1
|
He D, Tang H, Yang X, Liu X, Zhang Y, Shi J. Elaboration and validation of a prognostic signature associated with disulfidoptosis in lung adenocarcinoma, consolidated with integration of single-cell RNA sequencing and bulk RNA sequencing techniques. Front Immunol 2023; 14:1278496. [PMID: 37965333 PMCID: PMC10641741 DOI: 10.3389/fimmu.2023.1278496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/13/2023] [Indexed: 11/16/2023] Open
Abstract
Background Lung adenocarcinoma (LUAD), the predominant subtype of non-small cell lung cancer (NSCLC), remains a pervasive global public health concern. Disulfidoptosis, a nascent form of regulated cell death (RCD), presents an emerging field of inquiry. Currently, investigations into disulfidoptosis are in their initial stages. Our undertaking sought to integrate single-cell RNA sequencing (scRNA-seq) in conjunction with traditional bulk RNA sequencing (bulk RNA-seq) methodologies, with the objective of delineating genes associated with disulfidoptosis and subsequently prognosticating the clinical outcomes of LUAD patients. Methods Initially, we conducted an in-depth examination of the cellular composition disparities existing between LUAD and normal samples using scRNA-seq data sourced from GSE149655. Simultaneously, we scrutinized the expression patterns of disulfidoptosis-associated gene sets across diverse cell types. Subsequently, leveraging the bulk RNA-seq data, we formulated disulfidoptosis-related prognostic risk signatures (DRPS) employing LASSO-Cox regression. This was accomplished by focusing on genes implicated in disulfidoptosis that exhibited differential expression within endothelial cells (ECs). Sequentially, the robustness and precision of the DRPS model were rigorously verified through both internal and external validation datasets. In parallel, we executed single-cell trajectory analysis to delve into the differentiation dynamics of ECs. Concluding our study, we undertook a comprehensive investigation encompassing various facets. These included comparative assessments of enrichment pathways, clinicopathological parameters, immune cell abundance, immune response-associated genes, impacts of immunotherapy, and drug predictions among distinct risk cohorts. Results The scrutiny of scRNA-seq data underscored discernible disparities in cellular composition between LUAD and normal samples. Furthermore, disulfidoptosis-associated genes exhibited marked discrepancies within endothelial cells (ECs). Consequently, we formulated the Disulfidoptosis-Related Prognostic Signature (DRPS) to facilitate prognostic prediction. The prognostic nomogram based on the risk score effectively demonstrated DRPS's robust capacity to prognosticate survival outcomes. This assertion was corroborated by rigorous assessments utilizing both internal and external validation sets, thus affirming the commendable predictive accuracy and enduring stability of DRPS. Functional enrichment analysis shed light on the significant correlation of DRPS with pathways intrinsic to the cell cycle. Subsequent analysis unveiled correlations between DRPS and gene mutations characteristic of LUAD, as well as indications of an immunosuppressive status. Through drug prediction, we explored potential therapeutic agents for low-risk patients. Concluding our investigation, qRT-PCR experiments confirmed the heightened expression levels of EPHX1, LDHA, SHC1, MYO6, and TLE1 in lung cancer cell lines.
Collapse
Affiliation(s)
- Dabao He
- Department of Laboratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Hengfeng Tang
- Department of Laboratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Xiaoling Yang
- Department of Laboratory Medicine, Shenzhen Baoan District Songgang People’s Hospital, Shenzhen, China
| | - Xiaohong Liu
- Department of Oncology, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Yipeng Zhang
- Department of Laboratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Junzhu Shi
- Department of Laboratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, China
| |
Collapse
|
2
|
Zhang J, Liu X, Huang Z, Wu C, Zhang F, Han A, Stalin A, Lu S, Guo S, Huang J, Liu P, Shi R, Zhai Y, Chen M, Zhou W, Bai M, Wu J. T cell-related prognostic risk model and tumor immune environment modulation in lung adenocarcinoma based on single-cell and bulk RNA sequencing. Comput Biol Med 2023; 152:106460. [PMID: 36565482 DOI: 10.1016/j.compbiomed.2022.106460] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/06/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND T cells are present in all stages of tumor formation and play an important role in the tumor microenvironment. We aimed to explore the expression profile of T cell marker genes, constructed a prognostic risk model based on these genes in Lung adenocarcinoma (LUAD), and investigated the link between this risk model and the immunotherapy response. METHODS We obtained the single-cell sequencing data of LUAD from the literature, and screened out 6 tissue biopsy samples, including 32,108 cells from patients with non-small cell lung cancer, to identify T cell marker genes in LUAD. Combined with TCGA database, a prognostic risk model based on T-cell marker gene was constructed, and the data from GEO database was used for verification. We also investigated the association between this risk model and immunotherapy response. RESULTS Based on scRNA-seq data 1839 T-cell marker genes were identified, after which a risk model consisting of 9 gene signatures for prognosis was constructed in combination with the TCGA dataset. This risk model divided patients into high-risk and low-risk groups based on overall survival. The multivariate analysis demonstrated that the risk model was an independent prognostic factor. Analysis of immune profiles showed that high-risk groups presented discriminative immune-cell infiltrations and immune-suppressive states. Risk scores of the model were closely correlated with Linoleic acid metabolism, intestinal immune network for IgA production and drug metabolism cytochrome P450. CONCLUSION Our study proposed a novel prognostic risk model based on T cell marker genes for LUAD patients. The survival of LUAD patients as well as treatment outcomes may be accurately predicted by the prognostic risk model, and make the high-risk population present different immune cell infiltration and immunosuppression state.
Collapse
Affiliation(s)
- Jingyuan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xinkui Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhihong Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Chao Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Fanqin Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Aiqing Han
- School of Management, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Antony Stalin
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Shan Lu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Siyu Guo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jiaqi Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Pengyun Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Rui Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yiyan Zhai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Meilin Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wei Zhou
- Pharmacy Department, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Meirong Bai
- Key Laboratory of Mongolian Medicine Research and Development Engineering, Ministry of Education, Tongliao, 028000, China.
| | - Jiarui Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
3
|
Hei N, Chen Y, Peng S, Bao Y, Jin L. Circular RNA_0076977 contributes to oral squamous cell carcinoma progression through mediating microRNA-802 axis. Arch Oral Biol 2022; 144:105567. [DOI: 10.1016/j.archoralbio.2022.105567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 11/26/2022]
|
4
|
Feng D, Zhu W, Shi X, Wei W, Han P, Wei Q, Yang L. Leucine zipper protein 2 serves as a prognostic biomarker for prostate cancer correlating with immune infiltration and epigenetic regulation. Heliyon 2022; 8:e10750. [PMID: 36217461 PMCID: PMC9547219 DOI: 10.1016/j.heliyon.2022.e10750] [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: 07/11/2022] [Revised: 08/19/2022] [Accepted: 09/20/2022] [Indexed: 11/04/2022] Open
Abstract
Background We sought to determine whether leucine zipper protein 2 (LUZP2) could benefit men with prostate cancer (PCa) undergoing radical radiotherapy (RT) or prostatectomy (RP). Methods Analysis was done on differentiating expression, clinical prognosis, co-expressed genes, immune infiltration, and epigenetic changes. All of our analyses were done using the R software (version 3.6.3) and the appropriate packages. Results In terms of PCa, tumor samples expressed LUZP2 more than normal samples did. In the TCGA database and GSE116918, we found that LUZP2 was the only independent risk factor for PCa. The shared enriched pathways for patients undergoing RP or RT were cell-cell adhesion, regulation of filopodium assembly, and extracellular matrix containing collagen. With the exception of TNFRSF14, we discovered that LUZP2 was negatively correlated with 21 immune checkpoints in PCa patients receiving RT. We found a significant inverse relationship between LUZP2 expression and the tumor immune environment, which included B cells, CD4+ T cells, neutrophils, macrophages, dendritic cells, stromal score, immune score, and estimate score, in patients receiving RP or RT. Additionally, tumor purity was positively correlated with LUZP2. We found that the drug bortezomib may be susceptible to the LUZP2. DNA methylation was significantly associated with the mRNA expression of LUZP2 in PCa patients from the TCGA database, and LUZP2 methylation was positively correlated with immune cells. The proliferative activity of various PCa cells, which correlated to different stages of this disease, was also found to be significantly reduced by LUZP2 reduction, according to the results of our experimental work. Conclusions We proposed a relatively comprehensive understanding of the roles of LUZP2 on PCa from the fresh perspective of senescence.
Collapse
|
5
|
Zhang L, Yu R, Li C, Dang Y, Yi X, Wang L. Circ_0026416 downregulation blocks the development of colorectal cancer through depleting MYO6 expression by enriching miR-545-3p. World J Surg Oncol 2021; 19:299. [PMID: 34645476 PMCID: PMC8515727 DOI: 10.1186/s12957-021-02407-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/21/2021] [Indexed: 12/14/2022] Open
Abstract
Background Emerging evidence reveals that the initiation and development of human cancers, including colorectal cancer (CRC), are associated with the deregulation of circular RNAs (circRNAs). Our study intended to disclose the role of circ_0026416 in the malignant behaviors of CRC. Methods The detection for circ_0026416 expression, miR-545-3p expression, and myosin VI (MYO6) mRNA expression was performed using quantitative real-time PCR (qPCR). CCK-8 assay, colony formation assay, transwell assay, and flow cytometry assay were applied for functional analysis to monitor cell proliferation, migration, invasion, and apoptosis. The protein levels of MYO6 and epithelial mesenchymal-transition (EMT) markers were detected by western blot. Mouse models were used to determine the role of circ_0026416 in vivo. The potential relationship between miR-545-3p and circ_0026416 or MYO6 was verified by dual-luciferase reporter assay and RIP assay. Results The expression of circ_0026416 was increased in CRC tumor tissues and cell lines. Circ_0026416 downregulation inhibited CRC cell proliferation, colony formation, migration, invasion, and EMT but induced cell apoptosis in vitro, and circ_0026416 knockdown also blocked tumor growth in vivo. MiR-545-3p was a target of circ_0026416, and rescue experiments indicated that circ_0026416 knockdown blocked CRC development by enriching miR-545-3p. In addition, miR-545-3p targeted MYO6 and inhibited MYO6 expression. MiR-545-3p enrichment suppressed CRC cell malignant behaviors by sequestering MYO6. Importantly, circ_0026416 knockdown depleted MYO6 expression by enriching miR-545-3p. Conclusion Circ_0026416 downregulation blocked the development of CRC through depleting MYO6 expression by enriching miR-545-3p. Highlights Circ_0026416 downregulation inhibits CRC development in vitro and in vivo. Circ_0026416 regulates the expression of MYO6 by targeting miR-545-3p. Circ_0026416 governs the miR-545-3p/MYO6 axis to regulate CRC progression.
Supplementary Information The online version contains supplementary material available at 10.1186/s12957-021-02407-y.
Collapse
Affiliation(s)
- Lei Zhang
- Department of General Surgery, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingba Road, Shizhong District, Jinan City, 250001, Shandong Province, China
| | - Ranran Yu
- Department of Pathology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingba Road, Shizhong District, Jinan City, 250001, Shandong Province, China
| | - Chunhua Li
- Department of General Surgery, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingba Road, Shizhong District, Jinan City, 250001, Shandong Province, China
| | - Yu Dang
- Department of General Surgery, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingba Road, Shizhong District, Jinan City, 250001, Shandong Province, China
| | - Xiaoyu Yi
- Department of General Surgery, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingba Road, Shizhong District, Jinan City, 250001, Shandong Province, China
| | - Lei Wang
- Cancer Center, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No.1 Jingba Road, Shizhong District, Jinan City, 250001, Shandong Province, China.
| |
Collapse
|
6
|
Briolay T, Petithomme T, Fouet M, Nguyen-Pham N, Blanquart C, Boisgerault N. Delivery of cancer therapies by synthetic and bio-inspired nanovectors. Mol Cancer 2021; 20:55. [PMID: 33761944 PMCID: PMC7987750 DOI: 10.1186/s12943-021-01346-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/05/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND As a complement to the clinical development of new anticancer molecules, innovations in therapeutic vectorization aim at solving issues related to tumor specificity and associated toxicities. Nanomedicine is a rapidly evolving field that offers various solutions to increase clinical efficacy and safety. MAIN: Here are presented the recent advances for different types of nanovectors of chemical and biological nature, to identify the best suited for translational research projects. These nanovectors include different types of chemically engineered nanoparticles that now come in many different flavors of 'smart' drug delivery systems. Alternatives with enhanced biocompatibility and a better adaptability to new types of therapeutic molecules are the cell-derived extracellular vesicles and micro-organism-derived oncolytic viruses, virus-like particles and bacterial minicells. In the first part of the review, we describe their main physical, chemical and biological properties and their potential for personalized modifications. The second part focuses on presenting the recent literature on the use of the different families of nanovectors to deliver anticancer molecules for chemotherapy, radiotherapy, nucleic acid-based therapy, modulation of the tumor microenvironment and immunotherapy. CONCLUSION This review will help the readers to better appreciate the complexity of available nanovectors and to identify the most fitting "type" for efficient and specific delivery of diverse anticancer therapies.
Collapse
Affiliation(s)
- Tina Briolay
- Université de Nantes, Inserm, CRCINA, F-44000, Nantes, France
| | | | - Morgane Fouet
- Université de Nantes, Inserm, CRCINA, F-44000, Nantes, France
| | | | | | | |
Collapse
|
7
|
The Expressions and Mechanisms of Sarcomeric Proteins in Cancers. DISEASE MARKERS 2020; 2020:8885286. [PMID: 32670437 PMCID: PMC7346232 DOI: 10.1155/2020/8885286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/07/2020] [Accepted: 06/13/2020] [Indexed: 02/07/2023]
Abstract
The sarcomeric proteins control the movement of cells in diverse species, whereas the deregulation can induce tumours in model organisms and occurs in human carcinomas. Sarcomeric proteins are recognized as oncogene and related to tumor cell metastasis. Recent insights into their expressions and functions have led to new cancer therapeutic opportunities. In this review, we appraise the evidence for the sarcomeric proteins as cancer genes and discuss cancer-relevant biological functions, potential mechanisms by which sarcomeric proteins activity is altered in cancer.
Collapse
|
8
|
Duan Q, Xu M, Wu M, Zhang X, Gan M, Jiang H. Long noncoding RNA UCA1 promotes cell growth, migration, and invasion by targeting miR-143-3p in oral squamous cell carcinoma. Cancer Med 2020; 9:3115-3129. [PMID: 32130788 PMCID: PMC7196043 DOI: 10.1002/cam4.2808] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/10/2019] [Accepted: 12/14/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The long noncoding RNA (lncRNA) urothelial carcinoma-associated 1 (UCA1) is dysregulated in many types of tumors; however, its role in oral squamous cell carcinoma (OSCC) remains unclear. This study aims to determine the effect of lncRNA UCA1 on OSCC. METHODS Fifty-six paired OSCC and adjacent nontumorous tissues were collected and the levels of UCA1, miR-143-3p, and MYO6 in the tissues were evaluated by qRT-PCR. In in vitro experiments, cell viability, migration, and invasion were measured by, respectively, performing CCK-8, wound healing, and transwell assays. The target relationships among UCA1, miR-143-3p, and MYO6 were verified by dual-luciferase assay. Western blot and immunohistochemistry were carried out to determine the protein levels. Xenograft mouse model was established to explore the effects of UCA1 in vivo. RESULTS Levels of UCA1 and MYO6 were increased significantly in OSCC, while the level of miR-143-3p was decreased compared with the adjacent nontumorous tissues. UCA1 promoted OSCC cell growth, migration, and invasion both in vitro and in vivo, while miR-143-3p reversed the progression. MYO6 was validated as a target for miR-143-3p, and MYO6 overexpression reversed the effects of miR-143-3p mimic on OSCC cells. CONCLUSION LncRNA UCA1 contributes to the proliferation and metastasis of OSCC cells by targeting miR-143-3p and upregulating its downstream gene MYO6. UCA1 could serve as a promising novel target therapy for treatment of OSCC.
Collapse
Affiliation(s)
- Qingyun Duan
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical UniversityNanjingJiangsuChina
- Department of Oral and Maxillofacial SurgeryAffiliated Hangzhou First People's HospitalMedical College of Zhejiang UniversityHangzhouZhejiangChina
| | - Mei Xu
- Department of OphtalmologyHangzhou Jianggan District People's HospitalHangzhouZhejiangChina
| | - Meng Wu
- Department of Oral and Maxillofacial SurgeryThe Affiliated Huaian No.1 People's Hospital of Nanjing Medical UniversityHuaianJiangsuChina
| | - Xiong Zhang
- Department of Oral and Maxillofacial SurgeryAffiliated Hangzhou First People's HospitalMedical College of Zhejiang UniversityHangzhouZhejiangChina
| | - Min Gan
- Department of Oral and Maxillofacial SurgeryAffiliated Hangzhou First People's HospitalMedical College of Zhejiang UniversityHangzhouZhejiangChina
| | - Hongbing Jiang
- Jiangsu Key Laboratory of Oral DiseasesNanjing Medical UniversityNanjingJiangsuChina
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| |
Collapse
|
9
|
Labbé C, Grima N, Gautier T, Favier B, Byrne JA. Semi-automated fact-checking of nucleotide sequence reagents in biomedical research publications: The Seek & Blastn tool. PLoS One 2019; 14:e0213266. [PMID: 30822319 PMCID: PMC6396917 DOI: 10.1371/journal.pone.0213266] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 02/18/2019] [Indexed: 12/14/2022] Open
Abstract
Nucleotide sequence reagents are verifiable experimental reagents in biomedical publications, because their sequence identities can be independently verified and compared with associated text descriptors. We have previously reported that incorrectly identified nucleotide sequence reagents are characteristic of highly similar human gene knockdown studies, some of which have been retracted from the literature on account of possible research fraud. Because of the throughput limitations of manual verification of nucleotide sequences, we developed a semi-automated fact checking tool, Seek & Blastn, to verify the targeting or non-targeting status of published nucleotide sequence reagents. From previously described and unknown corpora of 48 and 155 publications, respectively, Seek & Blastn correctly extracted 304/342 (88.9%) and 1066/1522 (70.0%) nucleotide sequences and a predicted targeting/ non-targeting status. Seek & Blastn correctly predicted the targeting/ non-targeting status of 293/304 (96.4%) and 988/1066 (92.7%) of the correctly extracted nucleotide sequences. A total of 38/39 (97.4%) or 31/79 (39.2%) Seek & Blastn predictions of incorrect nucleotide sequence reagent use were correct in the two literature corpora. Combined Seek & Blastn and manual analyses identified a list of 91 misidentified nucleotide sequence reagents, which could be built upon through future studies. In summary, incorrect nucleotide sequence reagents represent an under-recognized source of error within the biomedical literature, and fact checking tools such as Seek & Blastn may help to identify papers and manuscripts affected by these errors.
Collapse
Affiliation(s)
- Cyril Labbé
- Univ. Grenoble Alpes, CNRS, Grenoble INP, LIG, Grenoble, France
| | - Natalie Grima
- Molecular Oncology Laboratory, Children’s Cancer Research Unit, Kids Research, The Children’s Hospital at Westmead, Westmead, New South Wales, Australia
| | - Thierry Gautier
- INSERM U1209/ CNRS UMR 5309, Univ. Grenoble Alpes, Grenoble, France
| | - Bertrand Favier
- Univ. Grenoble Alpes, Team GREPI, Etablissement Français du Sang, La Tronche, France
| | - Jennifer A. Byrne
- Molecular Oncology Laboratory, Children’s Cancer Research Unit, Kids Research, The Children’s Hospital at Westmead, Westmead, New South Wales, Australia
- Discipline of Child and Adolescent Health, Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
| |
Collapse
|
10
|
Byrne JA, Grima N, Capes-Davis A, Labbé C. The Possibility of Systematic Research Fraud Targeting Under-Studied Human Genes: Causes, Consequences, and Potential Solutions. Biomark Insights 2019; 14:1177271919829162. [PMID: 30783377 PMCID: PMC6366001 DOI: 10.1177/1177271919829162] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/08/2019] [Indexed: 12/27/2022] Open
Abstract
A major reason for biomarker failure is the selection of candidate biomarkers based on inaccurate or incorrect published results. Incorrect research results leading to the selection of unproductive biomarker candidates are largely considered to stem from unintentional research errors. The additional possibility that biomarker research may be actively misdirected by research fraud has been given comparatively little consideration. This review discusses what we believe to be a new threat to biomarker research, namely, the possible systematic production of fraudulent gene knockdown studies that target under-studied human genes. We describe how fraudulent papers may be produced in series by paper mills using what we have described as a 'theme and variations' model, which could also be considered a form of salami slicing. We describe features of these single-gene knockdown publications that may allow them to evade detection by journal editors, peer reviewers, and readers. We then propose a number of approaches to facilitate their detection, including improved awareness of the features of publications constructed in series, broader requirements to post submitted manuscripts to preprint servers, and the use of semi-automated literature screening tools. These approaches may collectively improve the detection of fraudulent studies that might otherwise impede future biomarker research.
Collapse
Affiliation(s)
- Jennifer A Byrne
- Molecular Oncology Laboratory, Children’s Cancer Research Unit, Kids Research, The Children’s Hospital at Westmead, Westmead, NSW, Australia
- Discipline of Child and Adolescent Health, The University of Sydney and The Children’s Hospital at Westmead, Westmead, NSW, Australia
| | - Natalie Grima
- Molecular Oncology Laboratory, Children’s Cancer Research Unit, Kids Research, The Children’s Hospital at Westmead, Westmead, NSW, Australia
| | - Amanda Capes-Davis
- CellBank Australia, Children’s Medical Research Institute and The University of Sydney, Westmead, NSW, Australia
| | - Cyril Labbé
- Univ Grenoble Alpes, CNRS, Grenoble INP, LIG, Grenoble, France
| |
Collapse
|
11
|
Recombinant Viruses for Cancer Therapy. Biomedicines 2018; 6:biomedicines6040094. [PMID: 30257488 PMCID: PMC6316473 DOI: 10.3390/biomedicines6040094] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/17/2018] [Accepted: 09/20/2018] [Indexed: 12/28/2022] Open
Abstract
Recombinant viruses are novel therapeutic agents that can be utilized for treatment of various diseases, including cancers. Recombinant viruses can be engineered to express foreign transgenes and have a broad tropism allowing gene expression in a wide range of host cells. They can be selected or designed for specific therapeutic goals; for example, recombinant viruses could be used to stimulate host immune response against tumor-specific antigens and therefore overcome the ability of the tumor to evade the host's immune surveillance. Alternatively, recombinant viruses could express immunomodulatory genes which stimulate an anti-cancer immune response. Oncolytic viruses can replicate specifically in tumor cells and induce toxic effects leading to cell lysis and apoptosis. However, each of these approaches face certain difficulties that must be resolved to achieve maximum therapeutic efficacy. In this review we discuss actively developing approaches for cancer therapy based on recombinant viruses, problems that need to be overcome, and possible prospects for further development of recombinant virus based therapy.
Collapse
|
12
|
miR-143 and miR-145 inhibit gastric cancer cell migration and metastasis by suppressing MYO6. Cell Death Dis 2017; 8:e3101. [PMID: 29022908 PMCID: PMC5682659 DOI: 10.1038/cddis.2017.493] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/26/2017] [Accepted: 08/28/2017] [Indexed: 01/07/2023]
Abstract
Metastasis is a major clinical obstacle responsible for the high mortality and poor prognosis of gastric cancer (GC). MicroRNAs (miRNAs) are critical mediators of metastasis that act by modulating their target genes. In this study, we found that miR-143 and miR-145 act via a common target gene, MYO6, to regulate the epithelial–mesenchymal transition (EMT) and inhibit metastasis. We determined that miR-143 and miR-145 were downregulated in GC, and the ectopic expression of miR-143 and/or miR-145 inhibited GC cell migration and metastasis. Furthermore, MYO6 was identified as a direct common target of miR-143 and miR-145 and was elevated in GC. Silencing of MYO6 resulted in a metastasis-suppressive activity similar to that of miR-143 and miR-145, while restoring MYO6 attenuated the anti-metastatic or anti-EMT effects caused by miR-143 and miR-145. Clinically, an inverse correlation was observed between miR-143/145 levels and MYO6 levels in GC tissues, and either miR-143/145 downregulation or MYO6 upregulation was associated with more malignant phenotypes in patients with GC. In conclusion, miR-143 and miR-145 suppress GC cell migration and metastasis by inhibiting MYO6 expression and the EMT, which provides a novel mechanism and promising therapeutic target for the treatment of GC metastasis.
Collapse
|
13
|
How myosin organization of the actin cytoskeleton contributes to the cancer phenotype. Biochem Soc Trans 2017; 44:1026-34. [PMID: 27528748 DOI: 10.1042/bst20160034] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Indexed: 12/29/2022]
Abstract
The human genome contains 39 genes that encode myosin heavy chains, classified on the basis of their sequence similarity into 12 classes. Most cells express at least 12 different genes, from at least 8 different classes, which are typically composed of several class 1 genes, at least one class 2 gene and classes 5, 6, 9, 10, 18 and 19. Although the different myosin isoforms all have specific and non-overlapping roles in the cell, in combination they all contribute to the organization of the actin cytoskeleton, and the shape and phenotype of the cell. Over (or under) expression of these different myosin isoforms can have strong effects on actin organization, cell shape and contribute to the cancer phenotype as discussed in this review.
Collapse
|
14
|
Wang Z, Ying M, Wu Q, Wang R, Li Y. Overexpression of myosin VI regulates gastric cancer cell progression. Gene 2016; 593:100-109. [PMID: 27515005 DOI: 10.1016/j.gene.2016.08.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/20/2016] [Accepted: 08/06/2016] [Indexed: 10/21/2022]
Abstract
Myosin VI (MYO6) is a unique member of the myosin superfamily. Although it has been reported to participate in human cancer progression, the role of MYO6 in gastric cancer remains unclear. In this study, we found the expression of MYO6 gene was higher in gastric cancer tissues than in the normal tissues by Oncomine database mining and affects patient overall survival using the Kaplan-Meier plotter online analysis. Additionally, the expression levels of MYO6 were widely expressed in gastric cancer cells by quantitative real-time Polymerase Chain Reaction (qRT-PCR) and western blot assay. Then knockdown of MYO6 significantly suppressed the proliferation and colony formation abilities of AGS and MGC80-3 cells. Moreover, cell cycle analysis showed that inhibition of MYO6 induced cell cycle arrested in G0/G1 phase in AGS and MGC80-3 cells. Further analysis showed knockdown of MYO6 downregulated cell-cycle activators cyclin A and cyclin D1 and upregulated cell-cycle inhibitor p21, as determined by qRT-PCR and western blot analysis in MGC80-3 cells. Meanwhile, MYO6 inhibition significantly induced apoptosis in AGS and MGC80-3 cells. Also, knockdown of MYO6 increased the expression of apoptosis-related proteins Bax and cleaved Caspase-3, and decreased Bcl-2 expression by western blot analysis in MGC80-3 cells. In addition, MYO6 knockdown also inhibited cell migration ability in MGC80-3 cells. Taken together, our study indicates that MYO6 may play an important role in gastric cancer tumorigenesis and may serve as a potential therapeutic target in human gastric cancer.
Collapse
Affiliation(s)
- Zishu Wang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, China.
| | - Mingzhen Ying
- Department of Oncology, Changhai Hospital, The Second Military Medical University, Shanghai 200433, China
| | - Qiong Wu
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, China.
| | - Rui Wang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, China
| | - Yumei Li
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, China
| |
Collapse
|
15
|
You W, Tan G, Sheng N, Gong J, Yan J, Chen D, Zhang H, Wang Z. Downregulation of myosin VI reduced cell growth and increased apoptosis in human colorectal cancer. Acta Biochim Biophys Sin (Shanghai) 2016; 48:430-6. [PMID: 27044563 DOI: 10.1093/abbs/gmw020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 02/25/2016] [Indexed: 12/19/2022] Open
Abstract
Colorectal cancer (CRC) is the third most commonly diagnosed cancer worldwide, with the mortality increasing steadily over the last decade. Myosin VI (MYO6) expression is found to be elevated in some types of human carcinoma cell types, suggesting that it may be a sensitive biomarker for the diagnosis and follow-up. In this study, we first used the Oncomine database to explore the expression of MYO6 in CRC tissues, and then constructed a plasmid of RNA interference targeting MYO6 gene. After transfection of lentivirus targeting MYO6 into SW1116 cells, cell viability and proliferation were measured with 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and colony formation assay. Cell cycle distribution was assayed by flow cytometry and apoptosis was evaluated by Annexin V. MYO6 expression was detected by quantitative real-time polymerase chain reaction and western blot analysis. It was found that MYO6 mRNA was upregulated in CRC tissues using data mining of public Oncomine microarray datasets. Depletion of MYO6 significantly inhibited cell proliferation and colony formation. In addition, knockdown of MYO6 slightly arrested cell cycle in G0/G1 phase, but remarkably increased the proportion of the sub-G1 phase of cell with the increase of apoptotic cells. These results suggest that MYO6 may promote cell growth and may be used as a potential target for anticancer therapy of CRC.
Collapse
Affiliation(s)
- Weiqiang You
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Gewen Tan
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Nengquan Sheng
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Jianfeng Gong
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Jun Yan
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Di Chen
- National Key Laboratory of Science and Technology on Nano/Micro Fabrication Technology, Research Institute Micro/Nano Science and Technology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Huizhen Zhang
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Zhigang Wang
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|