1
|
Li Y, Yu ND, Ye XL, Jiang MC, Chen XQ. Construction of lung cancer serum markers based on ReliefF feature selection. Comput Methods Biomech Biomed Engin 2024; 27:1215-1223. [PMID: 37489703 DOI: 10.1080/10255842.2023.2235045] [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: 05/05/2023] [Accepted: 07/03/2023] [Indexed: 07/26/2023]
Abstract
Serum miRNAs are available clinical samples for cancer screening. Identifying early serum markers in lung cancer (LC) is essential for patients' early diagnosis and clinical treatment. Expression data of serum miRNAs of lung adenocarcinoma (LUAD) patients and healthy individuals were downloaded from the Gene Expression Omnibus (GEO). These data were normalized and subjected to differential expression analysis to obtain differentially expressed miRNAs (DEmiRNAs). The DEmiRNAs were subsequently subjected to ReliefF feature selection, and subsets closely related to cancer were screened as candidate feature miRNAs. Thereafter, a Gaussian Naive Bayes (NB), Support Vector Machine (SVM), and Random Forest (RF) classifier were constructed based on these candidate feature miRNAs. Then the best diagnostic signature was constructed through NB combined with incremental feature selection (IFS). Thereafter, these samples were subjected to principal component analysis (PCA) based on miRNAs with optimal predictive performance. Finally, the peripheral serum miRNAs of 64 LUAD patients and 59 normal individuals were extracted for qRT-PCR analysis to validate the performance of the diagnostic model in respect of clinical detection. Finally, according to area under the curve (AUC) and accuracy values, the NB classifier composed of miR-5100 and miR-663a manifested the most outstanding diagnostic performance. The PCA results also revealed that the 2-miRNA diagnostic signature could effectively distinguish cancer patients from healthy individuals. Finally, qRT-PCR results of clinical serum samples revealed that miR-5100 and miR-663a expression in tumor samples was remarkably higher than that in normal samples. The AUC of the 2-miRNA diagnostic signature was 0.968. In summary, we identified markers (miR-5100 and miR-663a) in serum for early LUAD screening, providing ideas for developing early LUAD diagnostic models.
Collapse
Affiliation(s)
- Yong Li
- Department of Respiration Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Nan-Ding Yu
- Department of Respiration Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Xiang-Li Ye
- Department of Respiration Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Mei-Chen Jiang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Xiang-Qi Chen
- Department of Respiration Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| |
Collapse
|
2
|
Yang L, Zhu Z, Zheng Y, Yang J, Liu Y, Shen T, Li M, He H, Huang H, Dai W. RAB6A functions as a critical modulator of the stem-like subsets in cholangiocarcinoma. Mol Carcinog 2023; 62:1460-1473. [PMID: 37278569 DOI: 10.1002/mc.23589] [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: 02/22/2023] [Revised: 05/08/2023] [Accepted: 05/25/2023] [Indexed: 06/07/2023]
Abstract
RAB6A is a member of RAB GTPase family and plays an important role in the targeted transport of neurotrophic receptors and inflammatory cytokines. RAB6A-mediated secretory pathway is involved in many physiological and pathological processes. Defects in RAB6A-mediated secretory pathway may lead to the development of many diseases, including cancer. However, its role in cholangiocarcinoma (CCA) has not yet been revealed. We explored the regulatory role of RAB6A in the stem-like subsets of CCA. We showed that RAB6A knockdown (KD) impedes cancer stem cells (CSCs) properties and epithelial-mesenchymal transition in vitro and that suppression of RAB6A inhibits tumor growth in vivo. We screened target cargos of RAB6A in CCA cells and identified a extracellular matrix component as the target cargo. RAB6A binds directly to OPN, and RAB6A KD suppressed OPN secretion and inhibited the interaction between OPN and αV integrin receptor. Moreover, RAB6A KD inhibited the AKT signaling pathway, which is a downstream effector of the integrin receptor signaling. In addition, shRNA targeting OPN blocked endogenous expression of OPN and consequently weakened CSCs properties in RAB6A-formed spheres. Similarly, inhibitor of AKT signaling, MK2206 also impedes oncogenic function of RAB6A in the stem-like subsets of CCA cells. In conclusion, our findings showed that RAB6A sustains CSCs phenotype maintenance by modulating the secretion of OPN and consequentially activating the downstream AKT signaling pathway. Targeting the RAB6A/OPN axis may be an effective strategy for CCA therapy.
Collapse
Affiliation(s)
- Liangfang Yang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhiwen Zhu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yang Zheng
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiaqi Yang
- Institution of Plastic Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yuxin Liu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Tingyun Shen
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Mingyi Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Huijuan He
- Clinical Research Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Haili Huang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Institution of Plastic Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Wei Dai
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
3
|
Wu Y, Duan Y, Han W, Cao J, Ye B, Chen P, Li H, Wang Y, Liu J, Fang Y, Yue K, Wu Y, Wang X, Jing C. Deubiquitinase YOD1 suppresses tumor progression by stabilizing E3 ligase TRIM33 in head and neck squamous cell carcinoma. Cell Death Dis 2023; 14:517. [PMID: 37573347 PMCID: PMC10423255 DOI: 10.1038/s41419-023-06035-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 07/24/2023] [Accepted: 08/02/2023] [Indexed: 08/14/2023]
Abstract
Ubiquitination is a reversible process that not only controls protein synthesis and degradation, but also is essential for protein transport, localization and biological activity. Deubiquitinating enzyme (DUB) dysfunction leads to various diseases, including cancer. In this study, we aimed to explore the functions and mechanisms of crucial DUBs in head and neck squamous cell carcinoma (HNSCC). Based on bioinformatic analysis and immunohistochemistry detection, YOD1 was identified to be significantly downregulated in HNSCC specimens compared with adjacent normal tissues. Further analysis revealed that reduced YOD1 expression was associated with the malignant progression of HNSCC and indicated poor prognosis. The results of the in vitro and in vivo experiments verified that YOD1 depletion significantly promoted growth, invasion, and epithelial-mesenchymal transition in HNSCC. Mechanistically, YOD1 inhibited the activation of the ERK/β-catenin pathway by suppressing the ubiquitination and degradation of TRIM33, leading to the constriction of HNSCC progression. Overall, our findings reveal the molecular mechanism underlying the role of YOD1 in tumor progression and provide a novel potential therapeutic target for HNSCC treatment.
Collapse
Affiliation(s)
- Yue Wu
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yuansheng Duan
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Wei Han
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jiayan Cao
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Beibei Ye
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Peng Chen
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Hong Li
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yuxuan Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jin Liu
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yan Fang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Kai Yue
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yansheng Wu
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Xudong Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Chao Jing
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| |
Collapse
|
4
|
Tian Y, Cui X, Guan X, Meng X, Zheng M, Wang X, Cheng G, Xia Y, Ye M. Differential expression profile of microRNAs in the lung tissues of coal workers with pneumoconiosis and patients with silicosis. Toxicol Ind Health 2023; 39:204-217. [PMID: 36840710 DOI: 10.1177/07482337231156281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Abstract
The purpose of this study was to characterize the microRNA (miRNA) profile of the lung tissues from coal workers' pneumoconiosis (CWP) and silicosis and to analyze the changes in downstream genes, biological processes, and signaling pathways based on the differently expressed miRNAs. Lung tissues from three CWP patients, eight silicosis patients, and four healthy controls were collected and analyzed for their miRNA profiles using Affymetrix® GeneChip® miRNA Arrays. Differentially expressed miRNAs (DEMs) were identified between the different groups. The miRanda and TargetScan databases were used to predict the putative target genes, and volcano and heat maps were drawn. Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analyses were then performed to screen the DEMs-associated biological process and signaling pathways, respectively. Further identification with a comprehensive literature research involving particle exposure, fibrosis, inflammation and lung cancer were used to further screen DEMs of CWP and silicosis. Microarray data showed that 375 and 88 miRNAs were differentially expressed in CWP and silicosis lung tissues compared with healthy lung tissues, while 34 miRNAs were differentially expressed in CWP compared with silicosis lung tissues. The GO and KEGG pathway analyses showed that, the target genes were mainly enriched in the TGF-β, MAPK, p53 and other signal pathways. These results provided insight into the miRNA-related underlying mechanisms of CWP and silicosis, and they provided new clues for miRNAs as biomarkers for the diagnosis and differential diagnosis of these two diseases.
Collapse
Affiliation(s)
- Yilin Tian
- National Institute for Occupational Health and Poison Control, 12415Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiuqing Cui
- Hubei Provincial Key Laboratory for Applied Toxicology, 498598Hubei Provincial Center for Disease Control and Prevention, Hubei, China
| | - Xin Guan
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Xiang Meng
- National Institute for Occupational Health and Poison Control, 12415Chinese Center for Disease Control and Prevention, Beijing, China
| | - Min Zheng
- National Institute for Occupational Health and Poison Control, 12415Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xin Wang
- National Institute for Occupational Health and Poison Control, 12415Chinese Center for Disease Control and Prevention, Beijing, China
| | - Guoping Cheng
- Ward II of Respiratory and Critical Care Medicine, Huangshi Second Hospital, Huangshi, China
| | - Ying Xia
- Hubei Provincial Key Laboratory for Applied Toxicology, 498598Hubei Provincial Center for Disease Control and Prevention, Hubei, China
| | - Meng Ye
- National Institute for Occupational Health and Poison Control, 12415Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
5
|
Konoshenko M, Lansukhay Y, Krasilnikov S, Laktionov P. MicroRNAs as Predictors of Lung-Cancer Resistance and Sensitivity to Cisplatin. Int J Mol Sci 2022; 23:7594. [PMID: 35886942 PMCID: PMC9321818 DOI: 10.3390/ijms23147594] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Platinum-based chemotherapy, cisplatin (DDP) specifically, is the main strategy for treating lung cancer (LC). However, currently, there is a lack of predictive drug-resistance markers, and there is increased interest in the development of a reliable and sensitive panels of markers for DDP chemotherapy-effectiveness prediction. MicroRNAs represent a perspective pool of markers for chemotherapy effectiveness. OBJECTIVES Data on miRNAs associated with LC DDP chemotherapy response are summarized and analyzed. MATERIALS AND METHODS A comprehensive review of the data in the literature and an analysis of bioinformatics resources were performed. The gene targets of miRNAs, as well as their reciprocal relationships with miRNAs, were studied using several databases. RESULTS AND DISCUSSION The complex analysis of bioinformatics resources and the literature indicated that the expressions of 12 miRNAs have a high predictive potential for LC DDP chemotherapy responses. The obtained information was discussed from the point of view of the main mechanisms of LC chemoresistance. CONCLUSIONS An overview of the published data and bioinformatics resources, with respect to the predictive microRNA markers of chemotherapy response, is presented in this review. The selected microRNAs and gene panel have a high potential for predicting LC DDP sensitiveness or DDP resistance as well as for the development of a DDP co-therapy.
Collapse
Affiliation(s)
- Maria Konoshenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia;
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, 630055 Novosibirsk, Russia; (Y.L.); (S.K.)
| | - Yuriy Lansukhay
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, 630055 Novosibirsk, Russia; (Y.L.); (S.K.)
| | - Sergey Krasilnikov
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, 630055 Novosibirsk, Russia; (Y.L.); (S.K.)
| | - Pavel Laktionov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia;
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, 630055 Novosibirsk, Russia; (Y.L.); (S.K.)
| |
Collapse
|
6
|
Zhang H, Wang J, Wang Y, Li J, Zhao L, Zhang T, Liao X. Long Non-Coding LEF1-AS1 Sponge miR-5100 Regulates Apoptosis and Autophagy in Gastric Cancer Cells via the miR-5100/DEK/AMPK-mTOR Axis. Int J Mol Sci 2022; 23:4787. [PMID: 35563178 PMCID: PMC9101949 DOI: 10.3390/ijms23094787] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/21/2022] [Accepted: 04/24/2022] [Indexed: 12/16/2022] Open
Abstract
DEK and miR-5100 play critical roles in many steps of cancer initiation and progression and are directly or indirectly regulated by most promoters and repressors. LEF1-AS1 as a long non-coding RNA can regulate tumor development through sponge miRNA. The effect and regulatory mechanism of DEK on autophagy and apoptosis in gastric cancer (GC), and the role between miR-5100 and DEK or miR-5100 and LEF1-AS1 are still unclear. Our study found that DEK was highly expressed in gastric cancer tissues and cell lines, and knockdown of DEK inhibited the autophagy of cells, promoted apoptosis, and suppressed the malignant phenotype of gastric cancer. DEK regulates autophagy and apoptosis through the AMPK/mTOR signaling pathway. In addition, miR-5100 inhibits autophagy and promotes apoptosis in GC cells while LEF1-AS1 had the opposite effect. Studies have shown that miR-5100 acts by targeting the 3'UTR of DEK, and LEF1-AS1 regulates the expression of miR-5100 by sponging with mIR-5100. In conclusion, our results found that LEF1-AS1 and miR-5100 sponge function, and the miR-5100/DEK/AMPK/mTOR axis regulates autophagy and apoptosis in gastric cancer cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xinghua Liao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430000, China; (H.Z.); (J.W.); (Y.W.); (J.L.); (L.Z.); (T.Z.)
| |
Collapse
|
7
|
Enge AM, Sprenger H, Braeuning A, Hessel-Pras S. Identification of microRNAs Implicated in Modulating Senecionine-Induced Liver Toxicity in HepaRG Cells. Foods 2022; 11:foods11040532. [PMID: 35206009 PMCID: PMC8871147 DOI: 10.3390/foods11040532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 12/14/2022] Open
Abstract
1,2-unsaturated Pyrrolizidine Alkaloids (PAs) are secondary plant metabolites that occur as food contaminants. Upon consumption, they can cause severe liver damage. PAs have been shown to induce apoptosis, to have cytotoxic and genotoxic effects, and to impair bile acid homeostasis in the human hepatoma cell line HepaRG. The major mode of action of PAs is DNA- and protein-adduct formation. Beyond that, nuclear receptor activation has only been observed for one receptor and two PAs, yielding the possibility that other cellular mediators are involved in PA-mediated toxicity. Here, the mode of action of Senecionine (Sc), a prominent and ubiquitous representative of hepatotoxic PAs, was investigated by analyzing 7 hepatic microRNAs (miRNAs) in HepaRG cells. Ultimately, 11 target genes that were predicted with Ingenuity Pathway Analysis software (IPA) were found to be significantly downregulated, while their assigned miRNAs showed significant upregulation of gene expression. According to IPA, these targets are positively correlated with apoptosis and cellular death and are involved in diseases such as hepatocellular carcinoma. Subsequent antagomiR-inhibition analysis revealed a significant correlation between PA-induced miRNA-4434 induction and P21-Activated Kinase-1 (PAK1) downregulation. PAK1 downregulation is usually associated with cell cycle arrest, suggesting a new function of Sc-mediated toxicity in human liver cells.
Collapse
|
8
|
Mello-Grand M, Bruno A, Sacchetto L, Cristoni S, Gregnanin I, Dematteis A, Zitella A, Gontero P, Peraldo-Neia C, Ricotta R, Noonan DM, Albini A, Chiorino G. Two Novel Ceramide-Like Molecules and miR-5100 Levels as Biomarkers Improve Prediction of Prostate Cancer in Gray-Zone PSA. Front Oncol 2021; 11:769158. [PMID: 34868998 PMCID: PMC8640468 DOI: 10.3389/fonc.2021.769158] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/25/2021] [Indexed: 01/04/2023] Open
Abstract
Reliable liquid biopsy-based tools able to accurately discriminate prostate cancer (PCa) from benign prostatic hyperplasia (BPH), when PSA is within the “gray zone” (PSA 4–10), are still urgent. We analyzed plasma samples from a cohort of 102 consecutively recruited patients with PSA levels between 4 and 16 ng/ml, using the SANIST-Cloud Ion Mobility Metabolomic Mass Spectrometry platform, combined with the analysis of a panel of circulating microRNAs (miR). By coupling CIMS ion mobility technology with SANIST, we were able to reveal three new structures among the most differentially expressed metabolites in PCa vs. BPH. In particular, two were classified as polyunsaturated ceramide ester-like and one as polysaturated glycerol ester-like. Penalized logistic regression was applied to build a model to predict PCa, using six circulating miR, seven circulating metabolites, and demographic/clinical variables, as covariates. Four circulating metabolites, miR-5100, and age were selected by the model, and the corresponding prediction score gave an AUC of 0.76 (C.I. = 0.66–0.85). At a specified cut-off, no high-risk tumor was misclassified, and 22 out of 53 BPH were correctly identified, reducing by 40% the false positives of PSA. We developed and applied a novel, minimally invasive, liquid biopsy-based powerful tool to characterize novel metabolites and identified new potential non-invasive biomarkers to better predict PCa, when PSA is uninformative as a tool for precision medicine in genitourinary cancers.
Collapse
Affiliation(s)
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Lidia Sacchetto
- Department of Mathematical Sciences, Politecnico di Torino, Torino, Italy
| | - Simone Cristoni
- I.S.B.-Ion Source & Biotechnologies srl, Biotechnology, Bresso, Italy
| | - Ilaria Gregnanin
- Cancer Genomics Laboratory, Fondazione Edo ed Elvo Tempia, Biella, Italy
| | - Alessandro Dematteis
- Department of Urology, San Giovanni Battista Hospital of Torino, Corso Torino, Italy
| | - Andrea Zitella
- Department of Urology, San Giovanni Battista Hospital of Torino, Corso Torino, Italy
| | - Paolo Gontero
- Department of Urology, San Giovanni Battista Hospital of Torino, Corso Torino, Italy
| | | | - Riccardo Ricotta
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Douglas M Noonan
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy.,Unit of Molecular Pathology, Biochemistry, and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Adriana Albini
- Laboratory of Vascular Cell Biology and Angiogenesis Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Giovanna Chiorino
- Cancer Genomics Laboratory, Fondazione Edo ed Elvo Tempia, Biella, Italy
| |
Collapse
|
9
|
Mondal P, Meeran SM. microRNAs in cancer chemoresistance: The sword and the shield. Noncoding RNA Res 2021; 6:200-210. [PMID: 34977437 PMCID: PMC8669341 DOI: 10.1016/j.ncrna.2021.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer is a multifactorial disease and one of the leading causes of mortality worldwide. Cancer cells develop multiple strategies to reduce drug sensitivity and eventually lead to chemoresistance. Chemoresistance is initiated either by intrinsic factors or due to the prolonged use of chemotherapeutics as acquired resistance. Further, chemoresistance is also one of the major reasons behind tumor recurrence and metastasis. Therefore, overcoming chemoresistance is one of the primary challenges in cancer therapy. Several mechanisms are involved in chemoresistance. Among them, the key role of ABC transporters and tumor microenvironment have been well studied. Recently, microRNAs (miRNAs) regulation in tumor development, metastasis, and chemotherapy has got wider interest due to its role in regulating genes involved in cancer progression and therapy. Noncoding RNAs, including miRNAs, have been associated with the regulation of tumor-suppressor and tumor-promoter genes. Further, miRNA can also be used as a reliable diagnostic and prognostic marker to predict the stage and types of cancer. Recent evidences have revealed that miRNAs regulation also influences the function of drug transporters and the tumor microenvironment, which affects chemosensitivity to cancer cells. Therefore, miRNAs can be a promising target to reverse back chemosensitivity in cancer cells. This review comprehensively discusses the mechanisms involved in cancer chemoresistance and its regulation by miRNAs.
Collapse
Affiliation(s)
- Priya Mondal
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore, 570020, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Syed Musthapa Meeran
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore, 570020, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
10
|
Brunel A, Bégaud G, Auger C, Durand S, Battu S, Bessette B, Verdier M. Autophagy and Extracellular Vesicles, Connected to rabGTPase Family, Support Aggressiveness in Cancer Stem Cells. Cells 2021; 10:1330. [PMID: 34072080 PMCID: PMC8227744 DOI: 10.3390/cells10061330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/22/2022] Open
Abstract
Even though cancers have been widely studied and real advances in therapeutic care have been made in the last few decades, relapses are still frequently observed, often due to therapeutic resistance. Cancer Stem Cells (CSCs) are, in part, responsible for this resistance. They are able to survive harsh conditions such as hypoxia or nutrient deprivation. Autophagy and Extracellular Vesicles (EVs) secretion are cellular processes that help CSC survival. Autophagy is a recycling process and EVs secretion is essential for cell-to-cell communication. Their roles in stemness maintenance have been well described. A common pathway involved in these processes is vesicular trafficking, and subsequently, regulation by Rab GTPases. In this review, we analyze the role played by Rab GTPases in stemness status, either directly or through their regulation of autophagy and EVs secretion.
Collapse
|
11
|
Dong B, Li S, Zhu S, Yi M, Luo S, Wu K. MiRNA-mediated EMT and CSCs in cancer chemoresistance. Exp Hematol Oncol 2021; 10:12. [PMID: 33579377 PMCID: PMC7881653 DOI: 10.1186/s40164-021-00206-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/30/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) are a small group of cancer cells, which contribute to tumorigenesis and cancer progression. Cancer cells undergoing epithelial-to-mesenchymal transition (EMT) acquire the chemoresistant ability, which is regarded as an important feature of CSCs. Thus, there emerges an opinion that the generation of CSCs is considered to be driven by EMT. In this complex process, microRNAs (miRNAs) are found to play a key role. In order to overcome the drug resistance, inhibiting EMT as well as CSCs phenotype seem feasible. Thereinto, regulating the EMT- or CSCs-associated miRNAs is a crucial approach. Herein, we conduct this review to elaborate on the complicated interplay between EMT and CSCs in cancer chemoresistance, which is modulated by miRNAs. In addition, we elucidate the therapeutic strategy to overcome drug resistance through targeting EMT and CSCs.
Collapse
Affiliation(s)
- Bing Dong
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008 China
| | - Shiyu Li
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Shuangli Zhu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Suxia Luo
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008 China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008 China
| |
Collapse
|
12
|
Ashrafizadeh M, Zarrabi A, Hushmandi K, Hashemi F, Moghadam ER, Owrang M, Hashemi F, Makvandi P, Goharrizi MASB, Najafi M, Khan H. Lung cancer cells and their sensitivity/resistance to cisplatin chemotherapy: Role of microRNAs and upstream mediators. Cell Signal 2021; 78:109871. [PMID: 33279671 DOI: 10.1016/j.cellsig.2020.109871] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023]
Abstract
Cisplatin (CP) is a well-known chemotherapeutic agent with excellent clinical effects. The anti-tumor activity of CP has been demonstrated in different cancers such as breast, cervical, reproductive, lung, brain, and prostate cancers. However, resistance of cancer cells to CP chemotherapy has led to its failure in eradication of cancer cells, and subsequent death of patients with cancer. Fortunately, much effort has been put to identify molecular pathways and mechanisms involved in CP resistance/sensitivity. It seems that microRNAs (miRs) are promising candidates in mediating CP resistance/sensitivity, since they participate in different biological aspects of cells such as proliferation, migration, angiogenesis, and differentiation. In this review, we focus on miRs and their regulation in CP chemotherapy of lung cancer, as the most malignant tumor worldwide. Oncogenic miRs trigger CP resistance in lung cancer cells via targeting various pathways such as Wnt/β-catenin, Rab6, CASP2, PTEN, and Apaf-1. In contrast, onco-suppressor miRs inhibit oncogene pathways such as STAT3 to suppress CP resistance. These topics are discussed to determine the role of miRs in CP resistance/sensitivity. We also describe the upstream modulators of miRs such as lncRNAs, circRNAs, NF-κB, SOX2 and TRIM65 and their association with CP resistance/sensitivity in lung cancer cells. Finally, the effect of anti-tumor plant-derived natural compounds on miR expression during CP sensitivity of lung cancer cells is discussed.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla 34956, Istanbul, Turkey; Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul, Turkey
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul, Turkey
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ebrahim Rahmani Moghadam
- Department of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Owrang
- Department of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fardin Hashemi
- Student Research Committee, Department of Physiotherapy, Faculty of Rehabilitation, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia, Centre for Micro-BioRobotics, viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| | | | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran; Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan.
| |
Collapse
|
13
|
Yang L, Liu G, Fu L, Zhong W, Li X, Pan Q. DNA repair enzyme OGG1 promotes alveolar progenitor cell renewal and relieves PM2.5-induced lung injury and fibrosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 205:111283. [PMID: 32977282 DOI: 10.1016/j.ecoenv.2020.111283] [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: 06/19/2020] [Revised: 08/30/2020] [Accepted: 09/01/2020] [Indexed: 06/11/2023]
Abstract
Fine particulate matter (PM2.5) airborne pollution increases the risk of chronic respiratory diseases, such as idiopathic pulmonary fibrosis (IPF), which is characterized by non-specific inflammation of the interstitial lung and extensive deposition of collagen fibers. Type 2 alveolar epithelial cells (AEC2s) are alveolar stem cells in the adult lung that contribute to the lung repair process through complex signaling. Our previous studies demonstrated that OGG1, a kind of DNA repair enzyme, have a critical role in protecting cells from oxidative damage and apoptosis induced by PM2.5, but the contribution of OGG1 in proliferation and self-renewal of AEC2s is not known. Here, we constructed OGG1-/-mice to test the effect and mechanism of OGG1 on PM2.5-induced pulmonary fibrosis and injury in vivo. We detected proliferation and self-renewal of OGG1 overexpression or OGG1 knockout AEC2s after PM2.5 injury by flow cytometry and clone formation. We observed that knockout of OGG1 aggravated pulmonary fibrosis, oxidative stress, and AEC2 cell death in PM2.5-injured mice. In addition, OGG1 is required for the proliferation and renewal of AEC2s after PM2.5 injury. Overexpression of OGG1 promotes the proliferation and self-renewal of AEC2s by inhibiting PM2.5-mediated oxidative stress and NF-κB signaling hyperactivation in vitro. Furthermore, NF-κB inhibitors promoted proliferation and self-renewal of OGG1-deficient AEC2s cells after PM2.5 injury, and attenuated PM2.5-induced pulmonary fibrosis and injury in mice. These data establish OGG1 as a regulator of NF-κB signal that serves to regulate AEC2 cell proliferation and self-renewal, and suggest a mechanism that inhibition of the NF-κB signaling pathway may represent a potential therapeutic strategy for IPF patients with low-expression of OGG1.
Collapse
Affiliation(s)
- Lawei Yang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Gang Liu
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Liyuan Fu
- Guangdong Ocean University Cunjin College, Zhanjiang, 524086, China
| | - Weifeng Zhong
- Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xuenong Li
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Qingjun Pan
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| |
Collapse
|
14
|
Yang L, Liu G, Li X, Xia Z, Wang Y, Lin W, Zhang W, Zhang W, Li X. Small GTPase RAB6 deficiency promotes alveolar progenitor cell renewal and attenuates PM2.5-induced lung injury and fibrosis. Cell Death Dis 2020; 11:827. [PMID: 33012781 PMCID: PMC7533251 DOI: 10.1038/s41419-020-03027-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease characterized by chronic non-specific inflammation of the interstitial lung and extensive deposition of collagen fibers leading to destruction of lung function. Studies have demonstrated that exposure to fine particulate matter (PM2.5) increases the risk of IPF. In order to recover from PM2.5-induced lung injury, alveolar epithelial cells need to be repaired and regenerated to maintain lung function. Type 2 alveolar epithelial cells (AEC2) are stem cells in the adult lung that contribute to the lung repair process through complex signaling. Our previous studies demonstrated that RAB6, a RAS family member lowly expressed in lung cancer, inhibited lung cancer stem cell self-renewal, but it is unclear whether or not and how RAB6 may regulate AEC2 cell proliferation and self-renewal in PM2.5-induced pulmonary fibrosis. Here, we demonstrated that knockout of RAB6 inhibited pulmonary fibrosis, oxidative stress, and AEC2 cell death in PM2.5-injured mice. In addition, knockout of RAB6 decreased Dickkopf 1(DKK1) autocrine and activated proliferation, self-renewal, and wnt/β-catenin signaling of PM2.5-injured AEC2 cells. RAB6 overexpression increased DKK1 autocrine and inhibited proliferation, self-renewal and wnt/β-catenin signaling in AEC2 cells in vitro. Furthermore, DKK1 inhibitors promoted proliferation, self-renewal and wnt/β-catenin signaling of RAB6 overexpressing AEC2 cells, and attenuated PM2.5-induced pulmonary fibrosis in mice. These data establish RAB6 as a regulator of DKK1 autocrine and wnt/β-catenin signal that serves to regulate AEC2 cell proliferation and self-renewal, and suggest a mechanism that RAB6 disruption may promote AEC2 cell proliferation and self-renewal to enhance lung repair following PM2.5 injury.
Collapse
Affiliation(s)
- Lawei Yang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China.,Clinical Research Center, Affiliated Hospital of Guangdong Medical University, 524001, Zhanjiang, China
| | - Gang Liu
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, 524001, Zhanjiang, China
| | - Xiaomin Li
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Zhengyuan Xia
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, 524001, Zhanjiang, China.,Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524001, Zhanjiang, China.,Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| | - Yahong Wang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, 524001, Zhanjiang, China
| | - Weihao Lin
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Wei Zhang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Wenjuan Zhang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Xuenong Li
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China.
| |
Collapse
|
15
|
Wang S, Li MY, Liu Y, Vlantis AC, Chan JY, Xue L, Hu BG, Yang S, Chen MX, Zhou S, Guo W, Zeng X, Qiu S, van Hasselt CA, Tong MC, Chen GG. The role of microRNA in cisplatin resistance or sensitivity. Expert Opin Ther Targets 2020; 24:885-897. [PMID: 32559147 DOI: 10.1080/14728222.2020.1785431] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Cisplatin is a chemotherapy drug that has been used to treat a number of cancers for decades, and is still one of the most commonly used anti-cancer agents. However, some patients do not respond to cisplatin while other patients who were originally sensitive to cisplatin eventually develop chemoresistance, leading to treatment failure or/and tumor recurrence. AREAS COVERED Different mechanisms contribute to cisplatin resistance or sensitivity, involving multiple pathways or/and processes such as DNA repair, DNA damage response, drug transport, and apoptosis. Among the various mechanisms, it appears that microRNAs play an important role in determining the resistance or sensitivity. In this article, we analyzed and summarized recent findings in this area, with the aim that these data can aid further research and understanding, leading to the eventual reduction of cisplatin resistance. EXPERT COMMENTARY microRNAs can positively or negatively regulate cisplatin resistance by acting on molecules or/and pathways related to apoptosis, autophagy, hypoxia, cancer stem cells, NF-κB, and Notch1. It appears that the modulation of relevant microRNAs can effectively re-sensitize cancer cells to cisplatin regimen in certain types of cancers including breast, colorectal, gastric, liver, lung, ovarian, prostate, testicular, and thyroid cancers.
Collapse
Affiliation(s)
- Shanshan Wang
- School of Life Sciences and Biopharmaceutics, Guangdong, Pharmaceutical University , Guangzhou, China.,Department of Otorhinolaryngology, Head and Neck Surgery; The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT , Hong Kong, China
| | - Ming-Yue Li
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital , Hong Kong, China
| | - Yi Liu
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital , Hong Kong, China
| | - Alexander C Vlantis
- Department of Otorhinolaryngology, Head and Neck Surgery; The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT , Hong Kong, China.,The Chinese University of Hong Kong - Shenzhen Ear, Nose and Throat Joint Research Centre, Longgang ENT Hospital , Shenzhen, China
| | - Jason Yk Chan
- Department of Otorhinolaryngology, Head and Neck Surgery; The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT , Hong Kong, China.,The Chinese University of Hong Kong - Shenzhen Ear, Nose and Throat Joint Research Centre, Longgang ENT Hospital , Shenzhen, China
| | - Lingbin Xue
- Department of Otorhinolaryngology, Head and Neck Surgery; The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT , Hong Kong, China
| | - Bao-Guang Hu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Binzhou Medical University , Binzhou, Shenzhen, China
| | - Shucai Yang
- Department of Clinical Laboratory, Pingshan District People's Hospital of Shenzhen , Shenzhen, Guangdong, China
| | - Mo-Xian Chen
- Division of Gastroenterology, Shenzhen Children's Hospital , Shenzhen, China
| | - Shaoming Zhou
- Division of Gastroenterology, Shenzhen Children's Hospital , Shenzhen, China
| | - Wei Guo
- Shenzhen Ritzcon Biological Technology Co., LTD , Shenzhen, Guangdong, China
| | - Xianhai Zeng
- DShenzhen Key Laboratory of ENT, Institute of ENT & Longgang ENT Hospital , Shandong, China.,The Chinese University of Hong Kong - Shenzhen Ear, Nose and Throat Joint Research Centre, Longgang ENT Hospital , Shenzhen, China
| | - Shuqi Qiu
- DShenzhen Key Laboratory of ENT, Institute of ENT & Longgang ENT Hospital , Shandong, China.,The Chinese University of Hong Kong - Shenzhen Ear, Nose and Throat Joint Research Centre, Longgang ENT Hospital , Shenzhen, China
| | - C Andrew van Hasselt
- Department of Otorhinolaryngology, Head and Neck Surgery; The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT , Hong Kong, China.,The Chinese University of Hong Kong - Shenzhen Ear, Nose and Throat Joint Research Centre, Longgang ENT Hospital , Shenzhen, China
| | - Michael Cf Tong
- Department of Otorhinolaryngology, Head and Neck Surgery; The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT , Hong Kong, China.,The Chinese University of Hong Kong - Shenzhen Ear, Nose and Throat Joint Research Centre, Longgang ENT Hospital , Shenzhen, China
| | - George G Chen
- Department of Otorhinolaryngology, Head and Neck Surgery; The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT , Hong Kong, China.,The Chinese University of Hong Kong - Shenzhen Ear, Nose and Throat Joint Research Centre, Longgang ENT Hospital , Shenzhen, China
| |
Collapse
|
16
|
Zhang HM, Li H, Wang GX, Wang J, Xiang Y, Huang Y, Shen C, Dai ZT, Li JP, Zhang TC, Liao XH. MKL1/miR-5100/CAAP1 loop regulates autophagy and apoptosis in gastric cancer cells. Neoplasia 2020; 22:220-230. [PMID: 32315812 PMCID: PMC7167518 DOI: 10.1016/j.neo.2020.03.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 03/23/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE miR-5100 participates in the proliferation of lung cancer and pancreatic cancer cells, and participates in the differentiation of osteoblasts. However, the regulation of gastric cancer cells in gastric cancer cells remains unclear. EXPERIMENTAL DESIGN The blood of patients was collected to detect the expression level of miR-5100, and the apoptosis and autophagy levels of cells were detected using western blot, flow cytometry, and confocal. At the same time, in vitro tumor formation experiments in nude mice were used to verify the results of in vitro experiments. RESULTS The expression of miR-5100 is related to the prognosis of gastric cancer, miR-5100 can enhance the apoptosis level of gastric cancer cells and inhibit the occurrence of autophagy by targeting CAAP1. MKL1 can inhibit the apoptosis of gastric cancer cells and promote the occurrence of autophagy by targeting CAAP1. At the same time, MKL1 can also increase the expression of miR-5100. CONCLUSIONS Our research reveals the mechanism by which the MKL1/miR-5100/CAAP1 loop regulates apoptosis and autophagy levels in gastric cancer cells, and miR-5100 is expected to become a new potential target for gastric cancer treatment.
Collapse
Affiliation(s)
- Hui-Min Zhang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, 430000, PR China
| | - Hui Li
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, 430000, PR China
| | - Gen-Xin Wang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, 430000, PR China.
| | - Jun Wang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, 430000, PR China
| | - Yuan Xiang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, 430000, PR China
| | - You Huang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, 430000, PR China
| | - Chao Shen
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, 430000, PR China
| | - Zhou-Tong Dai
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, 430000, PR China
| | - Jia-Peng Li
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, 430000, PR China
| | - Tong-Cun Zhang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, 430000, PR China.
| | - Xing-Hua Liao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, 430000, PR China.
| |
Collapse
|
17
|
Zhou J, Kang Y, Chen L, Wang H, Liu J, Zeng S, Yu L. The Drug-Resistance Mechanisms of Five Platinum-Based Antitumor Agents. Front Pharmacol 2020; 11:343. [PMID: 32265714 PMCID: PMC7100275 DOI: 10.3389/fphar.2020.00343] [Citation(s) in RCA: 240] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 03/09/2020] [Indexed: 01/17/2023] Open
Abstract
Platinum-based anticancer drugs, including cisplatin, carboplatin, oxaliplatin, nedaplatin, and lobaplatin, are heavily applied in chemotherapy regimens. However, the intrinsic or acquired resistance severely limit the clinical application of platinum-based treatment. The underlying mechanisms are incredibly complicated. Multiple transporters participate in the active transport of platinum-based antitumor agents, and the altered expression level, localization, or activity may severely decrease the cellular platinum accumulation. Detoxification components, which are commonly increasing in resistant tumor cells, can efficiently bind to platinum agents and prevent the formation of platinum–DNA adducts, but the adducts production is the determinant step for the cytotoxicity of platinum-based antitumor agents. Even if adequate adducts have formed, tumor cells still manage to survive through increased DNA repair processes or elevated apoptosis threshold. In addition, autophagy has a profound influence on platinum resistance. This review summarizes the critical participators of platinum resistance mechanisms mentioned above and highlights the most potential therapeutic targets or predicted markers. With a deeper understanding of the underlying resistance mechanisms, new solutions would be produced to extend the clinical application of platinum-based antitumor agents largely.
Collapse
Affiliation(s)
- Jiabei Zhou
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yu Kang
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Lu Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hua Wang
- Department of Urology, Cancer Hospital of Zhejiang Province, Hangzhou, China
| | - Junqing Liu
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Lushan Yu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
18
|
Tang W, Yu X, Zeng R, Chen L. LncRNA-ATB Promotes Cisplatin Resistance in Lung Adenocarcinoma Cells by Targeting the miR-200a/β-Catenin Pathway. Cancer Manag Res 2020; 12:2001-2014. [PMID: 32256108 PMCID: PMC7090201 DOI: 10.2147/cmar.s240695] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 03/02/2020] [Indexed: 12/19/2022] Open
Abstract
Introduction Lung adenocarcinoma (LUAD), which is associated with high morbidity and mortality, is prone to cisplatin resistance, resulting in poor patient prognosis. Long non-coding RNAs (lncRNAs) have complex biological functions in a variety of tumors. Elucidating the underlying molecular mechanisms between lncRNA and cisplatin resistance in LUAD is expected to enable identification of new targets for drug development. Methods Cell proliferation was measured by CCK-8 assay and cell apoptosis was detected using flow cytometry analysis. Luciferase reporter assay was conducted to determine the interaction between lncRNA and MicroRNA. Gene expression was evaluated by Real-Time Quantitative Reverse Transcription Polymerase Chain Reaction and Western blot analysis. Results Long non-coding RNA activated by TGF-β (lncRNA-ATB) was shown to be significantly up-regulated in A549 cells resistant to cisplatin/cis-dichlorodiammineplatinum (II) (cis-DDP) (A549/CDDP cells), compared with corresponding levels in parental A549 cells. Overexpression of lncRNA-ATB significantly elevated cisplatin resistance in LUAD cell lines (A549 and H1975 cells), and this was associated with activation of apoptosis-related genes. Conversely, silencing of lncRNA-ATB decreased cisplatin resistance in LUAD cells. Mechanistically, lncRNA-ATB increased expression of β-catenin by directly binding to MicroRNA-200a (miR-200a), thereby promoting cell survival and cisplatin resistance. Transfection with a miR-200a mimic or treatment with the β-catenin downstream pathway inhibitor IWR-1 could reverse the phenotypes induced by lncRNA-ATB overexpression. Conclusion In summary, this study revealed that lncRNA-ATB is dramatically up-regulated in cisplatin-resistant LUAD cell lines, and that lncRNA-ATB facilitates cell survival by targeting the miR-200a/β-catenin pathway in these cells.
Collapse
Affiliation(s)
- Weiwei Tang
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, Cancer Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian Province 361003, People's Republic of China.,Teaching Hospital of Fujian Medical University, Xiamen, Fujian Province 361003, People's Republic of China
| | - Xiuyi Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, Fujian Province 361003, People's Republic of China
| | - Ru Zeng
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, Cancer Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian Province 361003, People's Republic of China.,Teaching Hospital of Fujian Medical University, Xiamen, Fujian Province 361003, People's Republic of China
| | - Lilin Chen
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, Cancer Center, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian Province 361003, People's Republic of China.,Teaching Hospital of Fujian Medical University, Xiamen, Fujian Province 361003, People's Republic of China
| |
Collapse
|
19
|
Yang LW, Wu XJ, Liang Y, Ye GQ, Che YC, Wu XZ, Zhu XJ, Fan HL, Fan XP, Xu JF. miR-155 increases stemness and decitabine resistance in triple-negative breast cancer cells by inhibiting TSPAN5. Mol Carcinog 2020; 59:447-461. [PMID: 32096299 DOI: 10.1002/mc.23167] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/20/2020] [Accepted: 01/31/2020] [Indexed: 12/24/2022]
Abstract
Effective therapeutic targets for triple-negative breast cancer (TNBC), a special type of breast cancer (BC) with rapid metastasis and poor prognosis, are lacking, especially for patients with chemotherapy resistance. Decitabine (DCA) is a Food and Drug Administration-approved DNA methyltransferase inhibitor that has been proven effective for the treatment of tumors. However, its antitumor effect in cancer cells is limited by multidrug resistance. Cancer stem cells (CSCs), which are thought to act as seeds during tumor formation, regulate tumorigenesis, metastasis, and drug resistance through complex signaling. Our previous study found that miR-155 is upregulated in BC, but whether and how miR-155 regulates DCA resistance is unclear. In this study, we demonstrated that miR-155 was upregulated in CD24- CD44+ BC stem cells (BCSCs). In addition, the overexpression of miR-155 increased the number of CD24- CD44+ CSCs, DCA resistance and tumor clone formation in MDA-231 and BT-549 BC cells, and knockdown of miR-155 inhibited DCA resistance and stemness in BCSCs in vitro. Moreover, miR-155 induced stemness and DCA resistance by inhibiting the direct target gene tetraspanin-5 (TSPAN5). We further confirmed that overexpression of TSPAN5 abrogated the effect of miR-155 in promoting stemness and DCA resistance in BC cells. Our data show that miR-155 increases stemness and DCA resistance in BC cells by targeting TSPAN5. These data provide a therapeutic strategy and mechanistic basis for future possible clinical applications targeting the miR-155/TSPAN5 signaling axis in the treatment of TNBC.
Collapse
Affiliation(s)
- La-Wei Yang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xian-Jin Wu
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Department of Clinical Laboratory, Huizhou Municipal Central Hospital, Huizhou, China
| | - Yi Liang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Guang-Qing Ye
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yu-Chuang Che
- Department of Clinical Laboratory, Huizhou Municipal Central Hospital, Huizhou, China
| | - Xue-Zhen Wu
- Department of Clinical Laboratory, Huizhou Municipal Central Hospital, Huizhou, China
| | - Xiao-Jie Zhu
- Department of Clinical Laboratory, Huizhou Municipal Central Hospital, Huizhou, China
| | - Huo-Liang Fan
- Department of Clinical Laboratory, Huizhou Municipal Central Hospital, Huizhou, China
| | - Xiang-Ping Fan
- Department of Clinical Laboratory, Huizhou Municipal Central Hospital, Huizhou, China
| | - Jun-Fa Xu
- Department of Clinical Immunology, Guangdong Medical University, Dongguan, China
| |
Collapse
|
20
|
Tang H, Han X, Li M, Li T, Hao Y. Linc00221 modulates cisplatin resistance in non-small-cell lung cancer via sponging miR-519a. Biochimie 2019; 162:134-143. [DOI: 10.1016/j.biochi.2019.04.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/24/2019] [Indexed: 10/27/2022]
|
21
|
Feng X, Bai X, Ni J, Wasinger VC, Beretov J, Zhu Y, Graham P, Li Y. CHTOP in Chemoresistant Epithelial Ovarian Cancer: A Novel and Potential Therapeutic Target. Front Oncol 2019; 9:557. [PMID: 31380263 PMCID: PMC6660285 DOI: 10.3389/fonc.2019.00557] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/07/2019] [Indexed: 01/14/2023] Open
Abstract
Objective: Chemoresistance is a major challenge in epithelial ovarian cancer (EOC) treatment. Chromatin target of protein arginine methyltransferase (CHTOP) was identified as a potential biomarker in chemoresistant EOC cell lines using label-free LC-MS/MS quantitative proteomics. Thus, the aim of this study is to investigate the role of CHTOP in chemoresistant EOC and the underlying mechanism. Methods: The expression of CHTOP in human ovarian cancer cells and tissues was detected using immunofluorescence (IF), western blot (WB), and immunohistochemistry (IHC), respectively. Flow cytometry and TUNEL assay were employed to detect the effect of CHTOP knockdown (KD) in chemoresistant EOC cell apoptosis, while colony and sphere formation assays were used to evaluate its effect on cell stemness. The association of CHTOP with cell metastasis was determined using Matrigel invasion and wound-healing assays. Results: The higher level expression of CHTOP protein was found in chemoresistant EOC cells as compared to their sensitive parental cells or normal epithelial ovarian cells. Results from IHC and bioinformatic analysis showed CHTOP was highly expressed in human ovarian cancer tissues and associated with a poor progression-free survival in patients. In addition, CHTOP KD significantly enhanced cisplatin-induced apoptosis, reduced the stemness of chemoresistant EOC cells, and decreased their metastatic potential. Conclusion: Our findings suggest that CHTOP is associated with apoptosis, stemness, and metastasis in chemoresistant EOC cells and might be a promising target to overcome chemoresistance in EOC treatment.
Collapse
Affiliation(s)
- Xiaojie Feng
- Department of Gynaecological Oncology, Henan Cancer Hospital, Zhengzhou, China.,Cancer Care Centre, St. George Hospital, Kogarah, NSW, Australia.,St. George and Sutherland Clinical School, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Xupeng Bai
- Cancer Care Centre, St. George Hospital, Kogarah, NSW, Australia.,St. George and Sutherland Clinical School, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Jie Ni
- Cancer Care Centre, St. George Hospital, Kogarah, NSW, Australia.,St. George and Sutherland Clinical School, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Valerie C Wasinger
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales Sydney, Sydney, NSW, Australia.,School of Medical Science, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Julia Beretov
- Cancer Care Centre, St. George Hospital, Kogarah, NSW, Australia.,St. George and Sutherland Clinical School, University of New South Wales Sydney, Sydney, NSW, Australia.,Anatomical Pathology, NSW Health Pathology, St. George Hospital, Kogarah, NSW, Australia
| | - Ying Zhu
- Cancer Care Centre, St. George Hospital, Kogarah, NSW, Australia.,St. George and Sutherland Clinical School, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Peter Graham
- Cancer Care Centre, St. George Hospital, Kogarah, NSW, Australia.,St. George and Sutherland Clinical School, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Yong Li
- Cancer Care Centre, St. George Hospital, Kogarah, NSW, Australia.,St. George and Sutherland Clinical School, University of New South Wales Sydney, Sydney, NSW, Australia.,School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
22
|
Eupatilin Inhibits Renal Cancer Growth by Downregulating MicroRNA-21 through the Activation of YAP1. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5016483. [PMID: 31179326 PMCID: PMC6507163 DOI: 10.1155/2019/5016483] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 04/02/2019] [Indexed: 02/07/2023]
Abstract
Renal cell carcinoma (RCC) is the second most common human urinary tumor. Eupatilin is the main active ingredient of the traditional Chinese medicine Artemisia asiatica. The effect of Eupatilin on RCC and the underlying mechanism remain unknown. Here, we investigated the anticancer effects and mechanisms of Eupatilin in RCC in vivo and in vitro, laying an experimental foundation for the clinical application of Eupatilin in the treatment of RCC. The results showed that Eupatilin significantly inhibited 786-O cell viability and migration and promoted apoptosis. Eupatilin inhibited the expression of miR-21 in 786-O cells, and overexpression of miR-21 suppressed the effect of Eupatilin on viability, apoptosis, and migration in 786-O cells. Eupatilin inhibited the growth of renal tumors in nude mice by downregulating miR-21. YAP1, which was identified as a target of miR-21, showed significantly lower expression in RCC tissues than in healthy tissues. miR-21 significantly inhibited YAP1 protein expression in 786-O cells and tumor tissues isolated from nude mice, and YAP1 attenuated the effect of miR-21 on the viability, apoptosis, and migration of 786-O cells. In conclusion, Eupatilin inhibited the expression of miR-21, which mediated the proapoptotic and antimigratory effects of Eupatilin by suppressing YAP1 in renal cancer cells. These results suggested that Eupatilin could be a potent agent for the treatment of RCC.
Collapse
|