1
|
Raigon Ponferrada A, Molina Ruiz JC, Romero Molina S, Rodriguez Garcia V, Guerrero Orriach JL. The Role of Anesthetic Drugs and Statins in Prostate Cancer Recurrence: Starting at the Actual Knowledge and Walking through a New Paradigm. Cancers (Basel) 2023; 15:cancers15113059. [PMID: 37297021 DOI: 10.3390/cancers15113059] [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: 04/19/2023] [Revised: 05/18/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Prostate cancer has become a major health problem in men. Its incidence is increasing as the average age of the affected population tends to be higher. Of all the possible treatments, surgery is the gold standard in its treatment. Surgery produces a deregulation in the immune system that can favour the development of distant metastases. Different anesthetic techniques have raised the hypothesis that different anesthetic drugs influence tumor recurrence and prognosis. Some mechanisms are beginning to be understood by which halogenated agents in cancer patients and the use of opioids may negatively affect patients. In this document, we group together all the available evidence on how the different anesthetic drugs affect tumor recurrence in prostate cancer.
Collapse
Affiliation(s)
- Aida Raigon Ponferrada
- Institute of Biomedical Research in Malaga [IBIMA], 29010 Malaga, Spain
- Department of Anaesthesiology, Virgen de la Victoria University Hospital, 29010 Malaga, Spain
- Department of Pharmacology and Pediatrics, School of Medicine, University of Malaga, 29010 Malaga, Spain
| | - Juan Carlos Molina Ruiz
- Department of Anaesthesiology, Virgen de la Victoria University Hospital, 29010 Malaga, Spain
| | - Salvador Romero Molina
- Department of Anaesthesiology, Virgen de la Victoria University Hospital, 29010 Malaga, Spain
| | | | - Jose Luis Guerrero Orriach
- Institute of Biomedical Research in Malaga [IBIMA], 29010 Malaga, Spain
- Department of Anaesthesiology, Virgen de la Victoria University Hospital, 29010 Malaga, Spain
- Department of Pharmacology and Pediatrics, School of Medicine, University of Malaga, 29010 Malaga, Spain
- Hospital Virgen de la Victoria, Campus Teatinos CP Malaga, 29010 Malaga, Spain
| |
Collapse
|
2
|
Araújo D, Ribeiro E, Amorim I, Vale N. Repurposed Drugs in Gastric Cancer. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010319. [PMID: 36615513 PMCID: PMC9822219 DOI: 10.3390/molecules28010319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/21/2022] [Accepted: 12/25/2022] [Indexed: 01/04/2023]
Abstract
Gastric cancer (GC) is one of the major causes of death worldwide, ranking as the fifth most incident cancer in 2020 and the fourth leading cause of cancer mortality. The majority of GC patients are in an advanced stage at the time of diagnosis, presenting a poor prognosis and outcome. Current GC treatment approaches involve endoscopic detection, gastrectomy and chemotherapy or chemoradiotherapy in an adjuvant or neoadjuvant setting. Drug development approaches demand extreme effort to identify molecular mechanisms of action of new drug candidates. Drug repurposing is based on the research of new therapeutic indications of drugs approved for other pathologies. In this review, we explore GC and the different drugs repurposed for this disease.
Collapse
Affiliation(s)
- Diana Araújo
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Eduarda Ribeiro
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Irina Amorim
- Institute of Biomedical Sciences Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Correspondence: ; Tel.: +351-220426537
| |
Collapse
|
3
|
Patrad E, Khalighfard S, Amiriani T, Khori V, Alizadeh AM. Molecular mechanisms underlying the action of carcinogens in gastric cancer with a glimpse into targeted therapy. Cell Oncol 2022; 45:1073-1117. [PMID: 36149600 DOI: 10.1007/s13402-022-00715-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric cancer imposes a substantial global health burden despite its overall incidence decrease. A broad spectrum of inherited, environmental and infectious factors contributes to the development of gastric cancer. A profound understanding of the molecular underpinnings of gastric cancer has lagged compared to several other tumors with similar incidence and morbidity rates, owing to our limited knowledge of the role of carcinogens in this malignancy. The International Agency for Research on Cancer (IARC) has classified gastric carcinogenic agents into four groups based on scientific evidence from human and experimental animal studies. This review aims to explore the potential comprehensive molecular and biological impacts of carcinogens on gastric cancer development and their interactions and interferences with various cellular signaling pathways. CONCLUSIONS In this review, we highlight recent clinical trial data reported in the literature dealing with different ways to target various carcinogens in gastric cancer. Moreover, we touch upon other multidisciplinary therapeutic approaches such as surgery, adjuvant and neoadjuvant chemotherapy. Rational clinical trials focusing on identifying suitable patient populations are imperative to the success of single-agent therapeutics. Novel insights regarding signaling pathways that regulate gastric cancer can potentially improve treatment responses to targeted therapy alone or in combination with other/conventional treatments. Preventive strategies such as control of H. pylori infection through eradication or immunization as well as dietary habit and lifestyle changes may reduce the incidence of this multifactorial disease, especially in high prevalence areas. Further in-depth understanding of the molecular mechanisms involved in the role of carcinogenic agents in gastric cancer development may offer valuable information and update state-of-the-art resources for physicians and researchers to explore novel ways to combat this disease, from bench to bedside. A schematic outlining of the interaction between gastric carcinogenic agents and intracellular pathways in gastric cancer H. pylori stimulates multiple intracellular pathways, including PI3K/AKT, NF-κB, Wnt, Shh, Ras/Raf, c-MET, and JAK/STAT, leading to epithelial cell proliferation and differentiation, apoptosis, survival, motility, and inflammatory cytokine release. EBV can stimulate intracellular pathways such as the PI3K/Akt, RAS/RAF, JAK/STAT, Notch, TGF-β, and NF-κB, leading to cell survival and motility, proliferation, invasion, metastasis, and the transcription of anti-apoptotic genes and pro-inflammatory cytokines. Nicotine and alcohol can lead to angiogenesis, metastasis, survival, proliferation, pro-inflammatory, migration, and chemotactic by stimulating various intracellular signaling pathways such as PI3K/AKT, NF-κB, Ras/Raf, ROS, and JAK/STAT. Processed meat contains numerous carcinogenic compounds that affect multiple intracellular pathways such as sGC/cGMP, p38 MAPK, ERK, and PI3K/AKT, leading to anti-apoptosis, angiogenesis, metastasis, inflammatory responses, proliferation, and invasion. Lead compounds may interact with multiple signaling pathways such as PI3K/AKT, NF-κB, Ras/Raf, DNA methylation-dependent, and epigenetic-dependent, leading to tumorigenesis, carcinogenesis, malignancy, angiogenesis, DNA hypermethylation, cell survival, and cell proliferation. Stimulating signaling pathways such as PI3K/Akt, RAS/RAF, JAK/STAT, WNT, TGF-β, EGF, FGFR2, and E-cadherin through UV ionizing radiation leads to cell survival, proliferation, and immortalization in gastric cancer. The consequence of PI3K/AKT, NF-κB, Ras/Raf, ROS, JAK/STAT, and WNT signaling stimulation by the carcinogenic component of Pickled vegetables and salted fish is the Warburg effect, tumorigenesis, angiogenesis, proliferation, inflammatory response, and migration.
Collapse
Affiliation(s)
- Elham Patrad
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Solmaz Khalighfard
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Taghi Amiriani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Vahid Khori
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Mohammad Alizadeh
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Breast Disease Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Tang YJ, Wu W, Chen QQ, Liu SH, Zheng ZY, Cui ZL, Xu JP, Xue Y, Lin DH. miR-29b-3p suppresses the malignant biological behaviors of AML cells via inhibiting NF-κB and JAK/STAT signaling pathways by targeting HuR. BMC Cancer 2022; 22:909. [PMID: 35986311 PMCID: PMC9392259 DOI: 10.1186/s12885-022-09996-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Background HuR/ELAVL1 (embryonic lethal abnormal vision 1) was a downstream target of miR-29b in some cancer cells. HuR protein exerts important prognostic effects of involving in the pathogenesis and development of acute myeloid leukemia (AML). This study aims to investigate the role of miR-29b-3p in biological behaviors of AML cells by targeting HuR and the involvement of the NF-κB and JAK/STAT signaling pathways. Methods The expressions of HuR and miR-29b-3p in AML cells were determined using RT-qPCR and Western blot, and the association between them was analyzed using the Spearman method. Next, the target relationship between HuR and miR-29b-3p was predicted by biological information databases and verified by the dual-luciferase reporter gene assay. MTS, methyl cellulose, flow cytometry and transwell assay were employed to detect the cell proliferation, clone formation, cell cycle and apoptosis, invasion and migration respectively, the effect of miR-29b-3p targeted HuR on the biological behaviors of AML cells was explored after over- /down-expression of miR-29b-3p and rescue experiment. Then, immunofluorescence assay and western blot were employed to detect location expression and phosphorylation levels of NF-κB and JAK/STAT signaling pathways related molecules respectively. Results HuR was negatively correlated with miR-29b-3p, and was the downstream target of miR-29b-3p in AML cells. When miR-29b-3p was overexpressed in AML cells, HuR was down-regulated, accompanied by cell viability decreased, cell cycle arrest, apoptosis increased, invasion and migration weakened. Moreover, the opposite result appeared after miR-29b-3p was down-regulated. The rescue experiment showed that miR-29b-3p inhibitor could reverse the biological effect of HuR down-regulation in AML cells. Molecular pathway results showed that miR-29b-3p could inhibit p65 expression in nucleus and phosphorylation levels of p65, IκBα, STAT1, STAT3 and STAT5. Conclusion miR-29b-3p can inhibit malignant biological behaviors of AML cells via the inactivation of the NF-κB and JAK/STAT signaling pathways by targeting HuR. miR-29b-3p and its target HuR can be used as a new potential molecular for AML treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09996-1.
Collapse
|
5
|
Effects of mir-195 Targeted Regulation of JAK2 on Proliferation, Invasion, and Apoptosis of Gastric Cancer Cells. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:5873479. [PMID: 35928970 PMCID: PMC9345721 DOI: 10.1155/2022/5873479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/06/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022]
Abstract
Background. Overexpression of miR-195 can make gastric cancer cells stay in G1/G2 phase. miR-195 has been shown to inhibit gastric cancer cell replication and accelerate cell death by targeting JAK2. However, the relationship between miR-195, JAK2, and gastric cancer is not clear. Objective. To observe the effect of mir-195 regulated by JAK2 on the growth, invasion, and death of gastric cancer cells. Methods. MGC803 and NCI gastric N87 cells were introduced into the negative control sequences of miR-195 and RNA, respectively. To detect the expression of miR-195 in cells, to detect the effect of miR-195 on mitosis and proliferation of tumor cells, to analyze the effect of miR-195 on cell invasion and metastasis, and to detect the regulation of miR-195 on JAK2 expression. Results. The level of miR-195 in miR-195-MIMICS group was significantly higher than that in miR-NC group. The cell survival rate of miR-195 mimic group was lower than that of miR-NC group (
). Compared with miR-NC group, the number of cells in G1 phase increased, the cells in G2 phase and S phase decreased, and the proportion of cells in G2 and S phase decreased in miR-195 mimic group. The scratch distance of miR-195 simulator group was larger than that of control group. The number of invasive cells in the miR-195 mimic group was significantly lower than that in the control group. The expression of JAK2 protein in miR-195 mimic group was lower than that in miR-NC group. There was a significant negative correlation between the expression level of miR-195 and JAK2 (rhabdomile 0.326 and record 0.00). There are continuous interaction fragments between JAK2 and miR-195. The luciferase activity of miR-195 mimic and wild type JAK2 sequence expression vector was significantly lower than that of wild type JAK2 sequence expression vector. Conclusion. miR-195 may inhibit the occurrence, metastasis, and invasion of gastric tumor by downregulating the expression of JAK2. miR-195/JAK2 may be a new molecular target for the treatment of gastrointestinal tumors.
Collapse
|
6
|
An Integrative Bioinformatics Analysis of the Potential Mechanisms Involved in Propofol Affecting Hippocampal Neuronal Cells. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:4911773. [PMID: 35515499 PMCID: PMC9064519 DOI: 10.1155/2022/4911773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 12/18/2022]
Abstract
The aim of this study is to probe the possible molecular mechanisms underlying the effects of propofol on HT22 cells. HT22 cells treated with different concentrations were sequenced, and then the results of the sequencing were analyzed for dynamic trends. Expression pattern clustering analysis was performed to demonstrate the expression of genes in the significant trend modules in each group of samples. We first chose the genes related to the trend module for WGCNA analysis, then constructed the PPI network of module genes related to propofol treatment group, and screened the key genes. Finally, GSEA analysis was performed on the key genes. Overall, 2,506 genes showed a decreasing trend with increasing propofol concentration, and 1,871 genes showed an increasing trend with increasing propofol concentration. WGCNA analysis showed that among them, turquoise panel genes were negatively correlated with propofol treatment, and genes with Cor R >0.9 in the turquoise panel were selected for PPI network construction. The MCC algorithm screened a total of five key genes (CD86, IL10RA, PTPRC, SPI1, and ITGAM). GSEA analysis showed that CD86, IL10RA, PTPRC, SPI1, and ITGAM are involved in the PRION_DISEASES pathway. Our study showed that propofol sedation can affect mRNA expression in the hippocampus, providing new ideas to identify treatment of nerve injury induced by propofol anesthesia.
Collapse
|
7
|
Duan L, Wang J, Zhang D, Yuan Y, Tang L, Zhou Y, Jiang X. Immune-Related miRNA-195-5p Inhibits the Progression of Lung Adenocarcinoma by Targeting Polypyrimidine Tract-Binding Protein 1. Front Oncol 2022; 12:862564. [PMID: 35600383 PMCID: PMC9117652 DOI: 10.3389/fonc.2022.862564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose Lung adenocarcinoma (LUAD) is the most common type of cancer and the leading cause of cancer-related death worldwide, resulting in a huge economic and social burden. MiRNA-195-5p plays crucial roles in the initiation and progression of cancer. However, the significance of the miRNA-195-5p/polypyrimidine tract-binding protein 1 (miRNA-195-5p/PTBP1) axis in the progression of lung adenocarcinoma (LUAD) remains unclear. Methods Data were collected from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. The starBase database was employed to examine the expression of miRNA-195-5p, while the Kaplan–Meier plotter, UALCAN, and Gene Expression Profiling Interactive Analysis (GEPIA) databases were utilized to analyze the tumor stage and prognostic value of miRNA and PTBP1. Quantitative reverse transcription-polymerase chain reaction assay was conducted to detect the expression levels of miRNA-195-5p in LUAD cell lines and tissues. The effects of miRNA-195-5p on cell proliferation and migration were examined using the cell growth curve, clone information, transwell assays, and wound healing assays. Results We found that miRNA-195-5p was down-regulated in LUAD cancer and cell lines. Importantly, its low levels were related to the tumor stage, lymph node metastasis, and poor prognosis in LUAD. Overexpression of miR-195-5p significantly inhibited cell growth and migration promotes cell apoptosis. Further study revealed that PTBP1 is a target gene of miRNA-195-5p, and overexpression of miRNA-195-5p inhibited the progression of LUAD by inhibiting PTBP1 expression. MiRNA-195-5p expression was related to immune infiltration in lung adenocarcinoma. Moreover, PTBP1 was negatively correlated with diverse immune cell infiltration and drug sensitivity. Conclusion Our findings uncover a pivotal mechanism that miRNA-195-5p by modulate PTBP1 expression to inhibit the progression of LUAD. MiRNA-195-5p could be a novel diagnostic and prognostic molecular marker for LUAD.
Collapse
Affiliation(s)
- Lincan Duan
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Juan Wang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Dahang Zhang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yixiao Yuan
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lin Tang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yongchun Zhou
- Molecular Diagnostic Center, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Yongchun Zhou, ; Xiulin Jiang,
| | - Xiulin Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, China
- *Correspondence: Yongchun Zhou, ; Xiulin Jiang,
| |
Collapse
|
8
|
Gao F, Li R, Wei PF, Ou L, Li M, Bai Y, Luo WJ, Fan Z. Synergistic anticancer effects of everolimus (RAD001) and Rhein on gastric cancer cells via phosphoinositide-3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway. Bioengineered 2022; 13:6332-6342. [PMID: 35209807 PMCID: PMC8973710 DOI: 10.1080/21655979.2021.2005988] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/09/2021] [Accepted: 11/09/2021] [Indexed: 01/26/2023] Open
Abstract
Everolimus (RAD001) is a mTOR inhibitor and is widely used for the treatment of gastric cancer (GC). Evidence suggests that Rhein has anticancer effect on GC. But the synergistic effect and mechanism of RAD001 and Rhein combination on GC is not clear. The current study aims to clarify the combination of RAD001 and Rhein in GC treatment. We found Rhein dose-dependently repressed MGC-803 cell viability (50% inhibition concentration (IC50) value = 94.26 μM). Rhein (80 μM) significantly suppressed GC cell proliferation and invasion. RAD001 dose-dependently repressed MGC-803 cells viability (IC50 value = 45.41 nM). The combination of Rhein and RAD001 repressed MGC-803 cells viability, invasion, and proliferation compared to the administration of Rhein or RAD001 alone. Protein levels of epithelial-mesenchymal transition (EMT)-related molecules E-cadherin, N-cadherin and Vimentin expressions were significantly affected by the combination of Rhein and RAD001. The combination of Rhein and RAD001 significantly facilitated cell apoptosis and up-regulated expressions of cell apoptosis and cycle-related protein p53, cyclin-dependent kinase 4 (CDK4) and cyclin D1 compared to the administration of Rhein or RAD001 alone. Moreover, the combination of Rhein and RAD001 repressed the expressions of phosphorylation-phosphoinositide-3-kinase (p-PI3K), p-protein kinase B (p-AKT) and p-mammalian target of rapamycin (p-mTOR). Finally, the combination of RAD001 and Rhein significantly decreased tumor weight and volume, suppressed the expressions of p-PI3K, p-Akt and p-mTOR, and repressed cell proliferation marker Ki-67 expression, which exerted synergistic cancer prevention in GC in vivo. Overall, the combination of Rhein and RAD001 exert synergistic cancer prevention in GC via PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Feng Gao
- Teaching and Research Office of Chinese Pharmacy, The College of Pharmacy of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Rui Li
- Department of Emergency, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Pei-Feng Wei
- The Office of Drug Clinical Trial Institution, The Second Affiliated Hospital of Shaanxi University of traditional Chinese Medicine, Xianyang, Shaanxi, China
| | - Li Ou
- Teaching and Research Office of Chinese Pharmacy, The College of Pharmacy of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Min Li
- Teaching and Research Office of Chinese Pharmacy, The College of Pharmacy of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yang Bai
- The Office of Drug Clinical Trial Institution, The Second Affiliated Hospital of Shaanxi University of traditional Chinese Medicine, Xianyang, Shaanxi, China
| | - Wen-Jia Luo
- Teaching and Research Office of Chinese Pharmacy, The College of Pharmacy of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Zheng Fan
- Department of Gastroenterology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang712000, Shaanxi, China
| |
Collapse
|
9
|
Bhattacharya S. The Incredible Potential of Exosomes as Biomarkers in the Diagnosis of Colorectal Cancer. Curr Drug Res Rev 2022; 14:188-202. [PMID: 35490434 DOI: 10.2174/2665998002666220501164429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/18/2021] [Accepted: 01/21/2022] [Indexed: 06/14/2023]
Abstract
Colorectal cancer (CRC) is common cancer that is one of the leading causes of cancerrelated deaths around the world. The creation of new biomarkers for this disease is an important public health strategy for lowering the disease's mortality rate. According to new research, exosomes may be important sources of biomarkers in CRC. Exosomes are nanometer-sized membrane vesicles (30-200 nm) secreted by normal and cancer cells that transport RNA and proteins between cells and are thought to help with intercellular communication. Exosomes have been linked to CRC initiation and progression, and some differentially expressed RNAs and proteins in exosomes have been identified as potential cancer detection candidates. As a result, studying the relationship between exosomes and CRC may aid in the development of new biomarkers for the disease. This article discusses the importance of exosomes as biomarkers in the diagnosis of CRC, as well as their use in the treatment of CRC metastasis, chemoresistance, and recrudescence. The benefits and drawbacks of using exosomes as tumour markers are also discussed.
Collapse
Affiliation(s)
- Sankha Bhattacharya
- Department of Pharmaceutics, NMIM'S School of Pharmacy & Technology Management, Deemed-to-be University, Shirpur, Maharashtra 425405, India
| |
Collapse
|
10
|
D-Limonene inhibits the occurrence and progression of LUAD through suppressing lipid droplet accumulation induced by PM 2.5 exposure in vivo and in vitro. Respir Res 2022; 23:338. [PMID: 36496421 PMCID: PMC9741803 DOI: 10.1186/s12931-022-02270-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND PM2.5 exposure is associated with lung adenocarcinoma (LUAD), but the mechanism is unclear. The lack of understanding impedes our effort on prevention. This study examined a possible mechanism of lung cancer caused by PM2.5 exposure, and aimed to find a potential intervention for people living in PM2.5 polluted regions. METHODS Electron microscopy and oil-red staining were conducted to examine the lipid droplet accumulation. Masson's trichrome staining, colony forming, scratch assay and transwell experiment were conducted to evaluate the effect of PM2.5 exposure and D-limonene intervention on the occurrence and progression of LUAD. Potential intervention targets were found by RNA-Seq and verified by luciferase reporter assay. MiR-195 KO mice constructed with CRISPR/Cas9 technology were used to investigate the pivotal role of D-limonene-miR-195-SREBP1/FASN axis. Cohort analysis of lung cancer patients, human LUAD tissues staining and human intervention trial were also conducted to validate the results of cell and animal experiments. RESULTS Our results showed that PM2.5 exposure induced accumulation of lipid droplets in LUAD cells which accompanied by increased malignant cellular behaviors. PM2.5 exposure led to cleaved N-SREBP1 translocation into nucleus, which activated the de novo lipogenesis pathway. Same changes were also observed in normal lung epithelial cells and normal lung tissue, and mice developed pulmonary fibrosis after long-term exposure to PM2.5. Furthermore, in a cohort of 11,712 lung cancer patients, significant lipid metabolism disorders were observed in higher PM2.5 polluted areas. In view of that, D-limonene was found to inhibit the changes in lipid metabolism through upregulating the expression of miR-195, which inhibited the expression of lipogenic genes (SREBF1/FASN/ACACA) specifically. And a small human intervention trial showed that serum miR-195 was upregulated after oral intake of D-limonene. CONCLUSION Our findings reveal a new mechanism of pulmonary fibrosis and LUAD that is related to PM2.5 exposure-induced lipid droplet accumulation. We also demonstrate that D-limonene-miR-195-SREBP1/FASN axis is a potential preventive intervention for mediating the progression and development of LUAD induced by PM2.5 exposure. Trial registration Chinese Clinical Trial Registry, ChiCTR2000030200. Registered 25 February 2020, http://www.chictr.org.cn/showproj.aspx?proj=48013.
Collapse
|
11
|
Liu YP, Qiu ZZ, Li XH, Li EY. Propofol induces ferroptosis and inhibits malignant phenotypes of gastric cancer cells by regulating miR-125b-5p/STAT3 axis. World J Gastrointest Oncol 2021; 13:2114-2128. [PMID: 35070046 PMCID: PMC8713308 DOI: 10.4251/wjgo.v13.i12.2114] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/10/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric cancer is a common malignancy with poor prognosis, in which ferroptosis plays a crucial function in its development. Propofol is a widely used anesthetic and has antitumor potential in gastric cancer. However, the effect of propofol on ferroptosis during gastric cancer progression remains unreported.
AIM To explore the function of propofol in the regulation of ferroptosis and malignant phenotypes of gastric cancer cells.
METHODS MTT assays, colony formation assays, Transwell assays, wound healing assay, analysis of apoptosis, ferroptosis measurement, luciferase reporter gene assay, and quantitative reverse transcription polymerase chain reaction were used in this study.
RESULTS Our data showed that propofol was able to inhibit proliferation and induce apoptosis of gastric cancer cells. Meanwhile, propofol markedly repressed the invasion and migration of gastric cancer cells. Importantly, propofol enhanced the erastin-induced inhibition of growth of gastric cancer cells. Consistently, propofol increased the levels of reactive oxygen species, iron, and Fe2+ in gastric cancer cells. Moreover, propofol suppressed signal transducer and activator of transcription (STAT)3 expression by upregulating miR-125b-5p and propofol induced ferroptosis by targeting STAT3 in gastric cancer cells. The miR-125b-5p inhibitor or STAT3 overexpression reversed propofol-attenuated malignant phenotypes of gastric cancer cells.
CONCLUSION Propofol induced ferroptosis and inhibited malignant phenotypes of gastric cancer cells by regulating the miR-125b-5p/STAT3 axis. Propofol may serve as a potential therapeutic candidate for gastric cancer.
Collapse
Affiliation(s)
- Yi-Ping Liu
- Department ofAnesthesiology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Zhong-Zhi Qiu
- Department ofAnesthesiology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Xu-Hui Li
- Department of Gastroenterology, Heilongjiang Forest Industry Federation (Red Cross) Hospital, Harbin 150008, Heilongjiang Province, China
| | - En-You Li
- Department ofAnesthesiology, First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| |
Collapse
|
12
|
Ye LL, Cheng ZG, Cheng XE, Huang YL. Propofol regulates miR-1-3p/IGF1 axis to inhibit the proliferation and accelerates apoptosis of colorectal cancer cells. Toxicol Res (Camb) 2021; 10:696-705. [PMID: 34745557 DOI: 10.1093/toxres/tfab047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 04/06/2021] [Accepted: 04/29/2021] [Indexed: 12/22/2022] Open
Abstract
This study aimed to clarify the mechanism of propofol on proliferation and apoptosis of colorectal cancer (CRC) cell. SW620 and HCT15 cells were exposed to different concentrations of propofol, the proliferation and apoptotic rate, were measured by MTT, colony formation and flow cytometry assays, respectively. The expressions of miR-1-3p and insulin-like growth factors 1 (IGF1) were examined by real-time polymerase chain reaction (RT-qPCR). Western bolt was employed to quantify the protein levels of IGF1 and apoptotic proteins. The molecular interaction between miR-1-3p and IGF1 was validated using dual-luciferase reporter assay. A xenograft tumor model was established to further assess the effects of propofol on CRC in vivo. Propofol dramatically decreased the proliferation and elevated apoptotic rate of CRC cells. RT-qPCR assay demonstrated that miR-1-3p was downregulated in CRC cells, and could be strikingly increased by propofol. Importantly, miR-1-3p inhibited IGF-1 expression through interacting with its 3'-UTR region, thus inactivating AKT/mTOR signals. Gain or loss of functional study revealed that miR-1-3p downregulation remarkedly diminished the anti-tumor roles of propofol by directly inhibiting IGF1. In vivo study showed that propofol inhibited tumor growth by regulating miR-1-3p/IGF1 axis. Our data eventually elucidated that propofol suppressed CRC progression by promoting miR-1-3p which targeted IGF1. These results might provide a scientific basis for the application of propofol on the clinical surgery and the prognosis of patients with CRC.
Collapse
Affiliation(s)
- Ling-Ling Ye
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zhong-Gui Cheng
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xiao-E Cheng
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yuan-Lu Huang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
13
|
Targeting miRNAs with anesthetics in cancer: Current understanding and future perspectives. Biomed Pharmacother 2021; 144:112309. [PMID: 34653761 DOI: 10.1016/j.biopha.2021.112309] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023] Open
Abstract
Anesthetics are extensively used during cancer surgeries. The progression of cancer can be influenced by perioperative events such as exposure to general or local anesthesia. However, whether they inhibit cancer or act as a causative factor for metastasis and exert deleterious effects on cancer growth differs based on the type of cancer and the therapy administration. Recent experimental data suggested that many of the most commonly used anesthetics in surgical oncology, whether general or local agents, can alter gene expression and cause epigenetic changes via modulating miRNAs. miRNAs are single-stranded non-coding RNAs that regulate gene expression at various levels, and their dysregulation contributes to the pathogenesis of cancers. However, anesthetics via regulating miRNAs can concurrently target several effectors of cellular signaling pathways involved in cell differentiation, proliferation, and viability. This review summarized the current research about the effects of different anesthetics in regulating cancer, with a particular emphasis on the role of miRNAs. A significant number of studies conducted in this area of research illuminate the effects of anesthetics on the regulation of miRNA expression; therefore, we hope that a thorough understanding of the underlying mechanisms involved in the regulation of miRNA in the context of anesthesia-induced cancer regulation could help to define optimal anesthetic regimens and provide better perspectives for further studies.
Collapse
|
14
|
Ota Y, Takahashi K, Otake S, Tamaki Y, Okada M, Yan I, Aso K, Fujii S, Patel T, Haneda M. Extracellular RNA transfer from non-malignant human cholangiocytes can promote cholangiocarcinoma growth. FEBS Open Bio 2021; 11:3276-3292. [PMID: 34510808 PMCID: PMC8634862 DOI: 10.1002/2211-5463.13294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 08/07/2021] [Accepted: 09/09/2021] [Indexed: 11/12/2022] Open
Abstract
Extracellular vesicles (EV) within the cellular secretome are emerging as modulators of pathological processes involved in tumor growth through their ability to transfer donor‐derived RNA into recipient cells. While the effects of tumor and stromal cell EVs within the tumor microenvironment have been studied, less is known about the contributions of normal, nontransformed cells. We examined the impact of EVs within the cellular secretome from nonmalignant cells on transformed cell growth and behavior in cholangiocarcinoma cells. These effects were enhanced in the presence of the pro‐fibrogenic mediator TGF‐β. We identified miR‐195 as a TGF‐β responsive miRNA in normal cells that can be transferred via EV to tumor cells and regulate cell growth, invasion, and migration. The effects of miR‐195 involve modulation of the epithelial–mesenchymal transition through direct effects on the transcription factor Snail. These studies provide in vitro and in vivo evidence for the impact of normal cellular secretome on transformed cell growth, show the importance of EV RNA transfer, and identify mechanisms of EV‐mediated transfer of miRNA as a contributor to tumor development, which may provide new therapeutic opportunities for targeting human cholangiocarcinoma.
Collapse
Affiliation(s)
- Yu Ota
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Japan
| | - Kenji Takahashi
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Japan
| | - Shin Otake
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Japan
| | - Yosui Tamaki
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Japan
| | - Mitsuyoshi Okada
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Japan
| | - Irene Yan
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Kazunobu Aso
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Japan
| | - Satoshi Fujii
- Department of Laboratory Medicine, Asahikawa Medical University, Japan
| | - Tushar Patel
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Masakazu Haneda
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Japan
| |
Collapse
|
15
|
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 PMCID: PMC8408744 DOI: 10.21873/invivo.12532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [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
|
16
|
Liu L, Dong T, Sheng J. Propofol Suppresses Gastric Cancer Progression by Regulating circPDSS1/miR-1324/SOX4 Axis. Cancer Manag Res 2021; 13:6031-6043. [PMID: 34377022 PMCID: PMC8349207 DOI: 10.2147/cmar.s312989] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
Background Propofol is a common intravenous anesthetic that exerts an antitumor role in human cancers. Circular RNAs (circRNAs) play crucial roles in the progression of various cancers. However, the relationship between propofol and circRNA decaprenyl diphosphate synthase subunit 1 (circPDSS1) in gastric cancer (GC) remains unclear. Methods Cell proliferation was evaluated by Cell Counting Kit-8 (CCK-8), colony formation, and 5-ethynyl-2ʹ-deoxyuridine (EdU) assays. Cell migration and invasion were assessed by transwell assay. Cell apoptosis was determined by flow cytometry. All protein levels were detected by Western blot assay. The expression levels of circPDSS1, microRNA-1324 (miR-1324), and SRY-box transcription factor 4 (SOX4) mRNA were determined by quantitative real-time PCR (qRT-PCR). The interaction between miR-1324 and circPDSS1 or SOX4 was confirmed by dual-luciferase reporter and RNA pull-down assays. The mice xenograft model was established to investigate the role of propofol and circPDSS1 in vivo. Results Propofol inhibited cell proliferation, migration and invasion and induced apoptosis in GC cells, which could be reversed by upregulating circPDSS1. MiR-1324 was a target of circPDSS1, and circPDSS1 promoted cell proliferation, migration and invasion and reduced apoptosis in propofol-treated cells by sponging miR-1324. Moreover, SOX4 was a direct target of miR-1324, and miR-1324 exerted anticancer role by targeting SOX4 in propofol-treated cells. CircPDSS1 acted as a sponge of miR-1324 to regulate SOX4 expression. Additionally, circPDSS1 overexpression weakened the anticancer role of propofol in vivo. Conclusion Propofol exerted anticancer role in GC through regulating circPDSS1/miR-1324/SOX4 axis, indicating that propofol might be an effective therapeutic medicine for GC treatment.
Collapse
Affiliation(s)
- Leyi Liu
- Department of Anesthesiology, Wuhu Hospital of Traditional Chinese Medicine, Wuhu, 241000, People's Republic of China
| | - Ting Dong
- Yijishan Hospital, First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, People's Republic of China
| | - Jun Sheng
- Department of Oncology, Wuhu Hospital of Traditional Chinese Medicine, Wuhu, 241000, People's Republic of China
| |
Collapse
|
17
|
Edgunlu TG, Avci CB, Ozates NP, Bagca BG, Celik SK, Boluk A, Ugur B. In Vitro Effects of Propofol on Cytotoxic, Apoptotic and PI3K-Akt Signaling Pathway Genes on Brain Cancer Cells. Anticancer Agents Med Chem 2021; 22:356-361. [PMID: 34238171 DOI: 10.2174/1871520621666210708094328] [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: 10/17/2020] [Revised: 05/24/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
AIM It was aimed to determine the cytotoxic and apoptotic effect of propofol on glioma cells. BACKGROUND Propofol [2,6-diisopropylphenol] is a commonly used intravenous anesthetic. Propofol is known to have a mechanism of action on the PI3K-AKT pathway. OBJECTIVE This study aimed to evaluate the effect of propofol on the proliferation and apoptosis of human glioma cells, as well as to investigate changes in expression levels of the PI3K-AKT signaling pathway genes. MATERIALS-METHODS The cytotoxic effect of propofol on the U-87 MG cell line was determined by WST-1 method. Annexin V-FITC and Mitoprobe JC-1 assay were used to measure apoptosis by flow cytometry. Expression levels of genes in the PI3K-AKT signaling pathway were investigated by qRT-PCR. RESULTS We have shown that propofol-induced apoptosis in U-87 MG cells by 17.1-fold compared to untreated control. Furthermore, significant differences were found in the expression levels of the PI3K-AKT signaling pathway genes. CONCLUSION As a result of our study, it was found that propofol caused differences in expression levels of PI3K-AKT signaling pathway genes, and it was suggested that these differences might be related to apoptosis induction.
Collapse
Affiliation(s)
- Tuba Gokdogan Edgunlu
- Muğla Sıtkı Koçman University Faculty of Medicine Department of Medical Biology, Turkey
| | - Cigir Biray Avci
- Ege University Faculty of Medicine Department of Medical Biology, Turkey
| | | | - Bakiye Goker Bagca
- Ege University Faculty of Medicine Department of Medical Biology, Turkey
| | - Sevim Karakas Celik
- Bülent Ecevit University Faculty of Medicine Department of Medical Genetic, Turkey
| | - Aydin Boluk
- Muğla Sıtkı Koçman University Faculty of Medicine, Turkey
| | - Bakiye Ugur
- Muğla Sıtkı Koçman University Faculty of Medicine Department of Anesthesiology and Reanimation, Turkey
| |
Collapse
|
18
|
Ji X, Sun W, Lv C, Huang J, Zhang H. Circular RNAs Regulate Glucose Metabolism in Cancer Cells. Onco Targets Ther 2021; 14:4005-4021. [PMID: 34239306 PMCID: PMC8259938 DOI: 10.2147/ott.s316597] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022] Open
Abstract
Circular RNAs (circRNAs) were originally thought to result from RNA splicing errors. However, it has been shown that circRNAs can regulate cancer onset and progression in various ways. They can regulate cancer cell proliferation, differentiation, invasion, and metastasis. Moreover, they modulate glucose metabolism in cancer cells through different mechanisms such as directly regulating glycolytic enzymes and glucose transporter (GLUT) or indirectly regulating signal transduction pathways. In this review, we elucidate on the role of circRNAs in regulating glucose metabolism in cancer cells, which partly explains the pathogenesis of malignant tumors, and provides new therapeutic targets or new diagnostic and prognostic markers for human cancers.
Collapse
Affiliation(s)
- Xiaoyu Ji
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, People's Republic of China
| | - Wei Sun
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, People's Republic of China
| | - Chengzhou Lv
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, People's Republic of China
| | - Jiapeng Huang
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, People's Republic of China
| | - Hao Zhang
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, People's Republic of China
| |
Collapse
|
19
|
Tan SH, Ding HJ, Mei XP, Liu JT, Tang YX, Li Y. Propofol suppressed cell proliferation and enhanced apoptosis of bladder cancer cells by regulating the miR-340/CDK2 signal axis. Acta Histochem 2021; 123:151728. [PMID: 34048990 DOI: 10.1016/j.acthis.2021.151728] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND As widely reported, propofol can effectively inhibit tumors development. However, little is known about the molecular mechanisms. Here, we proved that propofol regulated miR-340/CDK2 axis to suppress bladder cancer progression in vitro. METHODS MicroRNA (MiR)-340 expression in 5637 cells was examined using qRT-PCR. Cyclin-dependent kinase2 (CDK2) expression was detected using both qRT-PCR and western blot. The levels of apoptosis-related proteins and cell cycle-related proteins were evaluated using western blot. CCK-8 assay and BrdU assay were conducted to evaluate cell proliferation. Moreover, flow cytometry assay was employed to assess cell cycle and cell apoptosis. Finally, dual luciferase reporter assay was employed to verify the binding relationship between miR-340 and CDK2. RESULTS Here we showed that propofol treatment inhibited cell proliferation of 5637 cells but enhanced cell apoptosis. Propofol upregulated miR-340 in a dose and time dependent manner. MiR-340 inhibitor could reverse the effect of propofol on the proliferation and apoptosis of 5637 cells. Next, dual luciferase reporter assay displayed that miR-340 directly bound to the 3'-UTR of CDK2. Finally, inhibition of CDK2 could partly reversed the effect of miR-340 inhibitor on cell proliferation and cell apoptosis of propofol-treated 5637 cells. CONCLUSION In total, our results proved that targeting miR340/CDK2 axis was novel to enhance the anti-tumor effects of propofol in bladder cancer in vitro, and our study provided alternative therapeutic strategies for clinical treatment of bladder cancer.
Collapse
|
20
|
Du Y, Zhang X, Zhang H, Chen Y, Zhu S, Shu J, Pan H. Propofol modulates the proliferation, invasion and migration of bladder cancer cells through the miR‑145‑5p/TOP2A axis. Mol Med Rep 2021; 23:439. [PMID: 33846791 PMCID: PMC8060790 DOI: 10.3892/mmr.2021.12078] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 01/28/2021] [Indexed: 12/24/2022] Open
Abstract
Propofol‑based anesthesia has been reported to reduce the recurrence and metastasis of a number of cancer types following surgical resection. However, the effects of propofol in bladder cancer (BC) are yet to be fully elucidated. The aim of the present study was to investigate the functions of propofol in BC and their underlying mechanisms. In the study, the expression of microRNA (miR)‑145‑5p in BC tissues and cell lines was evaluated using reverse transcription‑quantitative PCR, and the effects of propofol on BC cells were determined using cell viability, wound healing and Transwell cell invasion assays, bioinformatics analysis, western blotting, immunohistochemistry and in vivo tumor xenograft models. It was found that propofol significantly suppressed the proliferation, migration and invasion of BC cells in vitro. In addition, propofol induced miR‑145‑5p expression in a time‑dependent manner, and miR‑145‑5p knockdown attenuated the inhibitory effects of propofol on the proliferation, migration and invasion of BC cells. Topoisomerase II α (TOP2A) was a direct target of miR‑145‑5p, and silencing TOP2A reversed the effects of miR‑145‑5p knockdown in propofol‑treated cells. Furthermore, propofol suppressed tumor xenograft growth, which was partially attenuated by miR‑145‑5p knockdown. The present study provided novel insight into the advantages of surgical intervention with propofol anesthesia in patients with BC.
Collapse
Affiliation(s)
- Yi Du
- Department of Anesthesiology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
| | - Xudong Zhang
- Department of Anesthesiology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
| | - Hongwei Zhang
- Department of Anesthesiology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
| | - Yiding Chen
- Department of Anesthesiology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
| | - Shuying Zhu
- Department of Anesthesiology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
| | - Jinjun Shu
- Department of Anesthesiology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
| | - Hui Pan
- Department of Anesthesiology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
21
|
Chen R, Yang M, Huang W, Wang B. Cascades between miRNAs, lncRNAs and the NF-κB signaling pathway in gastric cancer (Review). Exp Ther Med 2021; 22:769. [PMID: 34055068 PMCID: PMC8145527 DOI: 10.3892/etm.2021.10201] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 04/28/2021] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer is a common digestive tract malignancy that is mainly treated with surgery combined with perioperative adjuvant chemoradiotherapy and biological targeted therapy. However, the diagnosis rate of early gastric cancer is low and both postoperative recurrence and distant metastasis are thorny problems. Therefore, it is essential to study the pathogenesis of gastric cancer and search for more effective means of treatment. The nuclear factor-κB (NF-κB) signaling pathway has an important role in the occurrence and development of gastric cancer and recent studies have revealed that microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are able to regulate this pathway through a variety of mechanisms. Understanding these interrelated molecular mechanisms is helpful in guiding improvements in gastric cancer treatment. In the present review, the functional associations between miRNAs, lncRNAs and the NF-κB signaling pathway in the occurrence, development and prognosis of gastric cancer were discussed. It was concluded that miRNAs and lncRNAs have complex relations with the NF-κB signaling pathway in gastric cancer. miRNAs/target genes/NF-κB/target proteins, signaling molecules/NF-κB/miRNAs/target genes, lncRNAs/miRNAs/NF-κB/genes or mRNAs, lncRNAs/target genes/NF-Κb/target proteins, and lncRNAs/NF-κB/target proteins cascades are all important factors in the occurrence and development of gastric cancer.
Collapse
Affiliation(s)
- Risheng Chen
- Department of Anesthesiology, Affiliated Nanhua Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Mingxiu Yang
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology (2016TP1015), Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Weiguo Huang
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology (2016TP1015), Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Baiyun Wang
- Department of Anesthesiology, Affiliated Nanhua Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
22
|
Wang H, Sui ZL, Wu XX, Tang P, Zhang HD, Yu ZT. Reversal of Chemotherapy Resistance to Cisplatin in NSCLC by miRNA-195-5p via Targeting the FGF2 Gene. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2021; 14:497-508. [PMID: 33953601 PMCID: PMC8092352 DOI: 10.2147/pgpm.s302755] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/26/2021] [Indexed: 12/20/2022]
Abstract
Objective To explore the mechanism of miR-195-5p in the pathogenesis non-small cell lung cancer (NSCLC) and cisplatin resistance. Methods The function of miR-195-5p in NSCLC and cisplatin resistance were determined by MTT, scratch assay, transwell assay, and nude mice xenograft experiments. miR-195-5p target gene was identified by dual-luciferase reporter assays and real-time PCR analysis. Results miR-195-5p content was lower in A549/DDP than that in A549 cells, with reduced chemotherapy sensitivity and increased cell invasion and migration ability. The loss-of-function and gain-of-function assays illustrated that miR-195-5p might have increased the chemosensitivity to cisplatin in the A549/DDP cells and decreased cell migration and invasion. FGF2 is a negatively correlated action target of miR-195-5p. miR-195-5p might affect EMT by inhibiting FGF2. Overexpression of FGF2 resulted in enhanced cisplatin resistance in the cells, while miR-195-5p might have reversed this resistance. Conclusion Overall, miR-195-5p might target FGF2 to reduce cisplatin resistance in A549/DDP cells and enhance chemosensitivity.
Collapse
Affiliation(s)
- Hao Wang
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin City, Tianjin, 300060, People's Republic of China.,Department of Surgical Oncology, Baotou Cancer Hospital, Baotou, People's Republic of China
| | - Zhi-Lin Sui
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin City, Tianjin, 300060, People's Republic of China
| | - Xian-Xian Wu
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin City, Tianjin, 300060, People's Republic of China
| | - Peng Tang
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin City, Tianjin, 300060, People's Republic of China
| | - Hong-Dian Zhang
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin City, Tianjin, 300060, People's Republic of China
| | - Zhen-Tao Yu
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin City, Tianjin, 300060, People's Republic of China.,Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, People's Republic of China
| |
Collapse
|
23
|
Cao C, Zhang X, Xu Y. Propofol prevents the aggressive progression of oral squamous cell carcinoma via regulating circ_0005623/miR-195-5p/HOXB7 axis. Biotechnol Appl Biochem 2021; 69:1015-1028. [PMID: 33894003 DOI: 10.1002/bab.2172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/01/2021] [Indexed: 12/24/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is a general oral disease with high mortality. This study aimed to investigate the effects and underlying mechanism of propofol in OSCC. Propofol treatment inhibited cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT), but promoted apoptosis and induced cell cycle arrest in OSCC cells. miR-195-5p was a target of circ_0005623 and directly targeted to HOXB7. Circ_0005623 and HOXB7 were upregulated, while miR-195-5p was downregulated in OSCC tissues and cells. Overexpression of circ_0005623 partly reversed the effects of propofol on cell proliferation, migration invasion, EMT, cell cycle progression, and apoptosis in SCC-9 and CAL-27 cells. Meanwhile, further investigation uncovered that circ_0005623 could act as a sponge for miR-195-5p to regulate HOXB7 expression, thereby mediating the suppression effects of propofol on OSCC cells. In vivo assay suggested that overexpression of circ_0005623 promoted tumor growth, which was inhibited by propofol treatment. Taken together, propofol regulated aggressive progression of OSCC via the circ_0005623/miR-195-5p/HOXB7 axis, providing the new train of thoughts for diagnosis and therapy of human OSCC.
Collapse
Affiliation(s)
- Chen Cao
- Department of Anesthesiology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Xiaotong Zhang
- Department of Anesthesiology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Yi Xu
- Department of Stomatology, Zibo First Hospital, Zibo, Shandong, China
| |
Collapse
|
24
|
Wang H, Xu Y, Jin M, Li H, Li S. miR-383 reduces keratinocyte proliferation and induces the apoptosis in psoriasis via disruption of LCN2-dependent JAK/STAT pathway activation. Int Immunopharmacol 2021; 96:107587. [PMID: 33819732 DOI: 10.1016/j.intimp.2021.107587] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 11/25/2022]
Abstract
Psoriasis is a chronic and relapsing disorder with considerable negative effects on patients' quality of life. The finer details associated with the molecular mechanism of psoriasis and its pathogenesis remain somewhat elusive. Extensive studies have highlighted the crucial role of microRNAs (miRNAs) in the development of psoriasis. Hence, the current study aimed to investigate the effect of miR-383 on a psoriasis rat model and elucidate the underlying molecular mechanism. The rat psoriasis model was established via imiquimod (IMQ) induction followed by verification of miR-383 and LCN2 expression in the skin tissues of the models. ELISA was conducted to determine the secretion of inflammatory factors. Keratinocyte proliferation and apoptosis was evaluated by MTT assay and flow cytometric analysis. Down-regulation of miR-383 and up-regulation of LCN2 were detected in the psoriasis rat model. Our data indicated that miR-383 targeted LCN2 by binding to its 3'UTR and inhibited JAK/STAT pathway activation. Notably, miR-383 overexpression or LCN2 knockdown attenuated psoriasis-like symptoms, suppressed inflammatory response, reduced the expression of JAK3 and STAT3, ceased keratinocyte proliferation, and promoted the apoptosis. The findings of our study suggest that miR-383 may inhibit LCN2 and inactivate the JAK/STAT pathway, suppressing the progression of psoriasis in a rat model. This study provided novel insights into the pathogenesis of psoriasis and offered potential targets for psoriasis treatment.
Collapse
Affiliation(s)
- Hong Wang
- Department of Dermatology, The First Hospital of Jilin University, Changchun 130021, Jilin Province, PR China.
| | - Yangchun Xu
- Department of Dermatology, The Second Hospital of Jilin University, Changchun 130041, Jilin Province, PR China
| | - Meishan Jin
- Department of Pathology, The First Hospital of Jilin University, Changchun 130021, Jilin Province, PR China
| | - Hongxia Li
- Department of Dermatology, The First Hospital of Jilin University, Changchun 130021, Jilin Province, PR China
| | - Shanshan Li
- Department of Dermatology, The First Hospital of Jilin University, Changchun 130021, Jilin Province, PR China.
| |
Collapse
|
25
|
Bai ZM, Li XF, Yang Y, Yang YF, Lv DR, Tang LL. Propofol inhibited gastric cancer proliferation via the hsa-miR-328-3p/STAT3 pathway. Clin Transl Oncol 2021; 23:1866-1873. [PMID: 33772723 DOI: 10.1007/s12094-021-02595-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/12/2021] [Indexed: 11/25/2022]
Abstract
PURPOSE The aim of the present study was to elucidate the functional role of hsa-miR-328-3p/STAT3 pathway in the effects of propofol on gastric cancer proliferation. METHODS Bioinformatics was used to analyze the molecular expression differences of hsa-miR-328-3p/STAT3 axis in stomach adenocarcinoma (n = 435) and normal samples (n = 41) from TCGA database. The expression of the above molecules in gastric cancer cells SGC-7901 and normal gastric mucosal cells GES-1 was verified via qPCR. The dual-luciferase assay was carried out to confirm the interaction between hsa-miR-328-3p and STAT3. Subsequently, the cell proliferation and the expression of the above molecules in SGC-7901 and GES-1 cells were evaluated after 10 μM propofol treatment. Finally, we analyzed whether propofol still inhibited the proliferation of gastric cancer by suppressing STAT3 pathway after hsa-miR-328-3p down-regulation. RESULTS Compared with normal samples, the expression of hsa-miR-328-3p was significantly down-regulated in stomach adenocarcinoma samples, while the expression of STAT3 and downstream target genes (MMP2, CCND1 and COX2) was up-regulated. The results were consistent with those in GES-1 and SGC-7901 cell lines. Meanwhile, we found that hsa-miR-328-3p can bind to the 3'-UTR of the potential target gene STAT3. Furthermore, propofol significantly inhibited the proliferation of gastric cancer cell line SGC-7901, where hsa-miR-328-3p was up-regulated and the expression of STAT3 and downstream proliferation-related target genes were down-regulated. However, the growth inhibition of propofol on SGC-7901 cell was significantly reversed after the inhibition of hsa-miR-328-3p. CONCLUSIONS To sum up, propofol suppressed the STAT3 pathway via up-regulating hsa-miR-328-3p to inhibit gastric cancer proliferation.
Collapse
Affiliation(s)
- Z M Bai
- Department of Anesthesiology, Wuwei People's Hospital, North Side of Xuanwu Street, Liangzhou District, Wuwei, 733000, China
| | - X F Li
- Department of Neonatology, Wuwei People's Hospital, Wuwei, 733000, China
| | - Y Yang
- Department of Chinese Medicine, Rheumatology and Immunology, Wuwei Liangzhou Hospital, Wuwei, 733000, China
| | - Y F Yang
- Department of Neurocardiology, Wuwei Second People's Hospital, Wuwei, 733000, China
| | - D R Lv
- Department of Anesthesiology, Wuwei People's Hospital, North Side of Xuanwu Street, Liangzhou District, Wuwei, 733000, China
| | - L L Tang
- Department of Anesthesiology, Wuwei People's Hospital, North Side of Xuanwu Street, Liangzhou District, Wuwei, 733000, China.
| |
Collapse
|
26
|
Chang CY, Wu MY, Chien YJ, Su IM, Wang SC, Kao MC. Anesthesia and Long-term Oncological Outcomes: A Systematic Review and Meta-analysis. Anesth Analg 2021; 132:623-634. [PMID: 33105278 DOI: 10.1213/ane.0000000000005237] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Whether propofol elicits a survival benefit over volatile anesthetics during cancer surgery remains inconclusive. The primary aim of this systematic review and meta-analysis is to compare the effects of propofol-based total intravenous anesthesia (TIVA) with any volatile anesthesia on long-term oncological outcomes. The secondary aim is to compare propofol-based TIVA with specific volatile agents on long-term oncological outcomes. METHODS We searched PubMed, Embase, Scopus, Web of Science, and Cochrane Library from inception through March 3, 2020. Randomized control trials and observational studies that compared the effects of propofol-based TIVA and volatile anesthesia on long-term oncological outcomes, which also reported hazard ratios (HR) as effect estimates, were considered eligible for inclusion. Using the inverse variance method with a random-effects model, HR and 95% confidence intervals (CI) were calculated. Trial sequential analysis was incorporated to test if the results were subject to a type I or type II error. RESULTS Nineteen retrospective observational studies were included. Patients who received propofol-based TIVA during cancer surgery were associated with significantly better overall survival than those who received volatile anesthesia (HR = 0.79, 95% CI, 0.66-0.94, P = .008, I2 = 82%). In contrast, no statistically significant difference was observed in recurrence-free survival between patients who received propofol-based TIVA and volatile anesthesia during cancer surgery (HR = 0.81, 95% CI, 0.61-1.07, P = .137, I2 = 85%). In the subgroup analysis by different volatile anesthetics, patients who received propofol-based TIVA were associated with better overall survival than those who received desflurane (HR = 0.54, 95% CI, 0.36-0.80, P = .003, I2 = 80%). In contrast, there was no statistically significant difference in overall survival between patients who received propofol-based TIVA and those who received sevoflurane (HR = 0.92, 95% CI, 0.74-1.14, P = .439, I2 = 70%). In the trial sequential analysis of overall survival, the cumulative Z curve reached the required heterogeneity-adjusted information size and crossed the traditional significance boundary. In contrast, in the trial sequential analysis of recurrence-free survival, the cumulative Z curve did not cross the traditional significance boundary. However, the required heterogeneity-adjusted information size has not yet been reached. CONCLUSIONS Propofol-based TIVA is generally associated with better overall survival than volatile anesthesia during cancer surgery. Further large-scaled, high-quality randomized control trials are warranted to confirm our findings.
Collapse
Affiliation(s)
- Chun-Yu Chang
- From the Department of Anesthesiology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Meng-Yu Wu
- School of Medicine, Tzu Chi University, Hualien, Taiwan.,Department of Emergency Medicine
| | - Yung-Jiun Chien
- School of Medicine, Tzu Chi University, Hualien, Taiwan.,Department of Physical Medicine and Rehabilitation, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - I-Min Su
- School of Medicine, Tzu Chi University, Hualien, Taiwan.,Department of Anesthesiology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Shih-Ching Wang
- From the Department of Anesthesiology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Ming-Chang Kao
- From the Department of Anesthesiology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
27
|
Chen C, Ai QD, Wei YH. Kanglaite enhances the efficacy of cisplatin in suppression of hepatocellular carcinoma via inhibiting CKLF1 mediated NF-κB pathway and regulating transporter mediated drug efflux. JOURNAL OF ETHNOPHARMACOLOGY 2021; 264:113388. [PMID: 32918990 DOI: 10.1016/j.jep.2020.113388] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/30/2020] [Accepted: 09/07/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Kanglaite (KLT) is an active extract of the Coix lacryma-jobi seed, which can benefit Qi and nourish Yin, and disperse the accumulation of evils. It is used as a biphasic broad-spectrum anti-cancer drug, and shows synergistic effects with radiotherapy and chemotherapy. However, the mechanism of KLT combined with cisplatin (CDDP) against hepatocellular carcinoma (HCC) has not been elucidated. AIM OF THE STUDY The aim of present study was to investigate the potential synergistic effects of KLT and CDDP on HepG2 cells, discussing the possible mechanisms from the perspective of CKLF1 and NF-κB mediated inflammatory response and chemoresistance, and the involvement of drug efflux transporters. MATERIALS AND METHODS CDDP injured HepG2 cells were used to investigate the effects of KLT on chemotherapeutics treated HCC. Effects of KLT pretreatment on CDDP injured HepG2 cells were determined by MTT, wound healing assay, and transwell assay. Expression of chemokine-like factor 1 (CKLF1) and activation of nuclear factor κB (NF-κB) were examined by qPCR, western blot, and immunofluorescence staining. Furthermore, to study the role of CKLF1 in KLT mediated effects on this CDDP injured HCC cell model, HepG2 cells overexpressed with CKLF1 gene were used. Cell viability and NF-κB activation were investigated. Moreover, TNF-α and IL-1β levels were measured by Elisa analysis and western blot to evaluate the inflammatory response. Additionally, ATP-binding cassette (ABC) drug efflux transporters, MDR1, MRP2, and BCRP were also determined in present study. RESULTS KLT pretreatment followed by CDDP treatment was found to show synergistic effects, which showed by decreased cell viability, migration and invasion ability of HepG2 cells. Expression of CKLF1 enhanced significantly in CDDP treated HepG2 cells, and KLT decreased this elevation obviously. Furthermore, CDDP activated NF-κΒ and promoted translocation of NF-κB toward the nucleus. KLT inhibited the activation of NF-κΒ, which sensitized cancer cells. Overexpression of CKLF1 reversed the effects of KLT on CDDP injured HepG2 cells, which exhibited by increased cell viability and enhanced activation of NF-κΒ. CDDP induced NF-κΒ activation could also lead to excessive inflammatory response, and KLT can suppress the aggravating inflammation which may be beneficial for tumor progression. Furthermore, we found that ABC drug efflux transporters MDR1, MRP2, and BCRP in CDDP treated HepG2 cells were decreased when pretreated with KLT. CONCLUSIONS KLT pretreatment may increase the effects of CDDP on HepG2 cells, by exhibiting cooperative effects on suppression of HepG2 cells. The mechanisms may partly by inhibiting CKLF1 mediated NF-κB pathway, which may contribute to inflammation of tumor microenvironment and chemoresistance of CDDP. Inhibition of transporter-mediated drug efflux is also involved in KLT mediated sensitization effects of CDDP.
Collapse
Affiliation(s)
- Chen Chen
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| | - Qi-di Ai
- Hunan University of Traditional Chinese Medicine, Changsha, 410208, China.
| | - Yu-Hui Wei
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
28
|
Zhao H, Wei H, He J, Wang D, Li W, Wang Y, Ai Y, Yang J. Propofol disrupts cell carcinogenesis and aerobic glycolysis by regulating circTADA2A/miR-455-3p/FOXM1 axis in lung cancer. Cell Cycle 2020; 19:2538-2552. [PMID: 32857667 DOI: 10.1080/15384101.2020.1810393] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The involvement of propofol and circular RNAs (circRNAs) in lung cancer progression has been identified. However, the relationship between propofol and circRNAs as well as the underlying molecular mechanisms on lung cancer development remain unclear. Cell viability, migration and invasion were measured by cell counting kit-8 assay, 5-bromo-2-deoxyuridine (BrdU) and transwell assay. Glycolytic metabolism was calculated by measuring the glucose consumption, lactate production and extracellular acidification. Western blot was used to detect the protein of glucose transporter 1 (GLUT1), glycolysis enzymes, and forkhead box M1 (FOXM1). The expression of circRNA transcriptional adaptor 2A (circTADA2A), microRNA (miR)-455-3p and FOXM1 mRNA was detected by quantitative real-time polymerase chain reaction. The interaction between miR-455-3p and circTADA2A or FOXM1 was analyzed using the dual-luciferase reporter assay. Murine xenograft model was established to perform in vivo experiments. We found propofol treatment alleviated lung cancer cell proliferation, migration, invasion and aerobic glycolysis in vitro as well as inhibited tumor growth in vivo. Propofol decreased the level of circTADA2A and exerted anti-tumor effects by regulating circTADA2A. MiR-455-3p directly interacted with circTADA2A and FOXM1 in lung cancer cells, and circTADA2A could regulate FOXM1 expression by binding to miR-455-3p. Subsequently, rescue assay showed that propofol inhibited cell proliferation, migration, invasion and aerobic glycolysis by regulating circTADA2A/miR-455-3p/FOXM1 axis in lung cancer. Collectively, propofol suppressed cell carcinogenesis and aerobic glycolysis by regulating circTADA2A/miR-455-3p/FOXM1 axis in lung cancer, providing an effective clinical implication for propofol to prevent the development of lung cancer.
Collapse
Affiliation(s)
- Huaping Zhao
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, Henan, China
| | - Hua Wei
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, Henan, China
| | - Juan He
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, Henan, China
| | - Dongmei Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, Henan, China
| | - Weihao Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, Henan, China
| | - Yanping Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, Henan, China
| | - Yanqiu Ai
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, Henan, China
| | - Jianjun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University , Zhengzhou, Henan, China
| |
Collapse
|
29
|
IGF2-AS knockdown inhibits glycolysis and accelerates apoptosis of gastric cancer cells through targeting miR-195/CREB1 axis. Biomed Pharmacother 2020; 130:110600. [PMID: 34321174 DOI: 10.1016/j.biopha.2020.110600] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 07/30/2020] [Accepted: 08/02/2020] [Indexed: 02/08/2023] Open
Abstract
Dysregulation of long non-coding RNA (lncRNA) insulin growth factor 2 antisense (IGF2-AS) is being found to have relevance to tumorigenesis, including gastric cancer (GC). The purpose of this study was to further explore the detailed role and molecular mechanism of IGF2-AS in GC progression. The expression levels of IGF2-AS, miR-195 and cAMP responsive element binding protein 1 (CREB1) mRNA were assessed by qRT-PCR. Glucose consumption and lactate production were determined using a corresponding Commercial Assay Kit. Hexokinase 2 (HK2) and CREB1 protein levels were detected using western blot. Cell apoptosis was determined by flow cytometry. The targeted interaction between miR-195 and IGF2-AS or CREB1 was validated using dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. Our data revealed that IGF2-AS was upregulated in GC tissues and predicted poor prognosis. IGF2-AS knockdown hampered glycolysis and accelerated apoptosis of GC cells. Moreover, IGF2-AS acted as a sponge of miR-195 and CREB1 was a direct target of miR-195. MiR-195 mediated the regulatory effect of IGF2-AS knockdown on GC cell glycolysis and apoptosis. MiR-195 exerted its regulatory effect on GC cell glycolysis and apoptosis by CREB1. Furthermore, IGF2-AS regulated CREB1 expression via sponging miR-195. In conclusion, our study suggested that IGF2-AS knockdown suppressed glycolysis and facilitated apoptosis in GC cells at least partly through sponging miR-195 and modulating CREB1 expression, highlighting a novel therapeutic strategy for GC treatment.
Collapse
|
30
|
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
|
31
|
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: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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 GynecologyThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Shuya Pan
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Wenxiao Jiang
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Fang Xue
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xueqiong Zhu
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| |
Collapse
|
32
|
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
|
33
|
Liu F, Qiu F, Fu M, Chen H, Wang H. Propofol Reduces Epithelial to Mesenchymal Transition, Invasion and Migration of Gastric Cancer Cells through the MicroRNA-195-5p/Snail Axis. Med Sci Monit 2020; 26:e920981. [PMID: 32115570 PMCID: PMC7067052 DOI: 10.12659/msm.920981] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Gastric cancer (GC) is a life-threating malignancy worldwide. Accumulating studies suggest propofol has anti-tumor functions in addition to the anesthetic effect. This study aimed to figure out the effects of propofol treatment in GC development. MATERIAL AND METHODS Human GC SGC-7901 and NCI-N87 cells were treated with different doses of propofol. Then the invasion and migration of GC cells was measured. SGC-7901 cells following 10 μM propofol treatment were applied in the following experiments. MicroRNAs (miRNAs) with differential expression in cells with or without propofol treatment were analyzed. Expression of miR-195-5p, Snail, vimentin and E-cadherin in SGC-7901 cells was measured, and then loss-of-function of miR-195-5p and gain-of-function of Snail were performed. Target relation between miR-195-5p and Snail was confirmed using luciferase assay. Xenograft tumor was induced in nude mice to identify the effect of propofol on GC in vivo. RESULTS Propofol reduced epithelial to mesenchymal transition (EMT), invasion and migration of GC cells in a dose-dependent manner. Propofol elevated miR-195-5p expression but reduced Snail expression, and it reduced vimentin but increased E-cadherin expression in SGC-7901 cells. miR-195-5p directly bound to Snail. miR-195-5p inhibition or Snail promotion reversed propofol-inhibited malignant behaviors of SGC-7901 cells. In vitro results were reproduced in in vivo experiments. CONCLUSIONS Our study found that propofol could inhibit EMT, invasion, and migration of GC cells by promoting miR-195-5p expression and suppressing Snail expression. This study may provide novel insights in GC treatment.
Collapse
Affiliation(s)
- Fenghua Liu
- Department of Anesthesiology, Yidu Central Hospital, Weifang, Shandong, China (mainland)
| | - Fengyu Qiu
- Department of Anesthesiology, Yidu Central Hospital, Weifang, Shandong, China (mainland)
| | - Min Fu
- Department of Anesthesiology, Yidu Central Hospital, Weifang, Shandong, China (mainland)
| | - Huayong Chen
- Department of Anesthesiology, Yidu Central Hospital, Weifang, Shandong, China (mainland)
| | - Hui Wang
- Operating Room, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China (mainland)
| |
Collapse
|
34
|
Farooqi AA, Adylova A, Sabitaliyevich UY, Attar R, Sohail MI, Yilmaz S. Recent updates on true potential of an anesthetic agent as a regulator of cell signaling pathways and non-coding RNAs in different cancers: Focusing on the brighter side of propofol. Gene 2020; 737:144452. [PMID: 32044408 DOI: 10.1016/j.gene.2020.144452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/27/2020] [Accepted: 02/04/2020] [Indexed: 02/07/2023]
Abstract
There has always been a quest to search for synthetic and natural compounds having premium pharmacological properties and minimum off-target and/or side effects. Therefore, in accordance with this approach, scientists have given special attention to the molecules having remarkable ability to target oncogenic protein network, restore drug sensitivity and induce apoptosis in cancer cells. The mechanisms through which general anesthetics modulated wide-ranging deregulated cell signaling pathways and non-coding RNAs remained unclear. However, rapidly accumulating experimentally verified evidence has started to resolve this long-standing mystery and a knowledge about these important molecular targets has surfaced and how these drugs act at the molecular level is becoming more understandable. In this review we have given special attention to available evidence related to ability of propofol to modulate Wnt/β-catenin, JAK/STAT and mTOR-driven pathway. Excitingly, great strides have been made in sharpening our concepts related to potential of propofol to modulate non-coding RNAs in different cancers. Collectively, these latest findings offer interesting, unexplored opportunities to target deregulated signaling pathways to induce apoptosis in drug-resistant cancers.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan.
| | - Aima Adylova
- Department of Postgraduate Education and Research, Kazakhstan Medical University KSPH, Almaty, Kazakhstan
| | | | - Rukset Attar
- Department of Obstetrics and Gynecology, Yeditepe University, Turkey
| | | | - Seher Yilmaz
- Department of Anatomy, Yozgat Bozok University Faculty of Medicine, Yozgat, Turkey
| |
Collapse
|
35
|
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
|
36
|
Li C, Xia M, Wang H, Li W, Peng J, Jiang H. Propofol facilitates migration and invasion of oral squamous cell carcinoma cells by upregulating SNAI1 expression. Life Sci 2020; 241:117143. [DOI: 10.1016/j.lfs.2019.117143] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 02/07/2023]
|
37
|
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
|
38
|
Ashrafizadeh M, Ahmadi Z, Farkhondeh T, Samarghandian S. Anti-tumor Activity of Propofol: A Focus on MicroRNAs. Curr Cancer Drug Targets 2020; 20:104-114. [PMID: 31657687 DOI: 10.2174/1568009619666191023100046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [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
|
39
|
Zhao X, Hu GF, Shi YF, Xu W. Research Progress in microRNA-Based Therapy for Gastric Cancer. Onco Targets Ther 2019; 12:11393-11411. [PMID: 31920330 PMCID: PMC6935305 DOI: 10.2147/ott.s221354] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 12/10/2019] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer (GC) is one of the leading causes of tumor-related mortality. In addition to surgery and endoscopic resection, systemic therapy remains the main treatment option for GC, especially for advanced-stage disease and for cases not suitable for surgical therapy. Hence, improving the efficacy of systemic therapy is still an urgent problem to overcome. In the past decade, the essential roles of microRNAs (miRNAs) in tumor treatment have been increasingly recognized. In particular, miRNAs were recently shown to reverse the resistance to chemotherapy drugs such as 5-fluorouracil, cisplatin, and doxorubicin. Synthesized nanoparticles loaded with mimics or inhibitors of miRNAs can directly target tumor cells to suppress their growth. Moreover, exosomes may serve as promising safe carriers for mimics or inhibitors of miRNAs to treat GC. Some miRNAs have also been shown to play roles in the mechanism of action of other anti-tumor drugs. Therefore, in this review, we highlight the research progress on microRNA-based therapy in GC and discuss the challenges and prospects associated with this strategy. We believe that microRNA-based therapy has the potential to offer a clinical benefit to GC patients, and this review would contribute to and motivate further research to promote this field toward this ultimate goal.
Collapse
Affiliation(s)
- Xu Zhao
- Department of Hepatology, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| | - Gao-Feng Hu
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People's Republic of China.,Department of Clinical Laboratory, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| | - Yan-Fen Shi
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, People's Republic of China
| | - Wei Xu
- Department of Clinical Laboratory, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| |
Collapse
|
40
|
Cata JP, Owusu-Agyemang P, Kapoor R, Lonnqvist PA. Impact of Anesthetics, Analgesics, and Perioperative Blood Transfusion in Pediatric Cancer Patients: A Comprehensive Review of the Literature. Anesth Analg 2019; 129:1653-1665. [PMID: 31743187 DOI: 10.1213/ane.0000000000004314] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer is the leading cause of death by disease in developed countries. Children and adolescents with cancer need surgical interventions (ie, biopsy or major surgery) to diagnose, treat, or palliate their malignancies. Surgery is a period of high vulnerability because it stimulates the release of inflammatory mediators, catecholamines, and angiogenesis activators, which coincides with a period of immunosuppression. Thus, during and after surgery, dormant tumors or micrometastasis (ie, minimal residual disease) can grow and become clinically relevant metastasis. Anesthetics (ie, volatile agents, dexmedetomidine, and ketamine) and analgesics (ie, opioids) may also contribute to the growth of minimal residual disease or disease progression. For instance, volatile anesthetics have been implicated in immunosuppression and direct stimulation of cancer cell survival and proliferation. Contrarily, propofol has shown in vitro anticancer effects. In addition, perioperative blood transfusions are not uncommon in children undergoing cancer surgery. In adults, an association between perioperative blood transfusions and cancer progression has been described for some malignancies. Transfusion-related immunomodulation is one of the mechanisms by which blood transfusions can promote cancer progression. Other mechanisms include inflammation and the infusion of growth factors. In the present review, we discuss different aspects of tumorigenesis, metastasis, angiogenesis, the immune system, and the current studies about the impact of anesthetics, analgesics, and perioperative blood transfusions on pediatric cancer progression.
Collapse
Affiliation(s)
- Juan P Cata
- From the Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
- Anesthesiology and Surgical Oncology Research Group, Houston, Texas
| | - Pascal Owusu-Agyemang
- From the Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
- Anesthesiology and Surgical Oncology Research Group, Houston, Texas
| | - Ravish Kapoor
- From the Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
- Anesthesiology and Surgical Oncology Research Group, Houston, Texas
| | - Per-Arne Lonnqvist
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
41
|
Zhu F, Li Q, Yang Y, Wang L, Wang J. Propofol Suppresses Proliferation, Migration, Invasion And Promotes Apoptosis By Upregulating microRNA-140-5p In Gastric Cancer Cells. Onco Targets Ther 2019; 12:10129-10138. [PMID: 31819507 PMCID: PMC6885654 DOI: 10.2147/ott.s225360] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/16/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose This study aimed to investigate the anti-tumor effect of propofol on gastric cancer (GC) and its underlying mechanisms. Patients and methods SGC-7901 and MKN45 cells were transfected and divided into the following groups: Control group, Propofol group, Propofol+miR-140-5p inhibitor group and miR-140-5p inhibitor group. Moreover, cell proliferation, apoptosis, migration and invasion of SGC-7901 and MKN45 cells were evaluated by BrdU incorporation assay, Annexin V-FITC/PI double staining assay, wound healing assay and transwell assay, respectively. The mRNA expressions of matrix metalloproteinase 2 (MMP-2) and MMP-9 were detected by qRT-PCR. Cleaved caspase-3, Bcl-2, MMP-2 and MMP-9 expressions were detected by Western blot. Results Propofol inhibited cell proliferation, migration and invasion, but promoted cell apoptosis in SGC-7901 and MKN45 cells. Propofol also elevated the expression of miR-140-5p. Suppression of miR-140-5p could reverse the effects of propofol on the biological behavior of SGC-7901 and MKN45 cells. Meanwhile, propofol treatment increased the expression of cleaved caspase-3 but decreased Bcl-2, MMP-2 and MMP-9 in SGC-7901 and MKN45 cells. The expression of cleaved caspase-3 was downregulated while Bcl-2, MMP-2 and MMP-9 were upregulated by miR-140-5p suppression. Conclusion Propofol could inhibit cell proliferation, migration and invasion, as well as promote cell apoptosis by upregulating miR-140-5p in gastric cancer cells.
Collapse
Affiliation(s)
- Fengbo Zhu
- Department of Gastroenterology, Jinan Fifth People's Hospital, Jinan City, Shandong Province 250022, People's Republic of China
| | - Qiuxia Li
- Department of Gastroenterology, Jinan Fifth People's Hospital, Jinan City, Shandong Province 250022, People's Republic of China
| | - Ying Yang
- Department of Hyperbaric Oxygen, Jinan Fifth People's Hospital, Jinan City, Shandong Province 250022, People's Republic of China
| | - Liangui Wang
- Department of Gastroenterology, Jinan Fifth People's Hospital, Jinan City, Shandong Province 250022, People's Republic of China
| | - Jing Wang
- Department of Anesthesiology, Jinan Fifth People's Hospital, Jinan City, Shandong Province 250022, People's Republic of China
| |
Collapse
|
42
|
Han X, Sun F, Zhang Y, Wang J, Liu Q, Gao P, Zhang S. The protective effect of propofol on ionizing radiation-induced hematopoietic system damage in mice. RSC Adv 2019; 9:36366-36373. [PMID: 35540614 PMCID: PMC9075036 DOI: 10.1039/c9ra07262d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/19/2019] [Accepted: 10/26/2019] [Indexed: 12/18/2022] Open
Abstract
The hematopoietic system is highly sensitive to ionizing radiation (IR), and IR can cause injury to hematopoietic stem cells (HSCs); the main reason for this may be elevated reactive oxygen species (ROS) levels. Propofol is an anesthetic drug commonly used in clinical practice. The chemical structure of propofol is similar to that of vitamin E, and propofol has an antioxidant capacity. Therefore, in this work the effect of using propofol to protect against IR-induced hematopoietic system injury is evaluated. The data suggested that when the irradiated mice were treated with 20 mg kg-1 of propofol, the survival rate of lethally irradiated mice increased significantly, furthermore, the radiation-induced decrease of white blood cells (WBCs), red blood cells (RBCs), hemoglobin (HGC) and platelets (PLT) in peripheral blood is improved significantly. In addition, propofol could also increase the irradiated HSC and hematopoietic progenitor cell (HPC) frequencies, improving the self-renewal and differentiation abilities of HSCs and HPCs in irradiated mice. Next the ROS levels in HSCs and HPCs were measured, and the results showed that propofol could effectively decrease the ROS levels in these cells. The underlying ROS-scavenging mechanisms are further explored, and the results show that the Nrf2 pathway plays an important role in propofol's radiation protective effects, however, propofol can also increase the proliferation of the Nrf2 inhibitor-treated Lineage- cells after exposure to 4 Gy radiation. The data suggest that propofol has a radio-protective effect against IR-induced hematopoietic system damage through reducing cellular ROS in HSCs and HPCs partly through the Nrf2 pathway.
Collapse
Affiliation(s)
- Xiaoliang Han
- Affiliated Hospital, North China University of Science and Technology Tangshan Hebei 063000 China
| | - Fengtao Sun
- Affiliated Hospital, North China University of Science and Technology Tangshan Hebei 063000 China
| | - Ying Zhang
- Affiliated Hospital, North China University of Science and Technology Tangshan Hebei 063000 China
| | - Jinyan Wang
- Tangshan Gongren Hospital Tangshan Hebei 063000 China
| | - Qingguo Liu
- Tangshan Gongren Hospital Tangshan Hebei 063000 China
| | - Ping Gao
- Affiliated Hospital, North China University of Science and Technology Tangshan Hebei 063000 China
| | - Shubo Zhang
- Affiliated Hospital, North China University of Science and Technology Tangshan Hebei 063000 China
| |
Collapse
|
43
|
Sun Y, Sun H. Retracted
:Propofol exerts anticancer activity on hepatocellular carcinoma cells by raising lncRNA DGCR5. J Cell Physiol 2019; 235:2963-2972. [DOI: 10.1002/jcp.29202] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 09/03/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Yan Sun
- Department of Anesthesiology China‐Japan Union Hospital of Jilin University Changchun China
| | - Hai Sun
- Department of Anesthesiology China‐Japan Union Hospital of Jilin University Changchun China
| |
Collapse
|
44
|
Wang C, Qu Z, Kong L, Xu L, Zhang M, Liu J, Yang Z. RETRACTED: Quercetin ameliorates lipopolysaccharide-caused inflammatory damage via down-regulation of miR-221 in WI-38 cells. Exp Mol Pathol 2019; 108:1-8. [PMID: 30849307 DOI: 10.1016/j.yexmp.2019.03.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/21/2019] [Accepted: 03/05/2019] [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. Given the comments of Dr Elisabeth Bik regarding this article “… the Western blot bands in all 400+ papers are all very regularly spaced and have a smooth appearance in the shape of a dumbbell or tadpole, without any of the usual smudges or stains. All bands are placed on similar looking backgrounds, suggesting they were copy/pasted from other sources, or computer generated”, the journal requested the authors to provide the raw data. However, the authors were not able to fulfil this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Chong Wang
- Children's Medical Center, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Zhenghai Qu
- Children's Medical Center, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Lingpeng Kong
- Children's Medical Center, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Lei Xu
- Children's Medical Center, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Mengxue Zhang
- Children's Medical Center, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jianke Liu
- Children's Medical Center, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Zhaochuan Yang
- Children's Medical Center, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| |
Collapse
|
45
|
Ren YL, Zhang W. Propofol promotes apoptosis of colorectal cancer cells via alleviating the suppression of lncRNA HOXA11-AS on miRNA let-7i. Biochem Cell Biol 2019; 98:90-98. [PMID: 31013434 DOI: 10.1139/bcb-2018-0235] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
To date, surgical resection is the mainstay for the treatment of colorectal cancer (CRC). Propofol (2,6-diisopropylphenol), one of the most commonly used intravenous anaesthetic agents, has been reported to be involved in modulating the malignancy of a variety of human cancers. However, the underlying mechanisms remain poorly understood. In this study, using a cell counting kit (CCK-8), flow cytometry, and caspase-3 cleavage assays, we found that propofol promoted cell apoptosis and inhibited cell proliferation in both Colo205 and SW620 cells, through the down-regulation of HOXA11-AS and up-regulation of let-7i. Moreover, gain-of-function studies of HOXA11-AS or loss-of-function studies of let-7i also revealed a negative correlation between HOXA11-AS and let-7i in propofol-mediated biological functions of CRC cells. Furthermore, our mechanistic experiments revealed that HOXA11-AS acts as a molecular sponge for let-7i, thereby regulating the expression of ABCC10. We investigate the theory that propofol suppresses colorectal cancer tumorigenesis by modulating the HOXA11-AS-let-7i-ABCC10 regulatory network, indicating the potential for propofol to control CRC development.
Collapse
Affiliation(s)
- Yan-Ling Ren
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Wei Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.,Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
46
|
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: 35] [Impact Index Per Article: 5.8] [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
|
47
|
Chen X, Wang A. Clinical significance of miR-195 in hepatocellular carcinoma and its biological function in tumor progression. Onco Targets Ther 2019; 12:527-534. [PMID: 30666131 PMCID: PMC6330974 DOI: 10.2147/ott.s190108] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most lethal cancer types all over the world. Chronic viral hepatitis B and hepatitis C are risk factors that are associated with the development of HCC. The aim of this study is to identify the diagnostic role of serum miR-195 in HCC. Patients and methods The expression levels of miR-195 were detected in 120 HCC patients, 64 hepatitis only patients, and 118 healthy control as well as 4 HCC cell lines, by using quantitative real-time PCR. The association of miR-195 with clinicopathological parameters of patients was analyzed with the chi-squared test. The receiver operating characteristic (ROC) curve was adopted to estimate the potential diagnostic value of miR-195. The cell experiments were carried out to verify the functional role of miR-195. Results The expression of miR-195 was downregulated in HCC cells and serum of patients compared to the controls (all P<0.05). The miR-195 expression was associated with lymph node metastasis and TNM stage. The ROC curve analysis showed that miR-195 may be a noninvasive diagnostic marker for patients. By using miR-195 mimic or inhibitor, cell proliferation, migration, and invasion were inhibited by miR-195 overexpression but promoted by reduced expression of miR-195. Conclusion The downregulation of miR-195 may serve as a novel diagnostic biomarker for differentiating HCC patients, healthy individuals, and hepatitis patients, and may involve in the tumor progression of HCC.
Collapse
Affiliation(s)
- Xiaoyan Chen
- Department of Laboratory Medicine, Women and Children's Hospital of Linyi City, Shandong 276000, People's Republic of China,
| | - Angang Wang
- Department of Laboratory Medicine, People's Hospital of Yutai County, Shandong 272300, People's Republic of China
| |
Collapse
|
48
|
Translation elongation factor eEF1Bα is identified as a novel prognostic marker of gastric cancer. Int J Biol Macromol 2018; 126:345-351. [PMID: 30572058 DOI: 10.1016/j.ijbiomac.2018.12.126] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/15/2018] [Accepted: 12/15/2018] [Indexed: 11/24/2022]
Abstract
Gastric cancer (GC) is a common cancer in humans. Although overexpression of eukaryotic translation elongation factor eEF1Bα is associated with cancer onset and progression, little is known about its expression in GC and its prognostic significance. Here we used immunohistochemistry to analyze eEF1Bα expression in the following tissue types: GC, normal gastric, chronic gastritis, intestinal metaplasia, and intraepithelial neoplasia. These data were correlated with patients' clinical information. eEF1Bα was expressed at levels approximately three times higher in GC tissues compared with normal gastric tissues. High expression of eEF1Bα was significantly associated with histological type, TNM stage, tumor size, and distant metastases. GC patients with high eEF1Bα expression experienced significantly shorter overall survival. Bioinformatics analysis indicated that eEF1Bα may be associated with protein synthesis, energy metabolism, cell cycle, and the p53 signaling pathway. We identified the products of RPL10A and RPS13 as critical components of a network comprising eEF1Bα. We concluded that high eEF1Bα expression is associated with poor overall survival and may serve as an independent prognostic factor of GC. Further, we proposed that eEF1Bα likely mediates the development of GC through the cell cycle and p53 signaling pathway. Together, our findings suggest that eEF1Bα could be an effective prognostic biomarker for GC.
Collapse
|