Bim) were analyzed by western blotting. Propofol reduced viability, promoted apoptosis and decreased miR-374a expression levels in A2780 cells. In addition, the viability of A2780/DDP cells in the propofol + DDP treatment group was significantly inhibited, and the apoptotic rate was increased. In addition, miR-374a overexpression increased cell viability and the proportion of cells in the S phase, and decreased the proportion of cells in the G0/G1 phase. Conversely, genetic knockdown of miR-374a exerted the opposite effects on cell viability and cell cycle progression. Moreover, miR-374a was demonstrated to bind to FOXO1. Propofol promoted the expression of FOXO1, p27 and Bim, induced cell cycle arrest and decreased ovarian cancer cell viability. In addition, treatment with propofol and DDP regulated FOXO1 and increased apoptosis of ovarian cancer cells. In conclusion, propofol downregulated miR-374a and modulated the FOXO1 pathway to reduce proliferation and DDP resistance in ovarian cancer cells.
Collapse
Affiliation(s)
- Yang Sun
- Department of Anesthesiology, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330003, P.R. China
| | - Yong-Bao Peng
- Department of Anesthesiology, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330003, P.R. China
| | - Ling-Ling Ye
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Long-Xian Ma
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Mei-Yan Zou
- Department of Obstetrics and Gynecology, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi 330003, P.R. China
| | - Zhong-Gui Cheng
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Li Y, Dong W, Yang H, Xiao G. Propofol suppresses proliferation and metastasis of colorectal cancer cells by regulating miR-124-3p.1/AKT3. Biotechnol Lett 2020; 42:493-504. [PMID: 31894425 DOI: 10.1007/s10529-019-02787-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Propofol, an extensively used intravenous anesthetic agents during cancer resection surgery, has been confirmed to execute anti-tumor effect on multiple cancers, including colorectal cancer (CRC). Although the role of propofol in CRC has been previously reported, its action mechanism remains poorly understood. This study further explored the biological function and underlying mechanism of propofol in CRC cells. METHODS The cell proliferation, migration and invasion were assessed by methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay, wound healing assay and transwell assay, respectively. The expression levels microRNA-124-3p.1 (miR-124-3p.1) and AKT serine/threonine kinase 3 (AKT3) was analyzed by quantitative real-time polymerase chain reaction. Western blot assay was employed to measure the protein expression of MMP-9, Vimentin and Cyclin D1. The interaction between miR-124-3p.1 and AKT3 was predicted by TargetScan and confirmed by dual-luciferase reporter assay. RESULTS Propofol inhibited CRC cell proliferation, migration and invasion. Knockdown of miR-124-3p.1 or AKT3 upregulation reversed the inhibitory effects of propofol on CRC cell proliferation and metastasis. Besides, AKT3 was a direct target of miR-124-3p.1 and its overexpression abated the anti-tumor effect of miR-124-3p.1 on CRC cell proliferation and metastasis. CONCLUSION Propofol inhibited CRC cell proliferation, migration and invasion by upregulating miR-124-3p.1 and downregulating AKT3, providing a new sight for propofol treatment of CRC.
Collapse
Affiliation(s)
- Yujin Li
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, Jin Bi Road, Xishan District, Kunming, 650000, Yunnan, China
| | - Wangjun Dong
- Department of Anesthesiology, Yongping County People's Hospital, Dali, 672600, Yunnan, China
| | - Hao Yang
- Department of Anesthesiology, The Second People's Hospital of Kunming, Kunming, 650000, Yunnan, China
| | - Gaopeng Xiao
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, Jin Bi Road, Xishan District, Kunming, 650000, Yunnan, China.
| |
Collapse
|
29
|
Harrington BS, Annunziata CM. NF-κB Signaling in Ovarian Cancer. Cancers (Basel) 2019; 11:cancers11081182. [PMID: 31443240 PMCID: PMC6721592 DOI: 10.3390/cancers11081182] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/05/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022] Open
Abstract
The NF-κB signaling pathway is a master and commander in ovarian cancer (OC) that promotes chemoresistance, cancer stem cell maintenance, metastasis and immune evasion. Many signaling pathways are dysregulated in OC and can activate NF-κB signaling through canonical or non-canonical pathways which have both overlapping and distinct roles in tumor progression. The activation of canonical NF-κB signaling has been well established for anti-apoptotic and immunomodulatory functions in response to the tumor microenvironment and the non-canonical pathway in cancer stem cell maintenance and tumor re-initiation. NF-κB activity in OC cells helps to create an immune-evasive environment and to attract infiltrating immune cells with tumor-promoting phenotypes, which in turn, drive constitutive NF-κB activation in OC cells to promote cell survival and metastasis. For these reasons, NF-κB is an attractive target in OC, but current strategies are limited and broad inhibition of this major signaling pathway in normal physiological and immunological functions may produce unwanted side effects. There are some promising pre-clinical outcomes from developing research to target and inhibit NF-κB only in the tumor-reinitiating cancer cell population of OC and concurrently activate canonical NF-κB signaling in immune cells to promote anti-tumor immunity.
Collapse
|
30
|
Huang G, Liu X, Zhao X, Zhao J, Hao J, Ren J, Chen Y. MiR-9 promotes multiple myeloma progression by regulating TRIM56/NF-κB pathway. Cell Biol Int 2019; 43:1223-1233. [PMID: 30637864 DOI: 10.1002/cbin.11104] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 01/11/2019] [Indexed: 12/14/2022]
Abstract
miR-9 has been reported to play a pivotal role in multiple human cancers by acting as an oncogene or tumor suppressor. In this study, we explored the possible role and molecular mechanism of miR-9 in multiple myeloma (MM). The miR-9 expression was examined by quantitative real-time polymerase chain reaction assay. Transfection with miR-9-mimics, miR-9-inhibitor, pcDNA-TRIM56, or si-TRIM56 into cells was used to change the expression levels of miR-9 and TRIM56. Western blot analysis was used to detect the expression of TRIM56, p65, p-p65, IκBα, and p-IκBα. The potential target of miR-9 was confirmed by luciferase reporter assay. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT) assay, colony formation assay, and flow cytometry were used to assess the abilities of cell proliferation and apoptosis. miR-9 was upregulated in MM patients and cell lines, and miR-9 overexpression promoted proliferation and repressed apoptosis in MM cell lines. TRIM56 was confirmed as a target of miR-9. Moreover, TRIM56 reversed miR-9-mediated pro-proliferation and anti-apoptosis effect on MM cell lines. Furthermore, nuclear factor-κB (NF-κB) pathway was involved in miR-9/TRIM56-mediated regulation on MM cell lines. miR-9 promoted the development and progression of MM by regulating TRIM56/NF-κB pathway, thereby providing a potential microRNA-based target for MM therapy.
Collapse
Affiliation(s)
- Guoqiang Huang
- Department of Hematology, Hanzhong Central Hospital, Hanzhong, 723000, China
| | - Xiaopeng Liu
- Department of Hematology, Hanzhong Central Hospital, Hanzhong, 723000, China
| | - Xiaoying Zhao
- Department of Hematology, Hanzhong Central Hospital, Hanzhong, 723000, China
| | - Jing Zhao
- Department of Hematology, the First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China
| | - Jinxia Hao
- Department of Hematology, the First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China
| | - Juan Ren
- Department of Hematology, the First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China
| | - Ying Chen
- Department of Hematology, the First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China
| |
Collapse
|
31
|
Que YM, Fan XQ, Lin XJ, Jiang XL, Hu PP, Tong XY, Tan QY. Size dependent anti-invasiveness of silver nanoparticles in lung cancer cells. RSC Adv 2019; 9:21134-21138. [PMID: 35521328 PMCID: PMC9066013 DOI: 10.1039/c9ra03662h] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 06/20/2019] [Indexed: 02/04/2023] Open
Abstract
The molecular mechanism of cancer cell death caused by silver nanoparticles (AgNPs) of different sizes is investigated. Compared with the larger nanoparticles, 13 nm AgNPs significantly inhibit the migration and invasiveness of lung adenocarcinoma A549 cells, induce elevated reactive oxygen species and lead to NF-κB directed cellular apoptosis.
Collapse
Affiliation(s)
- Yu Mei Que
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University Chongqing 401331 PR China
| | - Xiao Qing Fan
- Department of Thoracic Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University Chongqing 400042 P. R. China
| | - Xiao Juan Lin
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University Chongqing 401331 PR China
| | - Xiao Li Jiang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University Chongqing 401331 PR China
| | - Ping Ping Hu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University Chongqing 401331 PR China
| | - Xiao Yong Tong
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University Chongqing 401331 PR China
| | - Qun You Tan
- Department of Thoracic Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University Chongqing 400042 P. R. China
| |
Collapse
|
32
|
Yu H, Ma M, Wang X, Zhou Z, Li R, Guo Q. Propofol suppresses proliferation, invasion, and migration of human melanoma cells via regulating microRNA‐137 and fibroblast growth factor 9. J Cell Physiol 2019; 234:23279-23288. [PMID: 31134615 DOI: 10.1002/jcp.28896] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/10/2019] [Accepted: 05/14/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Hong Yu
- Department of Anesthesiology Cangzhou Central Hospital Cangzhou Hebei People's Republic of China
| | - Meina Ma
- Department of Anesthesiology Cangzhou Central Hospital Cangzhou Hebei People's Republic of China
| | - Xupeng Wang
- Department of Anesthesiology Cangzhou Central Hospital Cangzhou Hebei People's Republic of China
| | - Zhenzhen Zhou
- Department of Anesthesiology Cangzhou Central Hospital Cangzhou Hebei People's Republic of China
| | - Rui Li
- Department of Anesthesiology Cangzhou Central Hospital Cangzhou Hebei People's Republic of China
| | - Qingduo Guo
- Department of Anesthesiology Cangzhou Central Hospital Cangzhou Hebei People's Republic of China
| |
Collapse
|
33
|
Gao Y, Yu X, Zhang F, Dai J. Propofol inhibits pancreatic cancer progress under hypoxia via ADAM8. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2019; 26:219-226. [PMID: 30945470 DOI: 10.1002/jhbp.624] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND To investigate the potential anti-tumoral properties of propofol in pancreatic cancer and elucidate the underlying mechanisms. METHODS The relative expression of ADAM metallopeptidase domain 8 (ADAM8) in response to hypoxia in Panc1 cells was analyzed by western blotting. The enzymatic activity was determined by fluorescence release from PEPDAB013 decomposition. Cell growth was measured via cell counting and cell viability was measured using CCK-8 kit. Cell migrative capacity was evaluated by transwell and adhesion assay. The relative abundance of angiogenesis-related markers including platelet-derived growth factor AA, angiogenin, endothelin-1 and vascular endothelial growth factor were determined by real-time polymerase chain reaction and western blotting. The anti-tumoral activity of propofol was investigated with Panc1-derived xenograft mice model. RESULTS ADAM8 was significantly induced by hypoxia and efficiently inhibited by co-treatment with propofol. Propofol suppressed proliferation and compromised viability of Panc1 cells. In addition, the migrative capacity was greatly inhibited by propofol dosage. Comprehensive profiling of angiogenesis-related markers demonstrated that propofol remarkably suppressed neovascularization response in Panc1 cells under hypoxia. We further uncovered that propofol administration via subcutaneous injection delayed xenograft tumor progression. CONCLUSION Propofol specifically inhibited ADAM8 expression and activation in response to hypoxia in pancreatic cancer, and held great value for therapeutic effects.
Collapse
Affiliation(s)
- Yutong Gao
- Department of Biomedicine, Guizhou University, Guiyang, Guizhou, China.,Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Xiangdi Yu
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Fangxiang Zhang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Jing Dai
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| |
Collapse
|
34
|
Yu X, Gao Y, Zhang F. Propofol inhibits pancreatic cancer proliferation and metastasis by up‐regulating miR‐328 and down‐regulating ADAM8. Basic Clin Pharmacol Toxicol 2019; 125:271-278. [PMID: 30861616 DOI: 10.1111/bcpt.13224] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 03/04/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Xiangdi Yu
- Department of Anesthesiology Guizhou Provincial People’s Hospital Guiyang China
| | - Yutong Gao
- Department of Anesthesiology Guizhou Provincial People’s Hospital Guiyang China
- Department of Biomedicine Guizhou University Guiyang China
| | - Fangxiang Zhang
- Department of Anesthesiology Guizhou Provincial People’s Hospital Guiyang China
| |
Collapse
|
35
|
Chen F, Li M, Zhu X. RETRACTED: Propofol suppresses proliferation and migration of papillary thyroid cancer cells by down-regulation of lncRNA ANRIL. Exp Mol Pathol 2019; 107:68-76. [PMID: 30703346 DOI: 10.1016/j.yexmp.2019.01.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/13/2018] [Accepted: 01/26/2019] [Indexed: 12/18/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief as there are concerns about the reliability of the results included in the article. The journal was initially contacted by the corresponding author to request the retraction of the article. Given the comments of Dr Elisabeth Bik https://scienceintegritydigest.com/2020/02/21/the-tadpole-paper-mill/ regarding this article, the journal requested the author to provide the raw data. However, the author was not able to fulfil this request.
Collapse
Affiliation(s)
- Fumei Chen
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Mengyuan Li
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Xiaoping Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China.
| |
Collapse
|
36
|
Xue JJ, Zhang LY, Hou HJ, Li Y, Liang WS, Yang KH. Protective effect of propofol on hydrogen peroxide-induced human esophageal carcinoma via blocking the Wnt/β-catenin signaling pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 21:1297-1304. [PMID: 30627375 PMCID: PMC6312680 DOI: 10.22038/ijbms.2018.29141.7039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective(s): To analyze the potential influences of propofol on the oxidative stress of H2O2-induced human esophageal squamous cell carcinoma (ESCC) Eca109 cell through mediating the Wnt/β-catenin signaling pathway. Materials and Methods: Eca109 cells were classified into 5 groups: Control group, H2O2 group, Propofol + H2O2 group, Dkk1 (Dickkopf-1, Wnt/β-catenin pathway antagonist) + H2O2 group, and Propofol + LiCl (Lithium chloride, Wnt/β-catenin pathway agonist) + H2O2 group. Western blotting was performed to determine the protein expressions, flow cytometry to measure the content of ROS, immunofluorescence staining to detect the oxidative DNA damage, as well as MTT, AnnexinV-FITC/PI, Wound-healing, and Transwell assays to test the biological characteristics of Eca109 cells. Results: H2O2 resulted in the increased nuclear and cytoplasmatic expression of β-catenin, reduced p-GSK3β expression, up-regulated ROS content, and induced oxidative DNA damage in Eca109 cells. Moreover, Eca109 cells treated with H2O2 alone had enhanced cell proliferation and metastasis but decreased cell apoptosis, as compared with those without any treatment; meanwhile, the declined Cyt C, Bax, and cleaved caspase-3, as well as the elevated Bcl-2 were also observed in Eca109 cells in the H2O2 group, which were reversed by Propofol or Dkk1. Moreover, Propofol could inhibit the effect of LiCl on activating the Wnt/β-catenin signaling pathway in H2O2-induced Eca109 cells. Conclusion: Propofol elicits protective effects to inhibit H2O2-induced proliferation and metastasis and promote apoptosis of Eca109 cells via blocking the Wnt/β-catenin pathway, offering a possible therapeutic modality for ESCC.
Collapse
Affiliation(s)
- Jian-Jun Xue
- Evidence Based Medicine Centre, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou 730000, China.,Gansu Provincial Hospital of TCM, Lanzhou 730050, China
| | | | - Huai-Jing Hou
- Gansu Provincial Hospital of TCM, Lanzhou 730050, China
| | - Yan Li
- Gansu Provincial Hospital of TCM, Lanzhou 730050, China
| | | | - Ke-Hu Yang
- Evidence Based Medicine Centre, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou 730000, China
| |
Collapse
|
37
|
Liu WZ, Liu N. Propofol Inhibits Lung Cancer A549 Cell Growth and Epithelial-Mesenchymal Transition Process by Upregulation of MicroRNA-1284. Oncol Res 2018; 27:1-8. [PMID: 29402342 PMCID: PMC7848461 DOI: 10.3727/096504018x15172738893959] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Propofol has been widely used in lung cancer resections. Some studies have demonstrated that the effects of propofol might be mediated by microRNAs (miRNAs). This study aimed to investigate the effects and mechanisms of propofol on lung cancer cells by regulation of miR-1284. A549 cells were treated with different concentrations of propofol, while transfected with miR-1284 inhibitor, si-FOXM1, and their negative controls. Cell viability, migration, and invasion, and the expression of miR-1284, FOXM1, and epithelial-mesenchymal transition (EMT) factors were detected by CCK-8, Transwell, qRT-PCR, and Western blot assays, respectively. In addition, the regulatory and binding relationships among propofol, miR-1284, and FOXM1 were assessed, respectively. Results showed that propofol suppressed A549 cell viability, migration, and invasion, upregulated E-cadherin, and downregulated N-cadherin, vimentin, and Snail expressions. Moreover, propofol significantly promoted the expression of miR-1284. miR-1284 suppression abolished propofol-induced decreases of cell viability, migration, and invasion, and increased FOXM1 expression and the luciferase activity of FOXM1-wt. Further, miR-1284 negatively regulated FOXM1 expression. FOXM1 knockdown reduced cell viability, migration, and invasion by propofol treatment plus miR-1284 suppression. In conclusion, our study indicated that propofol could inhibit cell viability, migration, invasion, and the EMT process in lung cancer cells by regulation of miR-1284.
Collapse
Affiliation(s)
- Wei-Zhen Liu
- Department of Anesthesia, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, P.R. China
| | - Nian Liu
- Department of Anesthesia, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, P.R. China
| |
Collapse
|
38
|
Sun H, Gao D. Propofol suppresses growth, migration and invasion of A549 cells by down-regulation of miR-372. BMC Cancer 2018; 18:1252. [PMID: 30547768 PMCID: PMC6295097 DOI: 10.1186/s12885-018-5175-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 12/03/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Propofol, a commonly used intravenous anesthetic during cancer resection surgery, has been found to exhibit tumor inhibitory effects in vitro and in vivo. The role of propofol in lung cancer has been previously reported, whereas its action mechanism remains unclear. This study further investigated the effects of propofol on lung cancer A549 cell growth, migration and invasion, as well as the underlying mechanisms. METHODS Cell viability, proliferation, migration, invasion and apoptosis were assessed by CCK-8 assay, BrdU assay, two chamber transwell assay and flow cytometry, respectively. The regulatory effect of propofol on microRNA-372 (miR-372) expression in A549 cells was analyzed by qRT-PCR. Cell transfection was used to change the expression of miR-372. The protein expression of key factors involving in cell proliferation, apoptosis, migration and invasion, as well as Wnt/β-catenin and mTOR pathways were analyzed by western blotting. RESULTS Propofol inhibited lung cancer A549 cell viability, proliferation, migration, and invasion, but promoted cell apoptosis. Moreover, miR-372 was down-regulated in propofol-treated A549 cells. Overexpression of miR-372 abrogated the effects of propofol on proliferation, migration, invasion and apoptosis of A549 cells. Knockdown of miR-372 had opposite effects. Furthermore, propofol suppressed Wnt/β-catenin and mTOR signaling pathways by down-regulating miR-372. CONCLUSION Propofol inhibits growth, migration and invasion of lung cancer A549 cells at least in part by down-regulating miR-372 and then inactivating Wnt/β-catenin and mTOR pathways.
Collapse
Affiliation(s)
- Hai Sun
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, No.126, Xiantai Street, Changchun, Jilin, 130033, China
| | - Dengyu Gao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, No.126, Xiantai Street, Changchun, Jilin, 130033, China.
| |
Collapse
|
39
|
Mao S, Ma J, Yu H. Sirtuin-7 knockdown inhibits the growth of endometrial cancer cells by inducing apoptosis via the NF-κB signaling pathway. Oncol Lett 2018; 17:937-943. [PMID: 30655851 PMCID: PMC6312928 DOI: 10.3892/ol.2018.9698] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 01/22/2018] [Indexed: 12/21/2022] Open
Abstract
Sirtuin-7 is an evolutionarily conserved NAD-dependent deacetylase, which serves an important role in carcinogenesis. However, the potential mechanism of sirtuin-7 in endometrial cancer has not yet been investigated. The purpose of the present study was to investigate whether sirtuin-7 exhibits inhibitory effects on endometrial cancer cells. The potential mechanisms mediated by sirtuin-7 in endometrial cancer cells were also investigated. The expression levels of sirtuin-7 in endometrial cancer cells were compared with normal endometrial cells using western blotting. The results demonstrated that sirtuin-7 is overexpressed in endometrial cancer cells compared with normal endometrial cells. The downregulation of sirtuin-7 inhibited the growth and invasiveness of endometrial cancer cells. The knockdown of sirtuin-7 was observed to increase the sensitivity of the endometrial cancer cells to cisplatin treatment in vitro. An investigation into the potential molecular mechanism demonstrated that sirtuin-7 knockdown promoted the apoptosis of endometrial cancer cells by regulating the nuclear factor (NF)-κB signaling pathway. The knockdown of sirtuin-7 inhibited NF-κB expression and resulted in a decrease in the expression of NF-κB target proteins that are anti-apoptotic: Bcl-xl, Bcl-2 and Mcl-1. Sirtuin-7 knockdown also resulted in an increase of the NF-κB target proteins that are pro-apoptotic: Caspase-3, Bad and Bax. In conclusion, the present study demonstrated that sirtuin-7 knockdown was able to markedly inhibit the growth of endometrial cancer cells, suggesting that sirtuin-7 may be a potential therapeutic target for endometrial cancer therapy.
Collapse
Affiliation(s)
- Shiqin Mao
- Department of Gynaecology, People's Hospital of Jingjiang, Jingjiang, Jiangsu 214500, P.R. China
| | - Jimin Ma
- Department of Gynaecology, The Third People's Hospital of Liaocheng, Liaocheng, Shandong 252000, P.R. China
| | - Hong Yu
- Department of Critical Care Medicine, Harbin Second Hospital, Harbin, Heilongjiang 150036, P.R. China
| |
Collapse
|
40
|
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
|
41
|
Yu B, Gao W, Zhou H, Miao X, Chang Y, Wang L, Xu M, Ni G. Propofol induces apoptosis of breast cancer cells by downregulation of miR-24 signal pathway. Cancer Biomark 2018; 21:513-519. [PMID: 29103019 DOI: 10.3233/cbm-170234] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND OBJECTIVE Propofol, an intravenous anesthetic agent, has been found to inhibit growth of breast cancer cells. However, the mechanisms underlying the antitumor are not known. A recent report has found that propofol could significantly downregulate miR-24 expression in the human malignant cancers. In breast cancer cells, overexpression of miR-24 promotes cell proliferation and inhibits cell apoptosis by downregulation of p27. The miR-24 has been reported to be overexpressed in breast cancer and breast cancer cell lines. In the present study, we hypothesized that propofol induces apoptosis of breast cancer cells by miR-24/p27 signal pathway. METHODS Breast cancer MDA-MB-435 cells were exposed to propofol (10 μM) for 6 hr and cell death was assessed using TUNEL staining, Flow cytometry and cleaved caspase-3 expression. microRNA-24 (miR-24) expression was assessed using quantitative reverse transcription polymerase chain reaction (qRT-PCR). miR-24 was overexpressed using a miR-24 mimic. P27 was knocked down using a small interfering RNA. p27 and cleaved caspase-3 expression was assessed by Western blot. RESULTS MDA-MB-435 exposed to propofol showed a significant increase in apoptotic cells, followed by the downregulation of miR-24, upregulation of p27 expression and cleaved caspase-3 expression. Targeting p27 inhibits propofol-induced cell apoptosis; miR-24 overexpression decreased propofol-induced cell apoptosis, cleaved caspase-3 and p27 expression. CONCLUSIONS Propofol inducescell death in MDA-MB-435 cells via inactivation of miR-24/p27 signal pathway.
Collapse
Affiliation(s)
- Benxia Yu
- Department of Imaging, Yantai Yuhuangding Hospital, Yantai, Shandong, China.,Department of Imaging, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Wei Gao
- Department of Imaging, Yantai Yuhuangding Hospital, Yantai, Shandong, China.,Department of Imaging, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Hui Zhou
- Department of Anesthesiology, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Xia Miao
- Department of Clinical Laboratory, The Affiliated Hospital of Weifang Medical College, Weifang, Shandong, China
| | - Yuan Chang
- Department of Anesthesiology, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Liping Wang
- Department of Imaging, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Miao Xu
- Department of Clinical Laboratory, The Affiliated Hospital of Weifang Medical College, Weifang, Shandong, China
| | - Guangzhen Ni
- Department of Clinical Laboratory, The Affiliated Hospital of Weifang Medical College, Weifang, Shandong, China
| |
Collapse
|
42
|
Jiang S, Liu Y, Huang L, Zhang F, Kang R. Effects of propofol on cancer development and chemotherapy: Potential mechanisms. Eur J Pharmacol 2018; 831:46-51. [DOI: 10.1016/j.ejphar.2018.04.009] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 04/08/2018] [Accepted: 04/09/2018] [Indexed: 12/30/2022]
|
43
|
Propofol attenuates mast cell degranulation via inhibiting the miR-221/PI3K/Akt/Ca 2+ pathway. Exp Ther Med 2018; 16:1426-1432. [PMID: 30116391 DOI: 10.3892/etm.2018.6317] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 03/29/2018] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to investigate the effect of propofol on immunoglobulin (Ig)E-activated mast cell degranulation and explore the underlying mechanisms responsible. RBL-2H3 cells were treated with propofol for at a variety of concentrations and different amounts of time. Cell viability was assessed using an MTT assay and microRNA (miR)-221 expression was quantified using reverse transcription-quantitative polymerase chain reaction. RBL-2H3 cells were transfected with miR-221 mimic or a negative control and degranulation, including the release of β-hexosaminidase and histamine, was evaluated using an ELISA kit. The effect of miR-221 overexpression on the phosphorylation of protein kinase B (Akt) was detected using western blotting and extracellular Ca2+ influx was measured via afura-2 assay. The phosphoinositide 3-kinase(PI3K) inhibitor LY294002 was used to investigate the association between PI3K/Akt signaling and Ca2+ influx in the presence of propofol. The results demonstrated that propofol treatment suppressed RBL-2H3 cell proliferation in a dose- and time-dependent manner. Propofol inhibited miR-221 expression in a dose-dependent manner compared with the control group; however, the inhibitive effect was significantly abrogated following transfection with miR-221 mimics. Furthermore, β-hexosaminidase and histamine release, PI3K/Akt signaling and Ca2+ influx were decreased following propofol application. miR-221 overexpression markedly ameliorated the suppressive effect of propofol. Treatment with LY294002 reversed the propofol-induced decrement of Ca2+ influx on IgE-mediated RBL-2H3 cells, suggesting an association between PI3K/Akt signaling and Ca2+ influx. In conclusion, the results of the present study suggest that propofol treatment attenuates mast cell degranulation via inhibiting the miR-221/PI3K/Akt/Ca2+ pathway. These results indicate that propofol may have a potential therapeutic effect as a treatment for allergic diseases.
Collapse
|
44
|
Xu K, Tao W, Su Z. Propofol prevents IL-13-induced epithelial-mesenchymal transition in human colorectal cancer cells. Cell Biol Int 2018; 42:985-993. [PMID: 29569786 DOI: 10.1002/cbin.10964] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/17/2018] [Indexed: 12/17/2022]
Abstract
Accumulating evidence showed that cytokines are involved in the development of cancer. IL-13 was showed to induce epithelial-mesenchymal transition and promote metastasis in colorectal cancer, providing a promising therapeutic target for cancer patients. Interestingly, recent studies showed that propofol, one of most common intravenous anesthetic agent, may have antitumor function in different cancer type. However, the impact of propofol on colorectal cancer and IL-13 induced epithelial-mesenchymal transition remains unknown. Herein, we found that propofol can effectively suppress cell proliferation in colorectal cell lines RKO and SW480 cells by using MTT assay. Furthermore, wound healing assay and migration assay demonstrated that propofol has the ability to inhibit epithelial-mesenchymal transition that induced by IL-13 in RKO and SW480 cells. Mechanistically, we found propofol treatment causes up-regulation of miR-361 and miR-135b, that suppress expression of STAT6 and thereafter leads to the inhibition of IL-13/STAT6/ZEB1 signaling pathway. In conclusion, our data for the first time demonstrated that propofol may serve as a novel therapeutic drug for targeting IL-13. The aggressive function of IL-13/STAT6/ZEB1 axis in colorectal cancer was impaired by propofol through miR-361 and miR-135b.
Collapse
Affiliation(s)
- Kejia Xu
- Department of Anaesthesiology, Tongren Hospital, Shanghai JiaoTong University School of Medicine, 1111 Xian Xia Road, Shanghai 200336, China
| | - Weimin Tao
- Department of Anaesthesiology, Tongren Hospital, Shanghai JiaoTong University School of Medicine, 1111 Xian Xia Road, Shanghai 200336, China
| | - Zhe Su
- Department of Anaesthesiology, Tongren Hospital, Shanghai JiaoTong University School of Medicine, 1111 Xian Xia Road, Shanghai 200336, China
| |
Collapse
|
45
|
Du Q, Liu J, Zhang X, Zhang X, Zhu H, Wei M, Wang S. Propofol inhibits proliferation, migration, and invasion but promotes apoptosis by regulation of Sox4 in endometrial cancer cells. ACTA ACUST UNITED AC 2018; 51:e6803. [PMID: 29490000 PMCID: PMC5856446 DOI: 10.1590/1414-431x20176803] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 12/14/2017] [Indexed: 01/27/2023]
Abstract
Propofol is an intravenous sedative hypnotic agent of which the growth-inhibitory effect has been reported on various cancers. However, the roles of propofol in endometrial cancer (EC) remain unclear. This study aimed to explore the effects of propofol on EC in vitro and in vivo. Different concentrations of propofol were used to treat Ishikawa cells. Colony number, cell viability, cell cycle, apoptosis, migration, and invasion were analyzed by colony formation, MTT, flow cytometry, and Transwell assays. In addition, the pcDNA3.1-Sox4 and Sox4 siRNA plasmids were transfected into Ishikawa cells to explore the relationship between propofol and Sox4 in EC cell proliferation. Tumor weight in vivo was measured by xenograft tumor model assay. Protein levels of cell cycle-related factors, apoptosis-related factors, matrix metalloproteinases 9 (MMP9), matrix metalloproteinases 2 (MMP2) and Wnt/β-catenin pathway were examined by western blot. Results showed that propofol significantly decreased colony numbers, inhibited cell viability, migration, and invasion but promoted apoptosis in a dose-dependent manner in Ishikawa cells. Moreover, propofol reduced the expression of Sox4 in a dose-dependent manner. Additionally, propofol significantly suppressed the proportions of Ki67+ cells, but Sox4 overexpression reversed the results. Furthermore, in vivo assay results showed that propofol inhibited tumor growth; however, the inhibitory effect was abolished by Sox4 overexpression. Moreover, propofol inhibited Sox4 expression via inactivation of Wnt/β-catenin signal pathway. Our study demonstrated that propofol inhibited cell proliferation, migration, and invasion but promoted apoptosis by regulation of Sox4 in EC cells. These findings might indicate a novel treatment strategy for EC.
Collapse
Affiliation(s)
- Qing Du
- Department of Anesthesiology, Qingdao University, Qingdao, China
| | - Jia Liu
- Department of Anesthesiology, Qingdao University, Qingdao, China
| | - Xuezhi Zhang
- Department of Emergency, Qingdao University, Qingdao, China
| | - Xin Zhang
- Department of Anesthesiology, Qingdao University, Qingdao, China
| | - He Zhu
- Department of Anesthesiology, Qingdao University, Qingdao, China
| | - Ming Wei
- Department of Anesthesiology, Qingdao University, Qingdao, China
| | - Shilei Wang
- Department of Anesthesiology, Qingdao University, Qingdao, China
| |
Collapse
|
46
|
Propofol Attenuates Inflammatory Response in LPS-Activated Microglia by Regulating the miR-155/SOCS1 Pathway. Inflammation 2017; 41:11-19. [DOI: 10.1007/s10753-017-0658-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
47
|
Yang Y, Ren M, Song C, Li D, Soomro SH, Xiong Y, Zhang H, Fu H. LINC00461, a long non-coding RNA, is important for the proliferation and migration of glioma cells. Oncotarget 2017; 8:84123-84139. [PMID: 29137410 PMCID: PMC5663582 DOI: 10.18632/oncotarget.20340] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 07/25/2017] [Indexed: 12/31/2022] Open
Abstract
An increasing number of reports have revealed that long non-coding RNAs are important players in tumorigenesis. Here we showed that long non-coding RNA LINC00461 is highly expressed in glioma tissues compared to non-neoplastic brain tissues. The knockdown of LINC00461 suppressed cyclinD1/A/E expression which led to G0/G1 cell cycle arrest and inhibited cell proliferation in glioma cells. LINC00461 suppression also inhibited glioma cell migration and invasion. The function of LINC00461 in glioma cells is partially mediated by MAPK/ERK and PI3K/AKT signaling pathways as down-regulation of LINC00461 expression suppressed ERK1/2 and AKT activities. Moreover, LINC00461 knockdown decreased expression levels of microRNA miR-9 and flanking genes MEF2C and TMEM161B. Taken together, our results demonstrate that LINC00461 is important for glioma progression affecting cell proliferation, migration and invasion via MAPK/ERK, PI3K/AKT, and possibly other signaling pathways.
Collapse
Affiliation(s)
- Yali Yang
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Mingxin Ren
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Chao Song
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Dan Li
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Shahid Hussain Soomro
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yajie Xiong
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Hongfeng Zhang
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Hui Fu
- Department of Anatomy and Embryology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| |
Collapse
|
48
|
魏 国, 杨 菁. MicroRNAs在结直肠癌中的研究进展. Shijie Huaren Xiaohua Zazhi 2017; 25:1928-1933. [DOI: 10.11569/wcjd.v25.i21.1928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
结直肠癌在我国恶性肿瘤中的发病率较高, 且其死亡率居高不下. 目前, 越来越多的学者十分关注微小RNAs(microRNAs, miRNAs)与结直肠癌的关系. miRNAs是普遍存在于生物体内的一类小分子非编码RNA, miRNAs的异常表达与结直肠癌的发生和进展密切相关. miRNAs可以通过转录后基因调控的方式, 来影响肿瘤细胞的增殖、调亡以及对化疗的敏感性等. 在这里, 我们回顾了近年来关于miRNAs与结直肠癌的相关文献, 了解miRNAs在结直肠癌中的表达、结直肠癌化疗耐药以及其与预后的关系, 从而更好的了解结直肠癌进展的生物学过程, 有助于结直肠癌的诊断与治疗, 提高结直肠癌患者的预后.
Collapse
|
49
|
Cui Y, Xue Y, Dong S, Zhang P. Plasma microRNA-9 as a diagnostic and prognostic biomarker in patients with esophageal squamous cell carcinoma. J Int Med Res 2017; 45:1310-1317. [PMID: 28587538 PMCID: PMC5625531 DOI: 10.1177/0300060517709370] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Purpose Emerging evidence indicates that circulating microRNAs (miRs) might act as noninvasive biomarkers for cancer diagnosis and prognosis. We examined the expression pattern and clinical significance of plasma miR-9 in patients with esophageal squamous cell carcinoma (ESCC). Methods Venous blood samples (6 mL) were collected from 131 patients with ESCC and 131 healthy controls, and the plasma miR-9 concentration was detected by reverse transcription polymerase chain reaction. The association of plasma miR-9 expression with clinicopathologic factors and survival of patients with ESCC was evaluated. Receiver operating characteristic (ROC) curve analysis was applied to evaluate the clinical value of plasma miR-9 for ESCC diagnosis. Results The plasma miR-9 expression levels in patients with ESCC were significantly upregulated compared with normal controls. High plasma miR-9 concentrations were significantly correlated with poor tumor differentiation, large tumor size, deep local invasion, lymph node metastasis, advanced clinical stage, and poor survival. ROC curve analysis showed that the plasma miR-9 concentration could efficiently distinguish patients with ESCC from healthy controls. Multivariate survival analysis confirmed plasma miR-9 as an independent prognostic factor for ESCC. Conclusions Plasma miR-9 expression was upregulated in ESCC and might act as a novel diagnostic and prognostic biomarker.
Collapse
Affiliation(s)
- Yuantao Cui
- 1 Department of Cardiothoracic Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, PR China
| | - Yuan Xue
- 2 Department of Cardiovascular Thoracic Surgery, People's Hospital of Rizhao, ShanDong, PR China
| | - Shangwen Dong
- 1 Department of Cardiothoracic Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, PR China
| | - Peng Zhang
- 1 Department of Cardiothoracic Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, PR China
| |
Collapse
|