101
|
Kuijpers TJM, Kleinjans JCS, Jennen DGJ. From multi-omics integration towards novel genomic interaction networks to identify key cancer cell line characteristics. Sci Rep 2021; 11:10542. [PMID: 34006939 PMCID: PMC8131752 DOI: 10.1038/s41598-021-90047-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 04/26/2021] [Indexed: 11/09/2022] Open
Abstract
Cancer is a complex disease where cancer cells express epigenetic and transcriptomic mechanisms to promote tumor initiation, progression, and survival. To extract relevant features from the 2019 Cancer Cell Line Encyclopedia (CCLE), a multi-layer nonnegative matrix factorization approach is used. We used relevant feature genes and DNA promoter regions to construct genomic interaction network to study gene-gene and gene-DNA promoter methylation relationships. Here, we identified a set of gene transcripts and methylated DNA promoter regions for different clusters, including one homogeneous lymphoid neoplasms cluster. In this cluster, we found different methylated transcription factors that affect transcriptional activation of EGFR and downstream interactions. Furthermore, the hippo-signaling pathway might not function properly because of DNA hypermethylation and low gene expression of both LATS2 and YAP1. Finally, we could identify a potential dysregulation of the CD28-CD86-CTLA4 axis. Characterizing the interaction of the epigenome and the transcriptome is vital for our understanding of cancer cell line behavior, not only for deepening insights into cancer-related processes but also for future disease treatment and drug development. Here we have identified potential candidates that characterize cancer cell lines, which give insight into the development and progression of cancers.
Collapse
Affiliation(s)
- T J M Kuijpers
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands.
| | - J C S Kleinjans
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands
| | - D G J Jennen
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, the Netherlands
| |
Collapse
|
102
|
Abstract
Significance: Genomic instability, a hallmark of cancer, renders cancer cells susceptible to genomic stress from both endogenous and exogenous origins, resulting in the increased tendency to accrue DNA damage, chromosomal instability, or aberrant DNA localization. Apart from the cell autonomous tumor-promoting effects, genomic stress in cancer cells could have a profound impact on the tumor microenvironment. Recent Advances: Recently, it is increasingly appreciated that harnessing genomic stress could provide a promising strategy to revive antitumor immunity, and thereby offer new therapeutic opportunities in cancer treatment. Critical Issues: Genomic stress is closely intertwined with antitumor immunity via mechanisms involving the direct crosstalk with DNA damage response components, upregulation of immune-stimulatory/inhibitory ligands, release of damage-associated molecular patterns, increase of neoantigen repertoire, and activation of DNA sensing pathways. A better understanding of these mechanisms will provide molecular basis for exploiting the genomic stress to boost antitumor immunity. Future Directions: Future research should pay attention to the heterogeneity between individual cancers in the genomic instability and the associated immune response, and how to balance the toxicity and benefit by specifying the types, potency, and treatment sequence of genomic stress inducer in therapeutic practice. Antioxid. Redox Signal. 34, 1128-1150.
Collapse
Affiliation(s)
- Congying Pu
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Siyao Tao
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jun Xu
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Min Huang
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
103
|
Arora R, Malla WA, Tyagi A, Mahajan S, Sajjanar B, Tiwari AK. Canine Parvovirus and Its Non-Structural Gene 1 as Oncolytic Agents: Mechanism of Action and Induction of Anti-Tumor Immune Response. Front Oncol 2021; 11:648873. [PMID: 34012915 PMCID: PMC8127782 DOI: 10.3389/fonc.2021.648873] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/30/2021] [Indexed: 12/15/2022] Open
Abstract
The exploration into the strategies for the prevention and treatment of cancer is far from complete. Apart from humans, cancer has gained considerable importance in animals because of increased awareness towards animal health and welfare. Current cancer treatment regimens are less specific towards tumor cells and end up harming normal healthy cells. Thus, a highly specific therapeutic strategy with minimal side effects is the need of the hour. Oncolytic viral gene therapy is one such specific approach to target cancer cells without affecting the normal cells of the body. Canine parvovirus (CPV) is an oncolytic virus that specifically targets and kills cancer cells by causing DNA damage, caspase activation, and mitochondrial damage. Non-structural gene 1 (NS1) of CPV, involved in viral DNA replication is a key mediator of cytotoxicity of CPV and can selectively cause tumor cell lysis. In this review, we discuss the oncolytic properties of Canine Parvovirus (CPV or CPV2), the structure of the NS1 protein, the mechanism of oncolytic action as well as role in inducing an antitumor immune response in different tumor models.
Collapse
Affiliation(s)
- Richa Arora
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Waseem Akram Malla
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Arpit Tyagi
- GB Pant University of Agriculture and Technology, Pantnagar, India
| | - Sonalika Mahajan
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Basavaraj Sajjanar
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Izatnagar, India
| | - Ashok Kumar Tiwari
- Division of Biological Standardisation, ICAR-Indian Veterinary Research Institute, Izatnagar, India.,ICAR - Central Avian Research Institute, Izatnagar, India
| |
Collapse
|
104
|
Targeting nutrient metabolism with FDA-approved drugs for cancer chemoprevention: Drugs and mechanisms. Cancer Lett 2021; 510:1-12. [PMID: 33857528 DOI: 10.1016/j.canlet.2021.03.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/21/2021] [Accepted: 03/30/2021] [Indexed: 12/14/2022]
Abstract
Proliferating cancer cells exhibit metabolic alterations and specific nutritional needs for adapting to their rapid growth. These changes include using aerobic glycolysis, lipid metabolic disorder, and irregular protein degradation. It may be useful to target metabolic abnormalities for cancer chemoprevention. Epidemiological and mechanism-related studies have indicated that many FDA-approved anti-metabolic drugs decrease tumor risk, inhibit tumor growth, or enhance the effect of chemotherapeutic drugs. Drugs targeting nutrient metabolism have fewer side effects with long-term use compared to chemotherapeutic drugs. The characteristics of these drugs make them promising candidates for cancer chemoprevention. Here, we summarize recent discoveries of the chemo-preventive effects of drugs targeting nutrient metabolic pathways and discuss future applications and challenges. Understanding the effects and mechanisms of anti-metabolic drugs in cancer has important implications for exploring strategies for cancer chemoprevention.
Collapse
|
105
|
Sharmin S, Zohura FT, Islam MS, Shimonty A, Khan MAAK, Parveen R, Sharmin F, Ahsan CR, Islam ABMMK, Yasmin M. Mutational profiles of marker genes of cervical carcinoma in Bangladeshi patients. BMC Cancer 2021; 21:289. [PMID: 33736612 PMCID: PMC7977314 DOI: 10.1186/s12885-021-07906-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 02/12/2021] [Indexed: 01/22/2023] Open
Abstract
Background Cervical cancer is a gynecologic cancer type that develops in the cervix, accounting for 8% mortality of all female cancer patients. Infection with specific human papillomavirus (HPV) types is considered the most severe risk factor for cervical cancer. In the context of our socioeconomic conditions, an increasing burden of this disease and high mortality rate prevail in Bangladesh. Although several researches related to the epidemiology, HPV vaccination, and treatment modalities were conducted, researches on the mutation profiles of marker genes in cervical cancer in Bangladesh remain unexplored. Methods In this study, five different genomic regions within the top three most frequently mutated genes (EGFR, KRAS and PIK3CA) in COSMIC database with a key role in the development of cervical cancers were selected to study the mutation frequency in Bangladeshi patients. In silico analysis was done in two steps: nucleotide sequence analysis and its corresponding amino acid analysis. Results DNA from 46 cervical cancer tissue samples were extracted and amplified by PCR, using 1 set of primers designed for EGFR and 2 sets of primers designed for two different regions of both PIK3CA and KRAS gene. In total, 39 mutations were found in 26 patient samples. Eleven different mutations (23.91%), twenty-four different mutations (52.17%) and four mutations (8.7%) were found in amplified EGFR, PIK3CA and KRAS gene fragments, respectively; among which 1 (EGFR) was common in seven patient samples and 2 (PIKCA) were found in more than 1 patient. Our study shows that except for KRAS, the frequency of observed mutations in our patients is higher than those reported earlier in other parts of the world. Most of the exonic mutations were found only in the PIK3CA and EGFR genes. Conclusions The study can be used as a basis to build a mutation database for cervical cancer in Bangladesh with the possibility of targetable oncogenic mutations. Further explorations are needed to establish future diagnostics, personalized medicine decisions, and other pharmaceutical applications for specific cancer subtypes. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-07906-5.
Collapse
Affiliation(s)
- Shahana Sharmin
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
| | - Fatima Tuj Zohura
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka, Bangladesh.,Current Affiliation: Internal Medicine OPD, Bangabandhu Sheikh Mujib Medical University Hospital, Dhaka, Bangladesh
| | - Md Sajedul Islam
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka, Bangladesh.,Current Affiliation: Department of Biochemistry and Biotechnology, University of Barisal, Barisal, Bangladesh
| | - Anika Shimonty
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
| | - Md Abdullah-Al-Kamran Khan
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka, Bangladesh.,Current Affiliation Department of Mathematics and Natural Sciences, BRAC University, Dhaka, Bangladesh
| | | | - Foujia Sharmin
- Department of Gynecological Oncology, National Institute of Cancer Research & Hospital, Dhaka, Bangladesh
| | | | | | - Mahmuda Yasmin
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh.
| |
Collapse
|
106
|
Łukasik P, Załuski M, Gutowska I. Cyclin-Dependent Kinases (CDK) and Their Role in Diseases Development-Review. Int J Mol Sci 2021; 22:ijms22062935. [PMID: 33805800 PMCID: PMC7998717 DOI: 10.3390/ijms22062935] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/07/2021] [Accepted: 03/09/2021] [Indexed: 12/13/2022] Open
Abstract
Cyclin-dependent kinases (CDKs) are involved in many crucial processes, such as cell cycle and transcription, as well as communication, metabolism, and apoptosis. The kinases are organized in a pathway to ensure that, during cell division, each cell accurately replicates its DNA, and ensure its segregation equally between the two daughter cells. Deregulation of any of the stages of the cell cycle or transcription leads to apoptosis but, if uncorrected, can result in a series of diseases, such as cancer, neurodegenerative diseases (Alzheimer’s or Parkinson’s disease), and stroke. This review presents the current state of knowledge about the characteristics of cyclin-dependent kinases as potential pharmacological targets.
Collapse
Affiliation(s)
- Paweł Łukasik
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstancow Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Michał Załuski
- Department of Pharmaceutical Chemistry, Pomeranian Medical University in Szczecin, Powstancow Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstancow Wlkp. 72 Av., 70-111 Szczecin, Poland;
- Correspondence:
| |
Collapse
|
107
|
Kalhori MR, Khodayari H, Khodayari S, Vesovic M, Jackson G, Farzaei MH, Bishayee A. Regulation of Long Non-Coding RNAs by Plant Secondary Metabolites: A Novel Anticancer Therapeutic Approach. Cancers (Basel) 2021; 13:cancers13061274. [PMID: 33805687 PMCID: PMC8001769 DOI: 10.3390/cancers13061274] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Cancer is caused by the rapid and uncontrolled growth of cells that eventually lead to tumor formation. Genetic and epigenetic alterations are among the most critical factors in the onset of carcinoma. Phytochemicals are a group of natural compounds that play an essential role in cancer prevention and treatment. Long non-coding RNAs (lncRNAs) are potential therapeutic targets of bioactive phytochemicals, and these compounds could regulate the expression of lncRNAs directly and indirectly. Here, we critically evaluate in vitro and in vivo anticancer effects of phytochemicals in numerous human cancers via regulation of lncRNA expression and their downstream target genes. Abstract Long non-coding RNAs (lncRNAs) are a class of non-coding RNAs that play an essential role in various cellular activities, such as differentiation, proliferation, and apoptosis. Dysregulation of lncRNAs serves a fundamental role in the progression and initiation of various diseases, including cancer. Precision medicine is a suitable and optimal treatment method for cancer so that based on each patient’s genetic content, a specific treatment or drug is prescribed. The rapid advancement of science and technology in recent years has led to many successes in this particular treatment. Phytochemicals are a group of natural compounds extracted from fruits, vegetables, and plants. Through the downregulation of oncogenic lncRNAs or upregulation of tumor suppressor lncRNAs, these bioactive compounds can inhibit metastasis, proliferation, invasion, migration, and cancer cells. These natural products can be a novel and alternative strategy for cancer treatment and improve tumor cells’ sensitivity to standard adjuvant therapies. This review will discuss the antineoplastic effects of bioactive plant secondary metabolites (phytochemicals) via regulation of expression of lncRNAs in various human cancers and their potential for the treatment and prevention of human cancers.
Collapse
Affiliation(s)
- Mohammad Reza Kalhori
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran;
| | - Hamid Khodayari
- International Center for Personalized Medicine, 40235 Düsseldorf, Germany; (H.K.); (S.K.)
- Breast Disease Research Center, Tehran University of Medical Sciences, Tehran 1419733141, Iran
| | - Saeed Khodayari
- International Center for Personalized Medicine, 40235 Düsseldorf, Germany; (H.K.); (S.K.)
- Breast Disease Research Center, Tehran University of Medical Sciences, Tehran 1419733141, Iran
| | - Miko Vesovic
- Department of Mathematics, Statistics, and Computer Science, University of Illinois at Chicago, Chicago, IL 60607, USA;
| | - Gloria Jackson
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
| | - Mohammad Hosein Farzaei
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6718874414, Iran
- Correspondence: (M.H.F.); or (A.B.)
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
- Correspondence: (M.H.F.); or (A.B.)
| |
Collapse
|
108
|
Vaghari-Tabari M, Ferns GA, Qujeq D, Andevari AN, Sabahi Z, Moein S. Signaling, metabolism, and cancer: An important relationship for therapeutic intervention. J Cell Physiol 2021; 236:5512-5532. [PMID: 33580511 DOI: 10.1002/jcp.30276] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 11/05/2022]
Abstract
In cancerous cells, significant changes occur in the activity of signaling pathways affecting a wide range of cellular activities ranging from growth and proliferation to apoptosis, invasiveness, and metastasis. Extensive changes also happen with respect to the metabolism of a cancerous cell encompassing a wide range of functions that include: nutrient acquisition, biosynthesis of macromolecules, and energy generation. These changes are important and some therapeutic approaches for treating cancers have focused on targeting the metabolism of cancerous cells. Oncogenes and tumor suppressor genes have a significant effect on the metabolism of cells. There appears to be a close interaction between metabolism and the signaling pathways in a cancerous cell, in which the interaction provides the metabolic needs of a cancerous cell for uncontrolled proliferation, resistance to apoptosis, and metastasis. In this review, we have reviewed the latest findings in this regard and briefly review the most recent research findings regarding targeting the metabolism of cancer cells as a therapeutic approach for treatment of cancer.
Collapse
Affiliation(s)
- Mostafa Vaghari-Tabari
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gordon A Ferns
- Department of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, Sussex, UK
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | - Ali Nosrati Andevari
- Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Zahra Sabahi
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soheila Moein
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
109
|
Zhang R, Li Q, Fu J, Jin Z, Su J, Zhang J, Chen C, Geng Z, Zhang D. Comprehensive analysis of genomic mutation signature and tumor mutation burden for prognosis of intrahepatic cholangiocarcinoma. BMC Cancer 2021; 21:112. [PMID: 33535978 PMCID: PMC7860034 DOI: 10.1186/s12885-021-07788-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 01/04/2021] [Indexed: 12/17/2022] Open
Abstract
Background Intrahepatic cholangiocarcinoma (iCCA) is a highly lethal malignancy of the biliary tract. Analysis of somatic mutational profiling can reveal new prognostic markers and actionable treatment targets. In this study, we explored the utility of genomic mutation signature and tumor mutation burden (TMB) in predicting prognosis in iCCA patients. Methods Whole-exome sequencing and corresponding clinical data were collected from the ICGC portal and cBioPortal database to detect the prognostic mutated genes and determine TMB values. To identify the hub prognostic mutant signature, we used Cox regression and Lasso feature selection. Mutation-related signature (MRS) was constructed using multivariate Cox regression. The predictive performances of MRS and TMB were assessed using Kaplan–Meier (KM) analysis and receiver operating characteristic (ROC). We performed a functional enrichment pathway analysis using gene set enrichment analysis (GSEA) for mutated genes. Based on the MRS, TMB, and the TNM stage, a nomogram was constructed to visualize prognosis in iCCA patients. Results The mutation landscape illustrated distributions of mutation frequencies and types in iCCA, and generated a list of most frequently mutated genes (such as Tp53, KRAS, ARID1A, and IDH1). Thirty-two mutated genes associated with overall survival (OS) were identified in iCCA patients. We obtained a six-gene signature using the Lasso and Cox method. AUCs for the MRS in the prediction of 1-, 3-, and 5-year OS were 0.759, 0.732, and 0.728, respectively. Kaplan–Meier analysis showed a significant difference in prognosis for patients with iCCA having a high and low MRS score (P < 0.001). GSEA was used to show that several signaling pathways, including MAPK, PI3K-AKT, and proteoglycan, were involved in cancer. Conversely, survival analysis indicated that TMB was significantly associated with prognosis. GSEA indicated that samples with high MRS or TMB also showed an upregulated expression of pathways involved in tumor signaling and the immune response. Finally, the predictive nomogram (that included MRS, TMB, and the TNM stage) demonstrated satisfactory performance in predicting survival in patients with iCCA. Conclusions Mutation-related signature and TMB were associated with prognosis in patients with iCCA. Our study provides a valuable prognostic predictor for determining outcomes in patients with iCCA. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-07788-7.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Qi Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jialu Fu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhechuan Jin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jingbo Su
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jian Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Chen Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhimin Geng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Dong Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
110
|
Choi JW, Hua TNM. Impact of Lifestyle Behaviors on Cancer Risk and Prevention. J Lifestyle Med 2021; 11:1-7. [PMID: 33763336 PMCID: PMC7957047 DOI: 10.15280/jlm.2021.11.1.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 12/03/2020] [Indexed: 12/21/2022] Open
Abstract
Cancer incidences are rising globally. Therefore, in order to prevent and treat cancer, understanding cancer pathology is crucial. Tumors reprogram their metabolic phenotype to meet their needs for bioenergy, biosynthesis, and redox control. Alteration of the metabolic pathway has been proposed as the hallmark of cancer and explains the distinction between normal and cancer cells concerning nutrient utilization. Changes in the metabolism of nutrients such as glucose, amino acid, and fatty acid are associated with cancer risk. Luckily, this can be controlled with lifestyle modifications. Improvements in lifestyle behaviors to reduce cancer risks include a healthy diet, calorie restriction, and regular physical activity. This review begins with the understandings of metabolic reprogramming in cancer. Then, there will be evidence on the correlation between lifestyle factors and altered nutrient metabolism suggesting an application of lifestyle intervention for cancer risk reduction.
Collapse
Affiliation(s)
- Jong-Whan Choi
- Department of Biochemistry, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Tuyen N M Hua
- Department of Biochemistry, Yonsei University Wonju College of Medicine, Wonju, Korea.,Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju, Korea.,Department of Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
111
|
Wang HY, Liu YN, Wu SG, Hsu CL, Chang TH, Tsai MF, Lin YT, Shih JY. MiR-200c-3p suppression is associated with development of acquired resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors in EGFR mutant non-small cell lung cancer via a mediating epithelial-to-mesenchymal transition (EMT) process. Cancer Biomark 2021; 28:351-363. [PMID: 32417760 DOI: 10.3233/cbm-191119] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND EGFR-mutant lung cancer inevitably develops resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs). OBJECTIVE To investigate the clinical relevance of microRNAs (miRNAs) in TKI therapy response and resistance. METHODS We performed a miRNA PCR array analysis and used The Cancer Genome Atlas (TCGA) database to identify potential miRNAs related to EGFR TKIs resistance. We then correlated miRNA expression in 70 surgical and 50 malignant pleural effusion specimens with patient outcomes in those with non-small cell lung carcinoma. Molecular manipulation was performed in EGFR mutant lung cancer cells to assess the effect of miR-200c-3p on cell migratory ability and EGFR-TKI sensitivity. RESULTS We identified miR-200c-3p and miR-203a-3p as potential EGFR TKI resistance regulators via their modulation of epithelial-to-mesenchymal transition (EMT). MiR-200c-3p and miR-203a-3p were down-regulated in EGFR TKI-resistant cell lines. Progression-free survival (PFS) with EGFR-TKI treatment of patients with high miR-200c-3p expression, but not miR-203a-3p, in the specimens was significantly longer than that of patients with low expression. MiR-200c-3p overexpression inhibited the EMT process in EGFR TKI resistance cell lines and promoted cell death. MiR-200c-3p silencing in EGFR TKI sensitive cell lines increased drug resistance. CONCLUSION MiR-200c-3p plays a role in sensitivity to EGFR TKIs via modulating EMT process.
Collapse
Affiliation(s)
- Hsin-Yi Wang
- Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Thoracic Medicine Center, Department of Medicine and Surgery, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Yi-Nan Liu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shang-Gin Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Tzu-Hua Chang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Meng-Feng Tsai
- Department of Biomedical Sciences, Da-Yeh University, Changhua, Taiwan
| | - Yen-Ting Lin
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jin-Yuan Shih
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
112
|
Rahman I, Athar MT, Islam M. Type 2 Diabetes, Obesity, and Cancer Share Some Common and Critical Pathways. Front Oncol 2021; 10:600824. [PMID: 33552973 PMCID: PMC7855858 DOI: 10.3389/fonc.2020.600824] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
Diabetes and cancer are among the most frequent and complex diseases. Epidemiological evidence showed that the patients suffering from diabetes are significantly at higher risk for a number of cancer types. There are a number of evidence that support the hypothesis that these diseases are interlinked, and obesity may aggravate the risk(s) of type 2 diabetes and cancer. Multi-level unwanted alterations such as (epi-)genetic alterations, changes at the transcriptional level, and altered signaling pathways (receptor, cytoplasmic, and nuclear level) are the major source which promotes a number of complex diseases and such heterogeneous level of complexities are considered as the major barrier in the development of therapeutic agents. With so many known challenges, it is critical to understand the relationships and the commonly shared causes between type 2 diabetes and cancer, which is difficult to unravel and understand. Furthermore, the real complexity arises from contended corroborations that specific drug(s) (individually or in combination) during the treatment of type 2 diabetes may increase or decrease the cancer risk or affect cancer prognosis. In this review article, we have presented the recent and most updated evidence from the studies where the origin, biological background, the correlation between them have been presented or proved. Furthermore, we have summarized the methodological challenges and tasks that are frequently encountered. We have also outlined the physiological links between type 2 diabetes and cancers. Finally, we have presented and summarized the outline of the hallmarks for both these diseases, diabetes and cancer.
Collapse
Affiliation(s)
- Ishrat Rahman
- Department of Basic Dental Sciences, College of Dentistry, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Md Tanwir Athar
- Scientific Research Center, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Mozaffarul Islam
- Scientific Research Center, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
113
|
Acute lymphoid leukemia etiopathogenesis. Mol Biol Rep 2021; 48:817-822. [PMID: 33438082 DOI: 10.1007/s11033-020-06073-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 12/08/2020] [Indexed: 12/30/2022]
Abstract
Acute lymphoid leukemia (ALL) is a type of hematological neoplasm that affects the precursor cells of strains B, T and NK, with a higher incidence in the pediatric range. The pathophysiology of ALL is characterized by chromosomal abnormalities and genetic alterations involved in the differentiation and proliferation of lymphoid precursor cells. Despite the lack of information in the literature, it is believed that leukemogenesis originates from a complex interaction between environmental and genetic factors, which combined lead to cellular modifications. Environmental factors have been evaluated as possible predisposing factors in the development of ALL but there are still conflicting results in the world literature. In this context, the aim of the present review is to discuss the major exogenous factors regarding ALL.
Collapse
|
114
|
Yang S, Xia J, Yang Z, Xu M, Li S. Lung cancer molecular mutations and abnormal glycosylation as biomarkers for early diagnosis. Cancer Treat Res Commun 2021; 27:100311. [PMID: 33465560 DOI: 10.1016/j.ctarc.2021.100311] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 06/12/2023]
Abstract
Lung cancer is the leading cause of mortality and morbidity in tumor-related deaths in the world. Early detection of tumors can greatly improve the survival rate of patients. However, the lack of reliable blood biomarkers remains a major challenge for early diagnosis. The blood proteins secreted by the lung bronchi and bronchial arteries may have characteristic glycosylation patterns associated with tumors, which are different from normal physiological and pathological conditions. In this review, we outline the oncogenic drivers, signaling pathways related to KRAS, gene and protein mutations, and oncogenic regulation of protein glycosylation. Based on to the TCGA transcriptomics and antibody-based proteomics data, we discussed oncogene and glycoproteins detected in the blood as tumor biomarkers. We hypothesize that glycoproteins whose glycosylation can be reversed by targeted drugs may serve as potential tumor biomarkers.
Collapse
Affiliation(s)
- Shuang Yang
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Jun Xia
- Clinical Laboratory Center, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, China
| | - Zeren Yang
- Applied Biomimetic, Gaithersburg, Maryland 20878, United States
| | - Mingming Xu
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shuwei Li
- Nanjing Apollomics Biotech, Inc., Nanjing, Jiangsu 210033, China
| |
Collapse
|
115
|
Zhu L, Yang X, Zhu R, Yu L. Identifying Discriminative Biological Function Features and Rules for Cancer-Related Long Non-coding RNAs. Front Genet 2021; 11:598773. [PMID: 33391350 PMCID: PMC7772407 DOI: 10.3389/fgene.2020.598773] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/23/2020] [Indexed: 01/17/2023] Open
Abstract
Cancer has been a major public health problem worldwide for many centuries. Cancer is a complex disease associated with accumulative genetic mutations, epigenetic aberrations, chromosomal instability, and expression alteration. Increasing lines of evidence suggest that many non-coding transcripts, which are termed as non-coding RNAs, have important regulatory roles in cancer. In particular, long non-coding RNAs (lncRNAs) play crucial roles in tumorigenesis. Cancer-related lncRNAs serve as oncogenic factors or tumor suppressors. Although many lncRNAs are identified as potential regulators in tumorigenesis by using traditional experimental methods, they are time consuming and expensive considering the tremendous amount of lncRNAs needed. Thus, effective and fast approaches to recognize tumor-related lncRNAs should be developed. The proposed approach should help us understand not only the mechanisms of lncRNAs that participate in tumorigenesis but also their satisfactory performance in distinguishing cancer-related lncRNAs. In this study, we utilized a decision tree (DT), a type of rule learning algorithm, to investigate cancer-related lncRNAs with functional annotation contents [gene ontology (GO) terms and KEGG pathways] of their co-expressed genes. Cancer-related and other lncRNAs encoded by the key enrichment features of GO and KEGG filtered by feature selection methods were used to build an informative DT, which further induced several decision rules. The rules provided not only a new tool for identifying cancer-related lncRNAs but also connected the lncRNAs and cancers with the combinations of GO terms. Results provided new directions for understanding cancer-related lncRNAs.
Collapse
Affiliation(s)
- Liucun Zhu
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Xin Yang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Rui Zhu
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Lei Yu
- Department of Medical Oncology, Shanghai Concord Medical Cancer Center, Shanghai, China
| |
Collapse
|
116
|
Eco-friendly sequential one-pot synthesis, molecular docking, and anticancer evaluation of arylidene-hydrazinyl-thiazole derivatives as CDK2 inhibitors. Bioorg Chem 2021; 108:104615. [PMID: 33484942 DOI: 10.1016/j.bioorg.2020.104615] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 11/19/2020] [Accepted: 12/28/2020] [Indexed: 11/20/2022]
Abstract
One current approach in the treatment of cancer is the inhibition of cyclin dependent kinase (CDK) enzymes with small molecules. CDK are a class of enzymes, which catalyze the transfer of the terminal phosphate of a molecule of ATP to a protein that acts as a substrate. Among CDK enzymes, CDK2 has been implicated in a variety of cancers, supporting its potential as a novel target for cancer therapy across many tumor types. Here the discovery and development of arylidene-hydrazinyl-thiazole as a potentially CDK2 inhibitors is described, including details of the design and successful synthesis of the series analogs (27a-r) using one-pot approach under eco-friendly ultrasound and microwave conditions. Most of the newly synthesized compounds showed good growth inhibition when assayed for their in-vitro anti-proliferative activity against three cancer cell lines (HepG2, MCF-7 and HCT-116) compared to the reference drug roscovitine, with little toxicity on the normal fibroblast cell lines (WI-38). Furthermore, the compounds exhibiting the highest anti-proliferative activities were tested against a panel of kinase enzymes. These derivatives displayed an outstanding CDK2 inhibitory potential with varying degree of inhibition in the range of IC50 0.35-1.49 μM when compared with the standard inhibitor roscovitine having an IC50 value 0.71 μM. The most promising CDK2 inhibitor (27f) was selected for further studies to determine its effect on the cell cycle progression and apoptosis in HepG2 cell line. The results indicated that this compound implied inhibition in the G2/M phase of the cell cycle, and it is a good apoptotic agent. Finally, Molecular docking study was performed to identify the structural elements which involved in the inhibitory activity with the prospective target, CDK2, and to rationalize the structure-activity relationship (SAR).
Collapse
|
117
|
Yang B, Zhang Y, Pang S, Shang X, Zhao X, Han M. Integrating Multi-Omic Data With Deep Subspace Fusion Clustering for Cancer Subtype Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:216-226. [PMID: 31689204 DOI: 10.1109/tcbb.2019.2951413] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
One type of cancer usually consists of several subtypes with distinct clinical implications, thus the cancer subtype prediction is an important task in disease diagnosis and therapy. Utilizing one type of data from molecular layers in biological system to predict is difficult to bridge the cancer genome to cancer phenotypes, since the genome is neither simple nor independent but rather complicated and dysregulated from multiple molecular mechanisms. Similarity Network Fusion (SNF) has been recently proposed to integrate diverse omics data for improving the understanding of tumorigenesis. SNF adopts Euclidean distance to measure the similarity between patients, which shows some limitations. In this article, we introduce a novel prediction technique as an extension of SNF, namely Deep Subspace Fusion Clustering (DSFC). DSFC utilizes auto-encoder and data self-expressiveness approaches to guide a deep subspace model, which can achieve effective expression of discriminative similarity between patients. As a result, the dissimilarity between inter-cluster is delivered and enhanced compactness of intra-cluster is achieved at the same time. The validity of DSFC is examined by extensive simulations over six different cancer through three levels omics data. The survival analysis demonstrates that DSFC delivers comparable or even better results than many state-of-the-art integrative methods.
Collapse
|
118
|
Ambite I, Butler D, Wan MLY, Rosenblad T, Tran TH, Chao SM, Svanborg C. Molecular determinants of disease severity in urinary tract infection. Nat Rev Urol 2021; 18:468-486. [PMID: 34131331 PMCID: PMC8204302 DOI: 10.1038/s41585-021-00477-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2021] [Indexed: 02/06/2023]
Abstract
The most common and lethal bacterial pathogens have co-evolved with the host. Pathogens are the aggressors, and the host immune system is responsible for the defence. However, immune responses can also become destructive, and excessive innate immune activation is a major cause of infection-associated morbidity, exemplified by symptomatic urinary tract infections (UTIs), which are caused, in part, by excessive innate immune activation. Severe kidney infections (acute pyelonephritis) are a major cause of morbidity and mortality, and painful infections of the urinary bladder (acute cystitis) can become debilitating in susceptible patients. Disease severity is controlled at specific innate immune checkpoints, and a detailed understanding of their functions is crucial for strategies to counter microbial aggression with novel treatment and prevention measures. One approach is the use of bacterial molecules that reprogramme the innate immune system, accelerating or inhibiting disease processes. A very different outcome is asymptomatic bacteriuria, defined by low host immune responsiveness to bacteria with attenuated virulence. This observation provides the rationale for immunomodulation as a new therapeutic tool to deliberately modify host susceptibility, control the host response and avoid severe disease. The power of innate immunity as an arbitrator of health and disease is also highly relevant for emerging pathogens, including the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Ines Ambite
- grid.4514.40000 0001 0930 2361Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Daniel Butler
- grid.4514.40000 0001 0930 2361Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Murphy Lam Yim Wan
- grid.4514.40000 0001 0930 2361Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Therese Rosenblad
- grid.4514.40000 0001 0930 2361Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Thi Hien Tran
- grid.4514.40000 0001 0930 2361Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Sing Ming Chao
- Nephrology Service, Department of Paediatrics, KK Hospital, Singapore, Singapore
| | - Catharina Svanborg
- grid.4514.40000 0001 0930 2361Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
119
|
O'Brien SJ, Netz U, Hallion J, Bishop C, Stephen V, Burton J, Paas M, Feagins K, Pan J, Rai SN, Galandiuk S. Circulating plasma microRNAs in colorectal neoplasia: A pilot study in assessing response to therapy. Transl Oncol 2020; 14:100962. [PMID: 33285367 PMCID: PMC7720092 DOI: 10.1016/j.tranon.2020.100962] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 10/17/2020] [Accepted: 11/16/2020] [Indexed: 12/31/2022] Open
Abstract
This pilot study examines a microRNA panel as a biomarker for response to surgical resection in colorectal cancer or colorectal advanced adenoma. A panel of 11 microRNAs was developed through screening and previous studies. Six miRNA are significantly increased following colorectal cancer resection. Three miRNA are significantly increased following colorectal advanced adenoma resection. The results of this study suggest that serum microRNA expression could be followed as a marker for response to therapy.
Introduction Current serological surveillance markers to monitor colorectal cancer (CRC) or colorectal advanced adenomas (CAA) are hampered by poor sensitivity and specificity. The aim of this study is to identify and validate a panel of plasma microRNAs which change in expression after resection of such lesions. Methods A prospectively maintained colorectal surgery database was queried for patients in whom both pre- and post-procedural serum samples had been obtained. An initial screening analysis of CRC and CAA patients (5 each) was conducted using screening cards for 380 miRNAs. Four identified miRNAs were combined with a previously described panel of 7 miRNAs that were diagnostically predictive of CRC and CAA. Differential miRNA expression was assessed using quantitative real-time polymerase chain reaction(qRT-PCR). Results Fifty patients were included (n = 27 CRC, n = 23 CAA). There was no difference in age, gender, or race profile of CRC patients compared to CAA patients. Six miRNA were significantly increased after CRC resection (miR-324, let7b, miR-454, miR-374a, miR-122, miR-19b, all p<0.05), while three miRNAs were significantly increased following CAA resection (miR-454, miR-374a, miR-122, all p<0.05). Three miRNA were increased in common for both (miR-454, miR-374a, miR-122). Discussion The expression of miRNAs associated with neoplasia (either CRC or CAA) was significantly increased following surgical resection or endoscopic removal of CRC or CAA. Future studies should focus on the evaluation of these miRNAs in CRC and CAA prognosis.
Collapse
Affiliation(s)
- Stephen J O'Brien
- Price Institute of Surgical Research, The Hiram C. Polk Jr. MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Uri Netz
- Price Institute of Surgical Research, The Hiram C. Polk Jr. MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY 40292, USA; Department of Surgery, Soroka University Medical Center, Beer Sheva, Israel
| | - Jacob Hallion
- Price Institute of Surgical Research, The Hiram C. Polk Jr. MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Campbell Bishop
- Price Institute of Surgical Research, The Hiram C. Polk Jr. MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Vincent Stephen
- Price Institute of Surgical Research, The Hiram C. Polk Jr. MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - James Burton
- Price Institute of Surgical Research, The Hiram C. Polk Jr. MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Mason Paas
- Price Institute of Surgical Research, The Hiram C. Polk Jr. MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Kayla Feagins
- Price Institute of Surgical Research, The Hiram C. Polk Jr. MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Jianmin Pan
- Department of Bioinformatics and Biostatistics, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Shesh N Rai
- Department of Bioinformatics and Biostatistics, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Susan Galandiuk
- Price Institute of Surgical Research, The Hiram C. Polk Jr. MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY 40292, USA.
| |
Collapse
|
120
|
Hayes SC, Hofmann SG, Ciarrochi J. A process-based approach to psychological diagnosis and treatment:The conceptual and treatment utility of an extended evolutionary meta model. Clin Psychol Rev 2020; 82:101908. [PMID: 32932093 PMCID: PMC7680437 DOI: 10.1016/j.cpr.2020.101908] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 08/18/2020] [Accepted: 08/31/2020] [Indexed: 12/16/2022]
Abstract
For half a century, the dominant paradigm in psychotherapy research has been to develop syndrome-specific treatment protocols for hypothesized but unproved latent disease entities, as defined by psychiatric nosological systems. While this approach provided a common language for mental health problems, it failed to achieve its ultimate goal of conceptual and treatment utility. Process-based therapy (PBT) offers an alternative approach to understanding and treating psychological problems, and promoting human prosperity. PBT targets empirically established biopsychosocial processes of change that researchers have shown are functionally important to long terms goals and outcomes. By building on concepts of known clinical utility, and organizing them into coherent theoretical models, an idiographic, functional-analytic approach to diagnosis is within our grasp. We argue that a multi-dimensional, multi-level extended evolutionary meta-model (EEMM) provides consilience and a common language for process-based diagnosis. The EEMM applies the evolutionary concepts of context-appropriate variation, selection, and retention to key biopsychosocial dimensions and levels related to human suffering, problems, and positive functioning. The EEMM is a meta-model of diagnostic and intervention approaches that can accommodate any set of evidence-based change processes, regardless of the specific therapy orientation. In a preliminary way, it offers an idiographic, functional analytic, and clinically useful alternative to contemporary psychiatric nosological systems.
Collapse
|
121
|
Characterization of PMI-5011 on the Regulation of Deubiquitinating Enzyme Activity in Multiple Myeloma Cell Extracts. Biochem Eng J 2020; 166. [PMID: 33716550 DOI: 10.1016/j.bej.2020.107834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Deubiquitinating enzyme (DUB)-targeted therapeutics have shown promise in recent years as alternative cancer therapeutics, especially when coupled with proteasome-based inhibitors. While a majority of DUB-based therapeutics function by inhibiting DUB enzymes, studies show that positive regulation of these enzymes can stabilize levels of protein degradation. Unfortunately, there are currently no clinically available therapeutics for this purpose. The goal of this work was to understand the effect of a botanical extract from Artemisia dracunculus L called PMI-5011 on DUB activity in cancer cells. Through a series of kinetic analyses and mathematical modeling, it was found that PMI-5011 positively regulated DUB activity in two model multiple myeloma cells line (OPM2 and MM.1S). This suggests that PMI-5011 interacts with the active domains of DUBs to enhance their activity directly or indirectly, without apparently affecting cellular viability. Similar kinetic profiles of DUB activity were observed with three bioactive compounds in PMI-5011 (DMC-1, DMC-2, davidigenin). Interestingly, a differential cell line-independent trend was observed at higher concentrations which suggested variances in inherent gene expressions of UCHL1, UCHL5, USP7, USP15, USP14, and Rpn11 in OPM2 and MM.1S cell lines. These findings highlight the therapeutic potential of PMI-5011 and its selected bioactive compounds in cancer.
Collapse
|
122
|
Kwok ML, Meng Q, Hu XL, Chung CT, Chan KM. Whole-transcriptome sequencing (RNA-seq) study of the ZFL zebrafish liver cell line after acute exposure to Cd 2+ ions. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2020; 228:105628. [PMID: 32971353 DOI: 10.1016/j.aquatox.2020.105628] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/03/2020] [Accepted: 09/06/2020] [Indexed: 06/11/2023]
Abstract
Cadmium (Cd) is a non-essential metal with no known biological function and a broad range of toxic effects in biological systems. We used whole-transcriptome sequencing (RNA-seq) to study the effects of Cd2+ toxicity in zebrafish liver cells, ZFL. The results of an RNA-Seq analysis of ZFL cells exposed to 5, 10 or 20 μM Cd2+ for 4- or 24-h. The differentially expressed genes affected by Cd2+ were analyzed by using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis to study the regulated pathways. Cd2+ regulated the expression of genes associated with cellular Cu, Zn, and Fe homeostasis, DNA replication leading to cell cycle arrest and apoptosis, and glutathione metabolism. Cd2+ boosted up the amino acid synthesis, possibly to support the glutathione metabolism for tackling the oxidative stress generated from Cd2+. Cd2+ stimulation was similar to heat or xenobiotics, based on the responses from ZFL such as endoplasmic reticulum stress and protein folding. We linked also those finding of gene activations relating to carcinogenesis of Cd. This paper provides a comprehensive analysis of the expression profiles induced by Cd2+ exposure in ZFL cells, as well as useful insights into the specific toxic effects.
Collapse
Affiliation(s)
- Man Long Kwok
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin., N.T., Hong Kong
| | - Qi Meng
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin., N.T., Hong Kong
| | - Xue Lei Hu
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin., N.T., Hong Kong
| | - Chun Ting Chung
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin., N.T., Hong Kong
| | - King Ming Chan
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin., N.T., Hong Kong.
| |
Collapse
|
123
|
Kamalabadi-Farahani M, Kia V. High percentage of cancer stem cells in metastatic locations; a comment on a claim “variation in cancer risk among tissues can be explained by the number of stem cell divisions”. Med Hypotheses 2020. [DOI: https://doi.org/10.1016/j.mehy.2020.110170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
124
|
Gebert M, Jaśkiewicz M, Moszyńska A, Collawn JF, Bartoszewski R. The Effects of Single Nucleotide Polymorphisms in Cancer RNAi Therapies. Cancers (Basel) 2020; 12:E3119. [PMID: 33113880 PMCID: PMC7694039 DOI: 10.3390/cancers12113119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/14/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
Tremendous progress in RNAi delivery methods and design has allowed for the effective development of siRNA-based therapeutics that are currently under clinical investigation for various cancer treatments. This approach has the potential to revolutionize cancer therapy by providing the ability to specifically downregulate or upregulate the mRNA of any protein of interest. This exquisite specificity, unfortunately, also has a downside. Genetic variations in the human population are common because of the presence of single nucleotide polymorphisms (SNPs). SNPs lead to synonymous and non-synonymous changes and they occur once in every 300 base pairs in both coding and non-coding regions in the human genome. Much less common are the somatic mosaicism variations associated with genetically distinct populations of cells within an individual that is derived from postzygotic mutations. These heterogeneities in the population can affect the RNAi's efficacy or more problematically, which can lead to unpredictable and sometimes adverse side effects. From a more positive viewpoint, both SNPs and somatic mosaicisms have also been implicated in human diseases, including cancer, and these specific changes could offer the ability to effectively and, more importantly, selectively target the cancer cells. In this review, we discuss how SNPs in the human population can influence the development and success of novel anticancer RNAi therapies and the importance of why SNPs should be carefully considered.
Collapse
Affiliation(s)
- Magdalena Gebert
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.G.); (M.J.); (A.M.)
| | - Maciej Jaśkiewicz
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.G.); (M.J.); (A.M.)
| | - Adrianna Moszyńska
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.G.); (M.J.); (A.M.)
| | - James F. Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Rafał Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdańsk, 80-416 Gdańsk, Poland; (M.G.); (M.J.); (A.M.)
| |
Collapse
|
125
|
Zhou Y, An H, Wu G. MicroRNA-6071 Suppresses Glioblastoma Progression Through the Inhibition of PI3K/AKT/mTOR Pathway by Binding to ULBP2. Onco Targets Ther 2020; 13:9429-9441. [PMID: 33061429 PMCID: PMC7520159 DOI: 10.2147/ott.s265791] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022] Open
Abstract
Objective The purpose of this study was to explore the effect of microRNA-6071 (miR-6071) on glioblastoma (GBM) and its potential mechanisms. Methods In this study, the expressions of miR-6071 and UL16 binding protein 2 (ULBP2) were measured by qRT-RCR in GBM tissues and cells. The prognostic values of miR-6071 and ULBP2 were evaluated by Kaplan–Meier methods using the data obtained from The Cancer Genome Atlas (TCGA) database. The cell clones, proliferation, apoptosis, migration and invasion in GBM cells were detected by colony formation assay, EdU assay, flow cytometry, wound-healing assay and transwell assay. The targeting relationship between miR-6071 and ULBP2 was predicted by Targetscan 7.2 and further verified by dual-luciferase reporter gene assay. Moreover, the expressions of Bax, caspase-3, Bcl-2, matrix metalloproteinases 2 (MMP-2), MMP-9, phosphatidylinositol 3′-kinase (PI3K), p-PI3K, protein kinase B (AKT), p-AKT, mammalian target of rapamycin (mTOR) and p-mTOR were measured by Western blot. Results miR-6071 was lowly expressed and ULBP2 was highly expressed in GBM tissues and cells. miR-6071 significantly repressed the proliferation, migration and invasion, and promoted apoptosis in GBM cells. Moreover, miR-6071 also inhibited the activation of PI3K/AKT/mTOR pathway in GBM cells. Additionally, miR-6071 has been shown to negatively regulate ULBP2 expression. We also confirmed that ULBP2 could reverse the effects of miR-6071 on GBM cells through regulating PI3K/AKT/mTOR pathway. Conclusion Our study demonstrated that miR-6071 could suppress cell proliferation, migration and invasion, as well as promote apoptosis through the inhibition of PI3K/Akt/mTOR pathway by binding to ULBP2 in GBM.
Collapse
Affiliation(s)
- Yunyan Zhou
- Second Department of Neurology, Rongcheng People's Hospital, Shandong Province, Rongcheng, Shandong 264300, People's Republic of China
| | - Hongwei An
- Surgery of Lingcheng, Hospital of Traditional Chinese Medicine in Dezhou City, Dezhou, Shandong 253500, People's Republic of China
| | - Gang Wu
- Department of Neurology, Yan'an Hospital of Kunming, Kunming, Yunnan 650051, People's Republic of China
| |
Collapse
|
126
|
Kamalabadi-Farahani M, Kia V. High percentage of cancer stem cells in metastatic locations; a comment on a claim "variation in cancer risk among tissues can be explained by the number of stem cell divisions". Med Hypotheses 2020; 144:110170. [PMID: 32795837 DOI: 10.1016/j.mehy.2020.110170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/23/2020] [Accepted: 08/06/2020] [Indexed: 11/16/2022]
Abstract
Metastasis is the major cause of cancer-related deaths. Cancer relapse and metastasis are associated with a part of cancer cells with stem cell properties. These cancer stem cells (CSCs) are resistant to treatments. In a recent survey, we observed that the population of cancer stem-like cells among metastatic tumor cells was significantly higher than that among the primary tumor cells. This high percentage can partly explain the reasons for chemoresistance and relapse in metastatic cancers. Analysis of the role of CSCs in metastasis has been mainly conceptual and speculative, and the reasons for a higher number of CSCs in the metastatic loci are questionable. Tomasetti and Vogelstein's claim can partly answer the question. They postulated that the proliferation rate of normal stem cells in some tissue is greater than that of other tissues, and accordingly, the incidence of cancer in these tissues is high. In compliance with CSCs paradigm, resident normal stem cells of tissues are the most probable source of CSCs. After homing of metastatic cancer cells in a tissue with high rate of normal stem cell proliferation, there is a big opportunity for cancer cells to convert normal stem cells to cancer stem cells. This is the powerful effect of cancerous microenvironment on resident stem cells of tissue. Therefore, in metastatic cancers, the number of CSCs in primary tumor or in each metastatic location is relevant to the proliferation rate of resident normal stem cells of the location. This concept is a confirmation of Tomasetti and Vogelstein's claim and can answer some fundamental questions about metastasis process.
Collapse
Affiliation(s)
| | - Vahid Kia
- Department of Medical Biotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
127
|
Tanase C, Gheorghisan-Galateanu AA, Popescu ID, Mihai S, Codrici E, Albulescu R, Hinescu ME. CD36 and CD97 in Pancreatic Cancer versus Other Malignancies. Int J Mol Sci 2020; 21:E5656. [PMID: 32781778 PMCID: PMC7460590 DOI: 10.3390/ijms21165656] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 07/31/2020] [Accepted: 08/04/2020] [Indexed: 02/06/2023] Open
Abstract
Starting from the recent identification of CD36 and CD97 as a novel marker combination of fibroblast quiescence in lung during fibrosis, we aimed to survey the literature in search for facts about the separate (or concomitant) expression of clusters of differentiation CD36 and CD97 in either tumor- or pancreatic-cancer-associated cells. Here, we provide an account of the current knowledge on the diversity of the cellular functions of CD36 and CD97 and explore their potential (common) contributions to key cellular events in oncogenesis or metastasis development. Emphasis is placed on quiescence as an underexplored mechanism and/or potential target in therapy. Furthermore, we discuss intricate signaling mechanisms and networks involving CD36 and CD97 that may regulate different subpopulations of tumor-associated cells, such as cancer-associated fibroblasts, adipocyte-associated fibroblasts, tumor-associated macrophages, or neutrophils, during aggressive pancreatic cancer. The coexistence of quiescence and activated states in cancer-associated cell subtypes during pancreatic cancer should be better documented, in different histological forms. Remodeling of the local microenvironment may also change the balance between growth and dormant state. Taking advantage of the reported data in different other tissue types, we explore the possibility to induce quiescence (similar to that observed in normal cells), as a therapeutic option to delay the currently observed clinical outcome.
Collapse
Affiliation(s)
- Cristiana Tanase
- Victor Babeș National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; (I.D.P.); (S.M.); (E.C.); (R.A.); (M.E.H.)
- Faculty of Medicine, Titu Maiorescu University, 001863 Bucharest, Romania
| | - Ancuta-Augustina Gheorghisan-Galateanu
- Department of Cellular and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 8 Eroilor Sanitari Str., 050474 Bucharest, Romania;
- ‘C.I. Parhon’ National Institute of Endocrinology, 001863 Bucharest, Romania
| | - Ionela Daniela Popescu
- Victor Babeș National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; (I.D.P.); (S.M.); (E.C.); (R.A.); (M.E.H.)
| | - Simona Mihai
- Victor Babeș National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; (I.D.P.); (S.M.); (E.C.); (R.A.); (M.E.H.)
| | - Elena Codrici
- Victor Babeș National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; (I.D.P.); (S.M.); (E.C.); (R.A.); (M.E.H.)
| | - Radu Albulescu
- Victor Babeș National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; (I.D.P.); (S.M.); (E.C.); (R.A.); (M.E.H.)
- National Institute for Chemical Pharmaceutical R&D, 001863 Bucharest, Romania
| | - Mihail Eugen Hinescu
- Victor Babeș National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; (I.D.P.); (S.M.); (E.C.); (R.A.); (M.E.H.)
- Department of Cellular and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 8 Eroilor Sanitari Str., 050474 Bucharest, Romania;
| |
Collapse
|
128
|
PTEN Function at the Interface between Cancer and Tumor Microenvironment: Implications for Response to Immunotherapy. Int J Mol Sci 2020; 21:ijms21155337. [PMID: 32727102 PMCID: PMC7432882 DOI: 10.3390/ijms21155337] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022] Open
Abstract
Mounting preclinical and clinical evidence indicates that rewiring the host immune system in favor of an antitumor microenvironment achieves remarkable clinical efficacy in the treatment of many hematological and solid cancer patients. Nevertheless, despite the promising development of many new and interesting therapeutic strategies, many of these still fail from a clinical point of view, probably due to the lack of prognostic and predictive biomarkers. In that respect, several data shed new light on the role of the tumor suppressor phosphatase and tensin homolog on chromosome 10 (PTEN) in affecting the composition and function of the tumor microenvironment (TME) as well as resistance/sensitivity to immunotherapy. In this review, we summarize current knowledge on PTEN functions in different TME compartments (immune and stromal cells) and how they can modulate sensitivity/resistance to different immunological manipulations and ultimately influence clinical response to cancer immunotherapy.
Collapse
|
129
|
Patel KR, Patel HD. p53: An Attractive Therapeutic Target for Cancer. Curr Med Chem 2020; 27:3706-3734. [PMID: 31223076 DOI: 10.2174/1573406415666190621094704] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 02/28/2019] [Accepted: 04/16/2019] [Indexed: 02/08/2023]
Abstract
Cancer is a leading cause of death worldwide. It initiates when cell cycle regulatory genes lose their function either by environmental and/or by internal factors. Tumor suppressor protein p53, known as "Guardian of genome", plays a central role in maintaining genomic stability of the cell. Mutation of TP53 is documented in more than 50% of human cancers, usually by overexpression of negative regulator protein MDM2. Hence, reactivation of p53 by blocking the protein-protein interaction between the murine double minute 2 (MDM2) and the tumor suppressor protein p53 has become the most promising therapeutic strategy in oncology. Several classes of small molecules have been identified as potent, selective and efficient p53-MDM2 inhibitors. Herein, we review the druggability of p53-MDM2 inhibitors and their optimization approaches as well as clinical candidates categorized by scaffold type.
Collapse
Affiliation(s)
- Krupa R Patel
- Department of Chemistry, School of Sciences, Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Hitesh D Patel
- Department of Chemistry, School of Sciences, Gujarat University, Ahmedabad 380009, Gujarat, India
| |
Collapse
|
130
|
TranScreen: Transfer Learning on Graph-Based Anti-Cancer Virtual Screening Model. BIG DATA AND COGNITIVE COMPUTING 2020. [DOI: 10.3390/bdcc4030016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Deep learning’s automatic feature extraction has proven its superior performance over traditional fingerprint-based features in the implementation of virtual screening models. However, these models face multiple challenges in the field of early drug discovery, such as over-training and generalization to unseen data, due to the inherently unbalanced and small datasets. In this work, the TranScreen pipeline is proposed, which utilizes transfer learning and a collection of weight initializations to overcome these challenges. An amount of 182 graph convolutional neural networks are trained on molecular source datasets and the learned knowledge is transferred to the target task for fine-tuning. The target task of p53-based bioactivity prediction, an important factor for anti-cancer discovery, is chosen to showcase the capability of the pipeline. Having trained a collection of source models, three different approaches are implemented to compare and rank them for a given task before fine-tuning. The results show improvement in performance of the model in multiple cases, with the best model increasing the area under receiver operating curve ROC-AUC from 0.75 to 0.91 and the recall from 0.25 to 1. This improvement is vital for practical virtual screening via lowering the false negatives and demonstrates the potential of transfer learning. The code and pre-trained models are made accessible online.
Collapse
|
131
|
Huo YH, Wang YN, Meng LB, Zhang AL, Liu B. Progress in the correlation between PTPN12 gene expression and human tumors. Medicine (Baltimore) 2020; 99:e20445. [PMID: 32541467 PMCID: PMC7302617 DOI: 10.1097/md.0000000000020445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND The global morbidity of cancer is rising rapidly. Despite advances in molecular biology, immunology, and cytotoxic and immune-anticancer therapies, cancer remains a major cause of death worldwide. Protein tyrosine phosphatase non-receptor type 12 (PTPN12) is a new member of the cytoplasmic protein tyrosine phosphatase family, isolated from a cDNA library of adult colon tissue. Thus far, no studies have reviewed the correlation between PTPN12 gene expression and human tumors. METHODS This article summarizes the latest domestic and international research developments on how the expression of PTPN12 relates to human tumors. The extensive search in Web of Science and PubMed with the keywords including PTPN12, tumor, renal cell carcinoma, proto-oncogenes, tumor suppressor genes was undertaken. RESULTS More and more studies have shown that a tumor is essentially a genetic disease, arising from a broken antagonistic function between proto-oncogenes and tumor suppressor genes. When their antagonistic effect is out of balance, it may cause uncontrolled growth of cells and lead to the occurrence of tumors. PTPN12 is a tumor suppressor gene, so inhibiting its activity will lead directly or indirectly to the occurrence of tumors. CONCLUSION The etiology, prevention, and treatment of tumors have become the focus of research around the world. PTPN12 is a tumor suppressor gene. In the future, PTPN12 might serve as a novel molecular marker to benefit patients, and even the development of tumor suppressor gene activation agents can form a practical research direction.
Collapse
Affiliation(s)
- Yu-hu Huo
- Department of Breast and Thyroid Surgery, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai
| | - Ya-ni Wang
- School of Basic Medical Sciences, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei
| | - Ling-bing Meng
- School of Basic Medical Sciences, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei
| | - Ai-li Zhang
- Department of Urinary Surgery, The fourth hospital of Hebei medical university, P. R. China
| | - Bin Liu
- Department of Urinary Surgery, The fourth hospital of Hebei medical university, P. R. China
| |
Collapse
|
132
|
Ji X, Mukherjee S, Landi MT, Bosse Y, Joubert P, Zhu D, Gorlov I, Xiao X, Han Y, Gorlova O, Hung RJ, Brhane Y, Carreras-Torres R, Christiani DC, Caporaso N, Johansson M, Liu G, Bojesen SE, Le Marchand L, Albanes D, Bickeböller H, Aldrich MC, Bush WS, Tardon A, Rennert G, Chen C, Byun J, Dragnev KH, Field JK, Kiemeney LF, Lazarus P, Zienolddiny S, Lam S, Schabath MB, Andrew AS, Bertazzi PA, Pesatori AC, Diao N, Su L, Song L, Zhang R, Leighl N, Johansen JS, Mellemgaard A, Saliba W, Haiman C, Wilkens L, Fernandez-Somoano A, Fernandez-Tardon G, Heijden EHFMVD, Kim JH, Davies MPA, Marcus MW, Brunnström H, Manjer J, Melander O, Muller DC, Overvad K, Trichopoulou A, Tumino R, Goodman GE, Cox A, Taylor F, Woll P, Wichmann E, Muley T, Risch A, Rosenberger A, Grankvist K, Johansson M, Shepherd F, Tsao MS, Arnold SM, Haura EB, Bolca C, Holcatova I, Janout V, Kontic M, Lissowska J, Mukeria A, Ognjanovic S, Orlowski TM, Scelo G, Swiatkowska B, Zaridze D, Bakke P, Skaug V, Butler LM, Offit K, Srinivasan P, Bandlamudi C, Hellmann MD, Solit DB, Robson ME, Rudin CM, Stadler ZK, Taylor BS, Berger MF, Houlston R, McLaughlin J, Stevens V, Nickle DC, Obeidat M, Timens W, Artigas MS, Shete S, Brenner H, Chanock S, Brennan P, McKay JD, Amos CI. Protein-altering germline mutations implicate novel genes related to lung cancer development. Nat Commun 2020; 11:2220. [PMID: 32393777 PMCID: PMC7214407 DOI: 10.1038/s41467-020-15905-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 03/25/2020] [Indexed: 01/24/2023] Open
Abstract
Few germline mutations are known to affect lung cancer risk. We performed analyses of rare variants from 39,146 individuals of European ancestry and investigated gene expression levels in 7,773 samples. We find a large-effect association with an ATM L2307F (rs56009889) mutation in adenocarcinoma for discovery (adjusted Odds Ratio = 8.82, P = 1.18 × 10-15) and replication (adjusted OR = 2.93, P = 2.22 × 10-3) that is more pronounced in females (adjusted OR = 6.81 and 3.19 and for discovery and replication). We observe an excess loss of heterozygosity in lung tumors among ATM L2307F allele carriers. L2307F is more frequent (4%) among Ashkenazi Jewish populations. We also observe an association in discovery (adjusted OR = 2.61, P = 7.98 × 10-22) and replication datasets (adjusted OR = 1.55, P = 0.06) with a loss-of-function mutation, Q4X (rs150665432) of an uncharacterized gene, KIAA0930. Our findings implicate germline genetic variants in ATM with lung cancer susceptibility and suggest KIAA0930 as a novel candidate gene for lung cancer risk.
Collapse
Affiliation(s)
- Xuemei Ji
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
| | - Semanti Mukherjee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yohan Bosse
- Institut universitaire de cardiologie et de pneumologie de Québec, Department of Molecular Medicine, Laval University, Québec, Canada
| | - Philippe Joubert
- Institut universitaire de cardiologie et de pneumologie de Québec, Department of Molecular Medicine, Laval University, Québec, Canada
| | - Dakai Zhu
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Ivan Gorlov
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Xiangjun Xiao
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Younghun Han
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Olga Gorlova
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System and University of Toronto, Toronto, Canada
| | - Yonathan Brhane
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System and University of Toronto, Toronto, Canada
| | | | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital/Harvard, Boston, MA, USA
| | - Neil Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mattias Johansson
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Geoffrey Liu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System and University of Toronto, Toronto, Canada
| | - Stig E Bojesen
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Demetrios Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Heike Bickeböller
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
| | - Melinda C Aldrich
- Department of Thoracic Surgery, Division of Epidemiology, Vanderbilt University Medical Center, Göttingen, Germany
| | - William S Bush
- Department of Epidemiology and Biostatistics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Adonina Tardon
- IUOPA. University of Oviedo and CIBERESP, Faculty of Medicine, Campus del Cristo s/n, Oviedo, Spain
| | - Gad Rennert
- Clalit National Cancer Control Center at Carmel Medical Center and Technion Faculty of Medicine, Haifa, Israel
| | - Chu Chen
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jinyoung Byun
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Konstantin H Dragnev
- The Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - John K Field
- Roy Castle lung Cancer Research Programme, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Lambertus Fa Kiemeney
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, WA, USA
| | | | - Stephen Lam
- British Columbia Cancer Agency, Vancouver, Canada
| | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Angeline S Andrew
- Department of Epidemiology, Geisel School of Medicine, Hanover, NH, USA
| | - Pier A Bertazzi
- Department of Preventive Medicine, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Angela C Pesatori
- Department of Preventive Medicine, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Nancy Diao
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Li Su
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lei Song
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ruyang Zhang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Natasha Leighl
- University Health Network- The Princess Margaret Cancer Centre, Toronto, CA, USA
| | - Jakob S Johansen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Anders Mellemgaard
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Walid Saliba
- Clalit National Cancer Control Center at Carmel Medical Center and Technion Faculty of Medicine, Haifa, Israel
| | - Christopher Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Lynne Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Ana Fernandez-Somoano
- IUOPA. University of Oviedo and CIBERESP, Faculty of Medicine, Campus del Cristo s/n, Oviedo, Spain
| | | | | | - Jin Hee Kim
- Department of Integrative Bioscience & Biotechnology, Sejong University, Gwangjin-gu, Seoul, Republic of Korea
| | - Michael P A Davies
- Roy Castle lung Cancer Research Programme, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Michael W Marcus
- Roy Castle lung Cancer Research Programme, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | | | - Jonas Manjer
- Faculty of Medicine, Lund University, Lund, Sweden
| | | | - David C Muller
- School of Public Health, St Mary's Campus, Imperial College London, London, UK
| | - Kim Overvad
- Faculty of Medicine, Lund University, Lund, Sweden
| | | | - Rosario Tumino
- Cancer Registry and Histopathology Department, "Civic - M.P. Arezzo" Hospital, Asp Ragusa, Italy
| | - Gary E Goodman
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Swedish Medical Group, Seattle, WA, USA
| | - Angela Cox
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Fiona Taylor
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Penella Woll
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Erich Wichmann
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Muley
- Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), Heidelberg, Germany
| | - Angela Risch
- University of Salzburg and Cancer Cluster Salzburg, Salzburg, Austria
| | - Albert Rosenberger
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
| | - Kjell Grankvist
- Department of Medical Biosciences, Umeå University, Umeå, Sweden
| | | | | | | | - Susanne M Arnold
- University of Kentucky, Markey Cancer Center, Lexington, KY, USA
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ciprian Bolca
- Institute of Pneumology "Marius Nasta", Bucharest, Romania
| | - Ivana Holcatova
- Charles University, 1st Faculty of Medicine, Prague, Czech Republic
| | - Vladimir Janout
- Faculty of Health Sciences, Palacky University, Olomouc, Czech Republic
| | - Milica Kontic
- Clinical Center of Serbia, Clinic for Pulmonology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Prevention, M. Sklodowska-Curie Institute - Oncology Center, Warsaw, Poland
| | - Anush Mukeria
- Department of Epidemiology and Prevention, Russian N.N.Blokhin Cancer Research Centre, Moscow, Russian Federation
| | - Simona Ognjanovic
- International Organization for Cancer Prevention and Research, Belgrade, Serbia
| | - Tadeusz M Orlowski
- Department of Surgery, National Tuberculosis and Lung Diseases Research Institute, Warsaw, Poland
| | - Ghislaine Scelo
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Beata Swiatkowska
- Nofer Institute of Occupational Medicine, Department of Environmental Epidemiology, Lodz, Poland
| | - David Zaridze
- Department of Epidemiology and Prevention, Russian N.N.Blokhin Cancer Research Centre, Moscow, Russian Federation
| | - Per Bakke
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Vidar Skaug
- National Institute of Occupational Health, Oslo, Norway
| | | | - Kenneth Offit
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Preethi Srinivasan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Chaitanya Bandlamudi
- Marie-Josée and Henry R. KravisCenter for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Matthew D Hellmann
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David B Solit
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Marie-Josée and Henry R. KravisCenter for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Mark E Robson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Barry S Taylor
- Marie-Josée and Henry R. KravisCenter for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Michael F Berger
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
- Marie-Josée and Henry R. KravisCenter for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
| | | | | | | | - David C Nickle
- Merck Research Laboratories, Genetics and Pharmacogenomics, Boston, MA, USA
| | - Ma'en Obeidat
- The University of British Columbia Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, GRIAC research institute, Groningen, The Netherlands
| | - María Soler Artigas
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, LE1 7RH, UK
- National Institute for Health Research (NIHR) Leicester Respiratory Biomedical Research Unit, Glenfield Hospital, Leicester, UK
| | - Sanjay Shete
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Paul Brennan
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - James D McKay
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Christopher I Amos
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA.
- Dan L Duncan Comprehensive Cancer Center, 7200 Cambridge St., 7th Floor, Houston, TX, 77030, USA.
| |
Collapse
|
133
|
Dhanasekaran S. Phytochemical characteristics of aerial part of Cissus quadrangularis (L) and its in-vitro inhibitory activity against leukemic cells and antioxidant properties. Saudi J Biol Sci 2020; 27:1302-1309. [PMID: 32346339 PMCID: PMC7183005 DOI: 10.1016/j.sjbs.2020.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/11/2019] [Accepted: 01/06/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Cissus quadrangularis Linn, is a rich bioresource for folk and traditional medicines from ancient times till date. The present study aimed to investigate the free radical scavenging and anticancer efficacy in vitro of the ethanolic and methanolic extract from the aerial parts of Cissus quadrangularis (L). MATERIAL AND METHODS In vitro cell-free antioxidant analyses were performed for the ethanolic extract of Cissus quadrangularis (L). (EECQ) and methanolic extract of Cissus quadrangularis (L). (MECQ) using different free radical scavenging assays includes DPPH, nitric oxide, superoxide, metal chelation, and hydrogen peroxide radical scavenging assays. In vitro leukemic cytotoxic assessment by MTT assay was performed both EECQ and MECQ extract against HL-60 cell lines. RESULTS Strong antioxidant effects were recorded in EECQ and MECQ in all the cell-free models. The ethanolic extract exhibited a significant dose-dependent free radical activity in comparison with methanolic extracts. The EECQ and MECQ possess pronounced anticancer efficacy against leukemic cells HL-60 with an IC50 value of 36 μg/mL and 40 μg/mL respectively. CONCLUSION Present data indicates the presence of marked antioxidant and anticancer behaviors in the extracts of aerial portions of Cissus quadrangularis (L). extracts. Thus, Cissus quadrangularis (L). poses as a promising safe chemopreventive plant to combat cancer.
Collapse
Affiliation(s)
- Sugapriya Dhanasekaran
- Department of Medical Laboratory Sciences (Pathology), College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Wadi-Al Dawaser, Riyadh, Saudi Arabia
| |
Collapse
|
134
|
Helm JS, Rudel RA. Adverse outcome pathways for ionizing radiation and breast cancer involve direct and indirect DNA damage, oxidative stress, inflammation, genomic instability, and interaction with hormonal regulation of the breast. Arch Toxicol 2020. [PMID: 32399610 DOI: 10.1007/s00204-020-02752-z)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Knowledge about established breast carcinogens can support improved and modernized toxicological testing methods by identifying key mechanistic events. Ionizing radiation (IR) increases the risk of breast cancer, especially for women and for exposure at younger ages, and evidence overall supports a linear dose-response relationship. We used the Adverse Outcome Pathway (AOP) framework to outline and evaluate the evidence linking ionizing radiation with breast cancer from molecular initiating events to the adverse outcome through intermediate key events, creating a qualitative AOP. We identified key events based on review articles, searched PubMed for recent literature on key events and IR, and identified additional papers using references. We manually curated publications and evaluated data quality. Ionizing radiation directly and indirectly causes DNA damage and increases production of reactive oxygen and nitrogen species (RONS). RONS lead to DNA damage and epigenetic changes leading to mutations and genomic instability (GI). Proliferation amplifies the effects of DNA damage and mutations leading to the AO of breast cancer. Separately, RONS and DNA damage also increase inflammation. Inflammation contributes to direct and indirect effects (effects in cells not directly reached by IR) via positive feedback to RONS and DNA damage, and separately increases proliferation and breast cancer through pro-carcinogenic effects on cells and tissue. For example, gene expression changes alter inflammatory mediators, resulting in improved survival and growth of cancer cells and a more hospitable tissue environment. All of these events overlap at multiple points with events characteristic of "background" induction of breast carcinogenesis, including hormone-responsive proliferation, oxidative activity, and DNA damage. These overlaps make the breast particularly susceptible to ionizing radiation and reinforce that these biological activities are important characteristics of carcinogens. Agents that increase these biological processes should be considered potential breast carcinogens, and predictive methods are needed to identify chemicals that increase these processes. Techniques are available to measure RONS, DNA damage and mutation, cell proliferation, and some inflammatory proteins or processes. Improved assays are needed to measure GI and chronic inflammation, as well as the interaction with hormonally driven development and proliferation. Several methods measure diverse epigenetic changes, but it is not clear which changes are relevant to breast cancer. In addition, most toxicological assays are not conducted in mammary tissue, and so it is a priority to evaluate if results from other tissues are generalizable to breast, or to conduct assays in breast tissue. Developing and applying these assays to identify exposures of concern will facilitate efforts to reduce subsequent breast cancer risk.
Collapse
Affiliation(s)
- Jessica S Helm
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA
| | - Ruthann A Rudel
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA.
| |
Collapse
|
135
|
Helm JS, Rudel RA. Adverse outcome pathways for ionizing radiation and breast cancer involve direct and indirect DNA damage, oxidative stress, inflammation, genomic instability, and interaction with hormonal regulation of the breast. Arch Toxicol 2020; 94:1511-1549. [PMID: 32399610 PMCID: PMC7261741 DOI: 10.1007/s00204-020-02752-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/16/2020] [Indexed: 12/15/2022]
Abstract
Knowledge about established breast carcinogens can support improved and modernized toxicological testing methods by identifying key mechanistic events. Ionizing radiation (IR) increases the risk of breast cancer, especially for women and for exposure at younger ages, and evidence overall supports a linear dose-response relationship. We used the Adverse Outcome Pathway (AOP) framework to outline and evaluate the evidence linking ionizing radiation with breast cancer from molecular initiating events to the adverse outcome through intermediate key events, creating a qualitative AOP. We identified key events based on review articles, searched PubMed for recent literature on key events and IR, and identified additional papers using references. We manually curated publications and evaluated data quality. Ionizing radiation directly and indirectly causes DNA damage and increases production of reactive oxygen and nitrogen species (RONS). RONS lead to DNA damage and epigenetic changes leading to mutations and genomic instability (GI). Proliferation amplifies the effects of DNA damage and mutations leading to the AO of breast cancer. Separately, RONS and DNA damage also increase inflammation. Inflammation contributes to direct and indirect effects (effects in cells not directly reached by IR) via positive feedback to RONS and DNA damage, and separately increases proliferation and breast cancer through pro-carcinogenic effects on cells and tissue. For example, gene expression changes alter inflammatory mediators, resulting in improved survival and growth of cancer cells and a more hospitable tissue environment. All of these events overlap at multiple points with events characteristic of "background" induction of breast carcinogenesis, including hormone-responsive proliferation, oxidative activity, and DNA damage. These overlaps make the breast particularly susceptible to ionizing radiation and reinforce that these biological activities are important characteristics of carcinogens. Agents that increase these biological processes should be considered potential breast carcinogens, and predictive methods are needed to identify chemicals that increase these processes. Techniques are available to measure RONS, DNA damage and mutation, cell proliferation, and some inflammatory proteins or processes. Improved assays are needed to measure GI and chronic inflammation, as well as the interaction with hormonally driven development and proliferation. Several methods measure diverse epigenetic changes, but it is not clear which changes are relevant to breast cancer. In addition, most toxicological assays are not conducted in mammary tissue, and so it is a priority to evaluate if results from other tissues are generalizable to breast, or to conduct assays in breast tissue. Developing and applying these assays to identify exposures of concern will facilitate efforts to reduce subsequent breast cancer risk.
Collapse
Affiliation(s)
- Jessica S Helm
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA
| | - Ruthann A Rudel
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA.
| |
Collapse
|
136
|
Laham-Karam N, Pinto GP, Poso A, Kokkonen P. Transcription and Translation Inhibitors in Cancer Treatment. Front Chem 2020; 8:276. [PMID: 32373584 PMCID: PMC7186406 DOI: 10.3389/fchem.2020.00276] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
Transcription and translation are fundamental cellular processes that govern the protein production of cells. These processes are generally up regulated in cancer cells, to maintain the enhanced metabolism and proliferative state of these cells. As such cancerous cells can be susceptible to transcription and translation inhibitors. There are numerous druggable proteins involved in transcription and translation which make lucrative targets for cancer drug development. In addition to proteins, recent years have shown that the "undruggable" transcription factors and RNA molecules can also be targeted to hamper the transcription or translation in cancer. In this review, we summarize the properties and function of the transcription and translation inhibitors that have been tested and developed, focusing on the advances of the last 5 years. To complement this, we also discuss some of the recent advances in targeting oncogenes tightly controlling transcription including transcription factors and KRAS. In addition to natural and synthetic compounds, we review DNA and RNA based approaches to develop cancer drugs. Finally, we conclude with the outlook to the future of the development of transcription and translation inhibitors.
Collapse
Affiliation(s)
- Nihay Laham-Karam
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Gaspar P. Pinto
- International Clinical Research Center, St. Anne University Hospital, Brno, Czechia
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| | - Antti Poso
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
- University Hospital Tübingen, Department of Internal Medicine VIII, University of Tübingen, Tübingen, Germany
| | - Piia Kokkonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
137
|
YÜKSEL BİLEN M, BERKÖZ M, YALIN AE, ÇALIKUŞU Z, EROĞLU P, ÇÖMELEKOĞLU Ü, YALIN S. PALB2 genetik varyasyonlarının meme kanseri yatkınlığı üzerindeki etkilerinin araştırılması. CUKUROVA MEDICAL JOURNAL 2020. [DOI: 10.17826/cumj.634598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
138
|
Leonardi L, Benassi MS, Pollino S, Locaputo C, Pazzaglia L. miR-106B-25 Cluster expression: a comparative human and canine osteosarcoma study. Vet Rec Open 2020; 7:e000379. [PMID: 32201579 PMCID: PMC7061892 DOI: 10.1136/vetreco-2019-000379] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/05/2019] [Accepted: 02/07/2020] [Indexed: 12/19/2022] Open
Abstract
Background Osteosarcoma (OS) is the most common primary malignant bone tumour in dogs and human beings, characterised by similar genetic and clinical features. With the aim to define similarities and differences in the biological aspects involved in OS progression, a comparative study was performed to create a model to improve patient outcome. Methods First, the expression of microRNAs (miRNAs) belonging to the cluster miR-106b-25 (miR-106b, miR-25 and miR-93-5p) in human and canine OS tissue was compared. Results miR-25 and miR-106b presented a variable expression not significantly different from the corresponding normal bone, while miR-93-5p expression was increased in all OS specimens, with higher levels in the canine subset compared with human. Accordingly, its target p21 presented a weaker and less homogeneous immunostaining distribution in the canine group. Given the high expression of miR-93-5p in all OS specimens, the functional response of human 143B and canine DAN OS cells to miRNA inhibition was evaluated. Although p21 expression increased after miR-93-5p inhibition both at mRNA and protein level, a more significant cell response in terms of proliferation and apoptosis was seen in canine OS cells. Conclusions In conclusion, canine OS tissue and cell line presented higher expression levels of miR-93-5p than human OS. In addition, the introduction of miR-93-5p inhibitor caused a cell response in 143B and DAN that differed for the more intense functional impact in the canine OS cell line.
Collapse
Affiliation(s)
- Leonardo Leonardi
- Department of Veterinary Medicine - Veterinary Pathology, Università degli Studi di Perugia, Perugia, PG, Italy
| | - Maria Serena Benassi
- Oncologia Sperimentale, Istituto Ortopedico Rizzoli Istituto di Ricovero e Cura a Carattere Scientifico, Bologna, Italy
| | - Serena Pollino
- Oncologia Sperimentale, Istituto Ortopedico Rizzoli Istituto di Ricovero e Cura a Carattere Scientifico, Bologna, Italy
| | - Carmen Locaputo
- Oncologia Sperimentale, Istituto Ortopedico Rizzoli Istituto di Ricovero e Cura a Carattere Scientifico, Bologna, Italy
| | - Laura Pazzaglia
- Oncologia Sperimentale, Istituto Ortopedico Rizzoli Istituto di Ricovero e Cura a Carattere Scientifico, Bologna, Italy
| |
Collapse
|
139
|
Large-Scale Transgenic Drosophila Resource Collections for Loss- and Gain-of-Function Studies. Genetics 2020; 214:755-767. [PMID: 32071193 DOI: 10.1534/genetics.119.302964] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/11/2020] [Indexed: 01/20/2023] Open
Abstract
The Transgenic RNAi Project (TRiP), a Drosophila melanogaster functional genomics platform at Harvard Medical School, was initiated in 2008 to generate and distribute a genome-scale collection of RNA interference (RNAi) fly stocks. To date, it has generated >15,000 RNAi fly stocks. As this covers most Drosophila genes, we have largely transitioned to development of new resources based on CRISPR technology. Here, we present an update on our libraries of publicly available RNAi and CRISPR fly stocks, and focus on the TRiP-CRISPR overexpression (TRiP-OE) and TRiP-CRISPR knockout (TRiP-KO) collections. TRiP-OE stocks express single guide RNAs targeting upstream of a gene transcription start site. Gene activation is triggered by coexpression of catalytically dead Cas9 fused to an activator domain, either VP64-p65-Rta or Synergistic Activation Mediator. TRiP-KO stocks express one or two single guide RNAs targeting the coding sequence of a gene or genes. Cutting is triggered by coexpression of Cas9, allowing for generation of indels in both germline and somatic tissue. To date, we have generated >5000 TRiP-OE or TRiP-KO stocks for the community. These resources provide versatile, transformative tools for gene activation, gene repression, and genome engineering.
Collapse
|
140
|
Reddy D, Ghosh P, Kumavath R. Strophanthidin Attenuates MAPK, PI3K/AKT/mTOR, and Wnt/β-Catenin Signaling Pathways in Human Cancers. Front Oncol 2020; 9:1469. [PMID: 32010609 PMCID: PMC6978703 DOI: 10.3389/fonc.2019.01469] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/09/2019] [Indexed: 12/16/2022] Open
Abstract
Lung cancer is the most prevalent in cancer-related deaths, while breast carcinoma is the second most dominant cancer in women, accounting for the most number of deaths worldwide. Cancers are heterogeneous diseases that consist of several subtypes based on the presence or absence of hormone receptors and human epidermal growth factor receptor 2. Several drugs have been developed targeting cancer biomarkers; nonetheless, their efficiency are not adequate due to the high reemergence rate of cancers and fundamental or acquired resistance toward such drugs, which leads to partial therapeutic possibilities. Recent studies on cardiac glycosides (CGs) positioned them as potent cytotoxic agents that target multiple pathways to initiate apoptosis and autophagic cell death in many cancers. In the present study, our aim is to identify the anticancer activity of a naturally available CG (strophanthidin) in human breast (MCF-7), lung (A549), and liver cancer (HepG2) cells. Our results demonstrate a dose-dependent cytotoxic effect of strophanthidin in MCF-7, A549, and HepG2 cells, which was further supported by DNA damage on drug treatment. Strophanthidin arrested the cell cycle at the G2/M phase; this effect was further validated by checking the inhibited expressions of checkpoint and cyclin-dependent kinases in strophanthidin-induced cells. Moreover, strophanthidin inhibited the expression of several key proteins such as MEK1, PI3K, AKT, mTOR, Gsk3α, and β-catenin from MAPK, PI3K/AKT/mTOR, and Wnt/β-catenin signaling. The current study adequately exhibits the role of strophanthidin in modulating the expression of various key proteins involved in cell cycle arrest, apoptosis, and autophagic cell death. Our in silico studies revealed that strophanthidin can interact with several key proteins from various pathways. Taken together, this study demonstrates the viability of strophanthidin as a promising anticancer agent, which may serve as a new anticancer drug.
Collapse
Affiliation(s)
- Dhanasekhar Reddy
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Kasaragod, India
| | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA, United States
| | - Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Kasaragod, India
| |
Collapse
|
141
|
Qazi AS, Akbar S, Saeed RF, Bhatti MZ. Translational Research in Oncology. 'ESSENTIALS OF CANCER GENOMIC, COMPUTATIONAL APPROACHES AND PRECISION MEDICINE 2020:261-311. [DOI: 10.1007/978-981-15-1067-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
142
|
Mokhtarian R, Tabatabaeian H, Saadatmand P, Azadeh M, Balmeh N, Yakhchali B, Ghaedi K. CD44 Gene rs8193 C Allele Is Significantly Enriched in Gastric Cancer Patients. CELL JOURNAL 2020; 21:451-458. [PMID: 31376327 PMCID: PMC6722445 DOI: 10.22074/cellj.2020.6389] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/23/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Gastric cancer is a multifactorial disease. In addition to environmental factors, many genes are involved in this malignancy. One of the genes associated with gastric cancer is CD44 gene and its polymorphisms. CD44 gene plays role in regulating cell survival, growth and mobility. The single nucleotide polymorphism (SNP) rs8193, located in the CD44 gene, has not been studied in gastric cancer patients of the Iranian population. The present study aims to study this polymorphism in 86 gastric cancer patients and 96 healthy individuals. MATERIALS AND METHODS In this cross-sectional case-control study, rs8193 polymorphism was genotyped by allele specific primer polymerase chain reaction (ASP-PCR) technique. The obtained data were statistically analyzed. To find the potential mechanism of action, rs8193 was bioinformatically investigated. RESULTS rs8193 C allele played a risk factor role for gastric cancer. Patients carrying this allele were more susceptible to have gastric cancer, with lymph node spread. On the other hand, rs8193 T allele, a protective factor, was associated with a higher chance of accumulation in the lower stages of cancer. C allele might impose its effect via destabilizing CD44 and miR-570 interaction. CONCLUSION rs8193 is statistically associated with the risk of malignancy, lymph node spread and stage of gastric cancer in Iranian population.
Collapse
Affiliation(s)
- Roya Mokhtarian
- Division of Cellular and Molecular Biology, Department of Biology, NourDanesh Institute of Higher Education, Meymeh, Iran
| | - Hossein Tabatabaeian
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | | | | | - Negar Balmeh
- Division of Cellular and Molecular Biology, Department of Biology, NourDanesh Institute of Higher Education, Meymeh, Iran
| | - Bagher Yakhchali
- Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology Institute, Tehran, Iran
| | - Kamran Ghaedi
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran. Electronic Address:
| |
Collapse
|
143
|
Bagger FO, Probst V. Single Cell Sequencing in Cancer Diagnostics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1255:175-193. [PMID: 32949400 DOI: 10.1007/978-981-15-4494-1_15] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Personalized medicine has been driven by improvements in genomic sequencing and analysis. For several diseases, in particular cancers, it has for nearly a decade been standard clinical practice to analyze the genome and expression of the genes of patients. The results are reflected directly in the treatment plan for the patient, in targeted medical inventions. This specialized mode of diagnostics has been restricted to account for averaged trends in the tumor. The approach sharply contrasts our knowledge on heterogeneity within tumors. Several studies further describe how treatment against one tumor subclone in some cases merely serves to provide space and support for uncontrolled growth of more aggressive subclones. In this chapter, we describe current possibilities for implementation of single cell sequencing of malignomas in clinic, as well as discuss hands-on practical advice for single cell routine diagnostics that allows for full delineation of tumor clonality.
Collapse
Affiliation(s)
- Frederik Otzen Bagger
- Department of Genomic Medicine, Rigshospitalet, Centre of Diagnostics, Section 4113, Copenhagen, Denmark.
| | - Victoria Probst
- Department of Genomic Medicine, Rigshospitalet, Centre of Diagnostics, Section 4113, Copenhagen, Denmark
| |
Collapse
|
144
|
Elhefnawi M, Salah Z, Soliman B. The Promise of miRNA Replacement Therapy for Hepatocellular Carcinoma. Curr Gene Ther 2019; 19:290-304. [DOI: 10.2174/1566523219666191023101433] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/25/2019] [Accepted: 10/09/2019] [Indexed: 12/15/2022]
Abstract
Hepatocellular carcinoma is a devastating tumor which accounts for death mortality rate
94% globally, and about 780,000 new cases each year. Tumor suppressor miRNAs represent a class of
noncoding RNAs, which exhibit decreased or inhibited expression in the case of carcinogenesis.
Therefore, the replacement of these molecules leads to post-transcriptional regulation of tens to hundreds
of oncogenic targets and limiting the tumor. Interestingly, there is a group of tumor silencer
miRNAs that have been highlighted in HCC and herein, our review will discuss the prominent examples
of these miRs in terms of their efficient delivery using vectors, nano-delivery systems, their successful
models either in vitro or in vivo and pre-clinical trials. Collectively, tumor suppressor miRNAs
can act as novel therapeutics for HCC and more studies should be directed towards these promising
therapeutics.
Collapse
Affiliation(s)
- Mahmoud Elhefnawi
- Biomedical Informatics and Chemo-Informatics Group Leader, Centre of Excellence for Medical Research, National Research Centre (NRC), Cairo, Egypt
| | - Zeinab Salah
- Biomedical Informatics and Chemo-Informatics Group Leader, Centre of Excellence for Medical Research, National Research Centre (NRC), Cairo, Egypt
| | - Bangly Soliman
- Biomedical Informatics and Chemo-Informatics Group Leader, Centre of Excellence for Medical Research, National Research Centre (NRC), Cairo, Egypt
| |
Collapse
|
145
|
Lynch KL, Gooding LR, Garnett-Benson C, Ornelles DA, Avgousti DC. Epigenetics and the dynamics of chromatin during adenovirus infections. FEBS Lett 2019; 593:3551-3570. [PMID: 31769503 DOI: 10.1002/1873-3468.13697] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 11/20/2019] [Accepted: 11/23/2019] [Indexed: 12/26/2022]
Abstract
The DNA genome of eukaryotic cells is compacted by histone proteins within the nucleus to form chromatin. Nuclear-replicating viruses such as adenovirus have evolved mechanisms of chromatin manipulation to promote infection and subvert host defenses. Epigenetic factors may also regulate persistent adenovirus infection and reactivation in lymphoid tissues. In this review, we discuss the viral proteins E1A and protein VII that interact with and alter host chromatin, as well as E4orf3, which separates host chromatin from sites of viral replication. We also highlight recent advances in chromatin technologies that offer new insights into virus-directed chromatin manipulation. Beyond the role of chromatin in the viral replication cycle, we discuss the nature of persistent viral genomes in lymphoid tissue and cell lines, and the potential contribution of epigenetic signals in maintaining adenovirus in a quiescent state. By understanding the mechanisms through which adenovirus manipulates host chromatin, we will understand new aspects of this ubiquitous virus and shed light on previously unknown aspects of chromatin biology.
Collapse
Affiliation(s)
- Kelsey L Lynch
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Linda R Gooding
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | | | - David A Ornelles
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Daphne C Avgousti
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
146
|
Lin LH, Chang KW, Cheng HW, Liu CJ. SMAD4 Somatic Mutations in Head and Neck Carcinoma Are Associated With Tumor Progression. Front Oncol 2019; 9:1379. [PMID: 31867281 PMCID: PMC6909744 DOI: 10.3389/fonc.2019.01379] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
As the incidence and the mortality rate of head and neck squamous cell carcinoma (HNSCC) is increasing worldwide, gaining knowledge about the genomic changes which happen in the carcinogenesis of HNSCC is essential for the diagnosis and therapy of the disease. SMAD4 (DPC4) is a tumor suppressor gene. It is located at chromosome 18q21.1 and a member of the SMAD family. Which mediates the TGF-β signaling pathway, thereby controlling the growth of epithelial cells. In the study presented here, we analyzed tumor samples by multiplex PCR-based next-generation sequencing (NGS) and found deleterious mutations of SMAD4 in 4.1% of the tumors. Knock-down experiments of endogenous and exogenous SMAD4 expression demonstrated that SMAD4 is involved in the migration and invasion of HNSCC cells. Functional analysis of a missense mutation in the MH1 domain of SMAD4 may be responsible for the loss of function in suppressing tumor progression. Missense SMAD4 mutations, therefore, could be useful prognostic determinants for patients affected by HNSCCs. This report is the first study where NGS analysis based on multiplex-PCR is used to demonstrate the imminent occurrence of missense SMAD4 mutations in HNSCC cells. The gene analysis that we performed may support the identification of SMAD4 mutations as a diagnostic marker or even as a potential therapeutic target in head and neck cancer. Moreover, the analytic strategy proposed for the detection of mutations in the SMAD4 gene may be validated as a platform to assist mutation screening.
Collapse
Affiliation(s)
- Li-Han Lin
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Kuo-Wei Chang
- Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Dentistry, Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan
| | - Hui-Wen Cheng
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chung-Ji Liu
- School of Dentistry, Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan.,Department of Oral and Maxillofacial Surgery, Taipei MacKay Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
147
|
Ohshima K, Fujiya K, Nagashima T, Ohnami S, Hatakeyama K, Urakami K, Naruoka A, Watanabe Y, Moromizato S, Shimoda Y, Ohnami S, Serizawa M, Akiyama Y, Kusuhara M, Mochizuki T, Sugino T, Shiomi A, Tsubosa Y, Uesaka K, Terashima M, Yamaguchi K. Driver gene alterations and activated signaling pathways toward malignant progression of gastrointestinal stromal tumors. Cancer Sci 2019; 110:3821-3833. [PMID: 31553483 PMCID: PMC6890443 DOI: 10.1111/cas.14202] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/17/2019] [Accepted: 09/22/2019] [Indexed: 12/28/2022] Open
Abstract
Mutually exclusive KIT and PDGFRA mutations are considered to be the earliest events in gastrointestinal stromal tumors (GIST), but insufficient for their malignant progression. Herein, we aimed to identify driver genes and signaling pathways relevant to GIST progression. We investigated genetic profiles of 707 driver genes, including mutations, gene fusions, copy number gain or loss, and gene expression for 65 clinical specimens of surgically dissected GIST, consisting of six metastatic tumors and 59 primary tumors from stomach, small intestine, rectum, and esophagus. Genetic alterations included oncogenic mutations and amplification‐dependent expression enhancement for oncogenes (OG), and loss of heterozygosity (LOH) and expression reduction for tumor suppressor genes (TSG). We assigned activated OG and inactivated TSG to 27 signaling pathways, the activation of which was compared between malignant GIST (metastasis and high‐risk GIST) and less malignant GIST (low‐ and very low‐risk GIST). Integrative molecular profiling indicated that a greater incidence of genetic alterations of driver genes was detected in malignant GIST (96%, 22 of 23) than in less malignant GIST (73%, 24 of 33). Malignant GIST samples groups showed mutations, LOH, and aberrant expression dominantly in driver genes associated with signaling pathways of PI3K (PIK3CA, AKT1, and PTEN) and the cell cycle (RB1, CDK4, and CDKN1B). Additionally, we identified potential PI3K‐related genes, the expression of which was upregulated (SNAI1 and TPX2) or downregulated (BANK1) in malignant GIST. Based on our observations, we propose that inhibition of PI3K pathway signals might potentially be an effective therapeutic strategy against malignant progression of GIST.
Collapse
Affiliation(s)
- Keiichi Ohshima
- Medical Genetics Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan.,Drug Discovery and Development Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan
| | - Keiichi Fujiya
- Division of Gastric Surgery, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Takeshi Nagashima
- Cancer Diagnostics Research Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan.,SRL, Inc., Tokyo, Japan
| | - Sumiko Ohnami
- Cancer Diagnostics Research Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan
| | - Keiichi Hatakeyama
- Medical Genetics Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan
| | - Kenichi Urakami
- Cancer Diagnostics Research Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan.,Region Resources Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan
| | - Akane Naruoka
- Drug Discovery and Development Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan
| | - Yuko Watanabe
- Medical Genetics Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan
| | - Sachi Moromizato
- Medical Genetics Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan
| | - Yuji Shimoda
- Cancer Diagnostics Research Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan.,SRL, Inc., Tokyo, Japan
| | - Shumpei Ohnami
- Cancer Diagnostics Research Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan
| | - Masakuni Serizawa
- Drug Discovery and Development Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan
| | - Yasuto Akiyama
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan
| | - Masatoshi Kusuhara
- Drug Discovery and Development Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan.,Region Resources Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan
| | - Tohru Mochizuki
- Medical Genetics Division, Shizuoka Cancer Center Research Institute, Shizuoka, Japan
| | - Takashi Sugino
- Division of Pathology, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Akio Shiomi
- Division of Colon and Rectal Surgery, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Yasuhiro Tsubosa
- Division of Esophageal Surgery, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Katsuhiko Uesaka
- Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Masanori Terashima
- Division of Gastric Surgery, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Ken Yamaguchi
- Shizuoka Cancer Center Hospital and Research Institute, Shizuoka, Japan
| |
Collapse
|
148
|
Abstract
miRNAs, a major class of small noncoding RNAs approximately 18-25 nucleotides in length, function by repressing the expression of target genes through binding to complementary sequences in the 3'-UTRs of target genes. Emerging evidence has highlighted their important roles in numerous diseases, including human cancers. Recently, miR-190 has been shown to be dysregulated in various types of human cancers that participates in cancer-related biological processes, including proliferation, apoptosis, metastasis, drug resistance, by regulating associated target genes, and to predict cancer diagnosis and prognosis. In this review, we summarized the roles of miR-190-5p in human diseases, especially in human cancers. Then we classified its target genes in tumorigenesis and progression, which might provide evidence for cancer diagnosis and prognosis, promising tools for cancer treatment, or leads for further investigation.
Collapse
Affiliation(s)
- Yue Yu
- 1The First Department of Breast Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huan-Hu-Xi Road, Hexi District, Tianjin, 300060 China.,2Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060 China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060 China.,4Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060 China
| | - Xu-Chen Cao
- 1The First Department of Breast Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Huan-Hu-Xi Road, Hexi District, Tianjin, 300060 China.,2Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060 China.,Tianjin's Clinical Research Center for Cancer, Tianjin, 300060 China.,4Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060 China
| |
Collapse
|
149
|
Xue Y, Cooper G, Cai C, Lu S, Hu B, Ma X, Lu X. Tumour-specific Causal Inference Discovers Distinct Disease Mechanisms Underlying Cancer Subtypes. Sci Rep 2019; 9:13225. [PMID: 31519988 PMCID: PMC6744493 DOI: 10.1038/s41598-019-48318-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/31/2019] [Indexed: 01/22/2023] Open
Abstract
Cancer is a disease mainly caused by somatic genome alterations (SGAs) that perturb cellular signalling systems. Furthermore, the combination of pathway aberrations in a tumour defines its disease mechanism, and distinct disease mechanisms underlie the inter-tumour heterogeneity in terms of disease progression and responses to therapies. Discovering common disease mechanisms shared by tumours would provide guidance for precision oncology but remains a challenge. Here, we present a novel computational framework for revealing distinct combinations of aberrant signalling pathways in tumours. Specifically, we applied the tumour-specific causal inference algorithm (TCI) to identify causal relationships between SGAs and differentially expressed genes (DEGs) within tumours from the Cancer Genome Atlas (TCGA) study. Based on these causal inferences, we adopted a network-based method to identify modules of DEGs, such that the member DEGs within a module tend to be co-regulated by a common pathway. Using the expression status of genes in a module as a surrogate measure of the activation status of the corresponding pathways, we divided breast cancers (BRCAs) into five subgroups and glioblastoma multiformes (GBMs) into six subgroups with distinct combinations of pathway aberrations. The patient groups exhibited significantly different survival patterns, indicating that our approach can identify clinically relevant disease subtypes.
Collapse
Affiliation(s)
- Yifan Xue
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, 15260, United States
| | - Gregory Cooper
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, 15260, United States
| | - Chunhui Cai
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, 15260, United States
| | - Songjian Lu
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, 15260, United States
| | - Baoli Hu
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, 15260, United States.,Paediatric Neurosurgery, UPMC Children's Hospital of Pittsburgh, Pittsburgh, 15213, United States.,Molecular and Cellular Cancer Biology Program, UPMC Hillman Cancer Centre, Pittsburgh, 15232, United States
| | - Xiaojun Ma
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, 15260, United States
| | - Xinghua Lu
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, 15260, United States.
| |
Collapse
|
150
|
Petracco R, Dias ACDO, Taylor H, Petracco Á, Badalotti M, Michelon JDR, Marinowic DR, Hentschke M, Azevedo PND, Zanirati G, Machado DC. Evaluation of miR-135a/b expression in endometriosis lesions. Biomed Rep 2019; 11:181-187. [PMID: 31565224 DOI: 10.3892/br.2019.1237] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 07/10/2019] [Indexed: 12/24/2022] Open
Abstract
The pathogenesis of endometriosis is not clear; however, microRNAs (miRNAs/miRs) are involved in the pathogenesis. miRNAs are short noncoding RNAs involved in post-transcriptional regulation of gene expression by silencing the expression of target genes. The expression of miR-135a/b is associated with endometrial receptivity and implantation; the expression is also associated with the expression of certain genes, including homeobox protein Hox-A10 (HOXA-10). The present study investigated the expression of miR-135a/b in eutopic and ectopic endometrium tissues throughout the different phases of the menstrual cycle. Samples of ectopic endometriosis lesions and eutopic endometrium tissue from 23 patients who underwent laparoscopic surgery were obtained and analyzed. miRNA was extracted and the expression levels of miR-135a/b were determined by reverse transcription quantitative polymerase chain reaction assays using U6 as a housekeeping control. The expression levels of miR-135a and miR-135b in endometriosis lesions were decreased compared with the levels in endometrium tissue. However, miR-135a/b expression levels were increased in the secretory phase compared with the proliferative phase in endometriosis lesions. The increased expression of miR-135a/b during the secretory phase compared with the proliferative phase suggested that these genes serve a determinant role in the homeostasis of reproductive tissue. Therefore, the expression of genes may affect endometrial functioning, impairing embryo implantation.
Collapse
Affiliation(s)
- Rafaella Petracco
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Fertilitat Center of Reproductive Medicine, Porto Alegre, Rio Grande do Sul 90610-000, Brazil
| | | | - Hugh Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, New Haven, CT 06520-8063, USA
| | - Álvaro Petracco
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Fertilitat Center of Reproductive Medicine, Porto Alegre, Rio Grande do Sul 90610-000, Brazil
| | - Mariângela Badalotti
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Fertilitat Center of Reproductive Medicine, Porto Alegre, Rio Grande do Sul 90610-000, Brazil
| | - João Da Rosa Michelon
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Fertilitat Center of Reproductive Medicine, Porto Alegre, Rio Grande do Sul 90610-000, Brazil
| | - Daniel Rodrigo Marinowic
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Fertilitat Center of Reproductive Medicine, Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-000, Brazil
| | - Marta Hentschke
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Fertilitat Center of Reproductive Medicine, Porto Alegre, Rio Grande do Sul 90610-000, Brazil
| | - Pamella Nunes De Azevedo
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-000, Brazil
| | - Gabriele Zanirati
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-000, Brazil
| | - Denise Cantarelli Machado
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-000, Brazil.,Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90610-000, Brazil
| |
Collapse
|