1
|
Zhong H, Shao X, Cao J, Huang J, Wang J, Yang N, Yuan B. Comparison of the Distribution Patterns of Microsatellites Across the Genomes of Reptiles. Ecol Evol 2024; 14:e70458. [PMID: 39575149 PMCID: PMC11581779 DOI: 10.1002/ece3.70458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 08/29/2024] [Accepted: 09/29/2024] [Indexed: 11/24/2024] Open
Abstract
Microsatellites or simple sequence repeats (SSRs) are prevalent across various organisms' genomes. However, their distribution patterns and evolutionary dynamics in reptile genomes are rarely studied systematically. We herein conducted a comprehensive analysis of SSRs in the genomes of 36 reptile species. Our findings revealed that the total number of SSRs ranged from 1,840,965 to 7,664,452, accounting for 2.16%-8.19% of the genomes analyzed. The relative density ranged from 21,567.82 to 81,889.41 bp per megabase (Mbp). The abundance of different SSR categories followed the pattern of imperfect SSR (I-SSR) > perfect SSR (P-SSR) > compound SSR (C-SSR). A significant positive correlation was observed between the number of SSRs and genome size (p = 0.0034), whereas SSR frequency (p = 0.013) or density (p = 0.0099) showed a negative correlation with genome size. Furthermore, no correlation was found between SSR length and genome size. Mononucleotide repeats were the most common P-SSRs in crocodilians and turtles, whereas mononucleotides, trinucleotides, or tetranucleotides were the most common P-SSRs in snakes, lizards, and tuatara. P-SSRs of varying motif sizes showed nonrandom distribution across different genic regions, with AT-rich repeats being predominant. The genomic SSR content of the squamate lineage ranked the highest in abundance and variability, whereas crocodilians and turtles showed a slowly evolving and reduced microsatellite landscape. Gene ontology enrichment and Kyoto Encyclopedia of Genes and Genomes pathway analyses indicated that genes harboring P-SSRs in the coding DNA sequence regions were primarily involved in the regulation of transcription and translation processes. The SSR dataset generated in this study provides potential candidates for functional analysis and calls for broader-scale analyses across the evolutionary spectrum.
Collapse
Affiliation(s)
- Huaming Zhong
- College of Biology and FoodShangqiu Normal UniversityShangqiuHenanChina
- Key Laboratory on Agricultural Microorganism Resources Development of ShangqiuScience and Technology Bureau of Shangqiu CityShangqiuHenanChina
| | - Xuan Shao
- College of Biology and FoodShangqiu Normal UniversityShangqiuHenanChina
- Key Laboratory on Agricultural Microorganism Resources Development of ShangqiuScience and Technology Bureau of Shangqiu CityShangqiuHenanChina
| | - Jing Cao
- College of Biology and FoodShangqiu Normal UniversityShangqiuHenanChina
| | - Jie Huang
- College of Biology and FoodShangqiu Normal UniversityShangqiuHenanChina
| | - Jing Wang
- College of Biology and FoodShangqiu Normal UniversityShangqiuHenanChina
| | - Nuo Yang
- College of Biology and FoodShangqiu Normal UniversityShangqiuHenanChina
| | - Baodong Yuan
- College of Life ScienceLiaocheng UniversityLiaochengShandongChina
| |
Collapse
|
2
|
Smith K, Zubair RU, Smith RV, Augustine S, Schlecht NF, Ow TJ, Prystowsky MB, Belbin TJ. ZNF671 Silencing Affects Signaling Pathways in Head and Neck Cancer via Activation of Oncogenic Non-Coding RNAs. Biomedicines 2024; 12:2482. [PMID: 39595048 PMCID: PMC11592198 DOI: 10.3390/biomedicines12112482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Novel ZNF genes, such as ZNF671, that are located on chromosome 19q13 are known to be hypermethylated at a high frequency in HNSCC as well as in other epithelial solid tumors. Their function is largely unknown. RESULTS Here, we show that ZNF671 is epigenetically silenced in HNSCC primary tumors compared to matched adjacent normal tissue. Moreover, low expression of ZNF671 is significantly associated with decreased survival in HNSCC patients. Over-expression of ZNF671 in UM-SCC-1 oral cancer cells resulted in a significant reduction in tumor cell mobility and invasion compared to the empty-vector control cells. Transcriptomic analysis showed that ZNF671 re-expression resulted in a significant decrease in the expression of a major oncogenic long non-coding RNA LINC00665. CONCLUSIONS Together, these results suggest that epigenetic silencing of ZNF671 may activate multiple oncogenic signaling pathways via the resulting up-regulation of LINC00665.
Collapse
Affiliation(s)
- Kendra Smith
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada (R.U.Z.)
- Discipline of Oncology, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada
| | - Rufa’i Umar Zubair
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada (R.U.Z.)
| | - Richard V. Smith
- Department of Otorhinolaryngology—Head and Neck Surgery, Montefiore Med Center, 3400 Bainbridge Ave., Bronx, NY 10467, USA
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA;
| | - Stelby Augustine
- Department of Otorhinolaryngology—Head and Neck Surgery, Montefiore Med Center, 3400 Bainbridge Ave., Bronx, NY 10467, USA
| | - Nicholas F. Schlecht
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Thomas J. Ow
- Department of Otorhinolaryngology—Head and Neck Surgery, Montefiore Med Center, 3400 Bainbridge Ave., Bronx, NY 10467, USA
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA;
| | - Michael B. Prystowsky
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA;
| | - Thomas J. Belbin
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada (R.U.Z.)
- Discipline of Oncology, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6, Canada
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA;
| |
Collapse
|
3
|
Xiao N, Liu H, Zhang C, Chen H, Li Y, Yang Y, Liu H, Wan J. Applications of single-cell analysis in immunotherapy for lung cancer: Current progress, new challenges and expectations. J Adv Res 2024:S2090-1232(24)00462-4. [PMID: 39401694 DOI: 10.1016/j.jare.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 06/28/2024] [Accepted: 10/11/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Lung cancer is a prevalent form of cancer worldwide, presenting a substantial risk to human well-being. Lung cancer is classified into two main types: non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). The advancement of tumor immunotherapy, specifically immune checkpoint inhibitors and adaptive T-cell therapy, has encountered substantial obstacles due to the rapid progression of SCLC and the metastasis, recurrence, and drug resistance of NSCLC. These challenges are believed to stem from the tumor heterogeneity of lung cancer within the tumor microenvironment. AIM OF REVIEW This review aims to comprehensively explore recent strides in single-cell analysis, a robust sequencing technology, concerning its application in the realm of tumor immunotherapy for lung cancer. It has been effectively integrated with transcriptomics, epigenomics, genomics, and proteomics for various applications. Specifically, these techniques have proven valuable in mapping the transcriptional activity of tumor-infiltrating lymphocytes in patients with NSCLC, identifying circulating tumor cells, and elucidating the heterogeneity of the tumor microenvironment. KEY SCIENTIFIC CONCEPTS OF REVIEW The review emphasizes the paramount significance of single-cell analysis in mapping the immune cells within NSCLC patients, unveiling circulating tumor cells, and elucidating the tumor microenvironment heterogeneity. Notably, these advancements highlight the potential of single-cell analysis to revolutionize lung cancer immunotherapy by characterizing immune cell fates, improving therapeutic strategies, and identifying promising targets or prognostic biomarkers. It is potential to unravel the complexities within the tumor microenvironment and enhance treatment strategies marks a significant step towards more effective therapies and improved patient outcomes.
Collapse
Affiliation(s)
- Nan Xiao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hongyang Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Chenxing Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Huanxiang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yang Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ying Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hongchun Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Junhu Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
4
|
Ivancevic A, Simpson DM, Joyner OM, Bagby SM, Nguyen LL, Bitler BG, Pitts TM, Chuong EB. Endogenous retroviruses mediate transcriptional rewiring in response to oncogenic signaling in colorectal cancer. SCIENCE ADVANCES 2024; 10:eado1218. [PMID: 39018396 PMCID: PMC466953 DOI: 10.1126/sciadv.ado1218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/13/2024] [Indexed: 07/19/2024]
Abstract
Cancer cells exhibit rewired transcriptional regulatory networks that promote tumor growth and survival. However, the mechanisms underlying the formation of these pathological networks remain poorly understood. Through a pan-cancer epigenomic analysis, we found that primate-specific endogenous retroviruses (ERVs) are a rich source of enhancers displaying cancer-specific activity. In colorectal cancer and other epithelial tumors, oncogenic MAPK/AP1 signaling drives the activation of enhancers derived from the primate-specific ERV family LTR10. Functional studies in colorectal cancer cells revealed that LTR10 elements regulate tumor-specific expression of multiple genes associated with tumorigenesis, such as ATG12 and XRCC4. Within the human population, individual LTR10 elements exhibit germline and somatic structural variation resulting from a highly mutable internal tandem repeat region, which affects AP1 binding activity. Our findings reveal that ERV-derived enhancers contribute to transcriptional dysregulation in response to oncogenic signaling and shape the evolution of cancer-specific regulatory networks.
Collapse
Affiliation(s)
- Atma Ivancevic
- BioFrontiers Institute and Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - David M. Simpson
- BioFrontiers Institute and Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Olivia M. Joyner
- BioFrontiers Institute and Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Stacey M. Bagby
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lily L. Nguyen
- BioFrontiers Institute and Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ben G. Bitler
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Todd M. Pitts
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Edward B. Chuong
- BioFrontiers Institute and Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
5
|
Jiang YH, Liu YS, Wei YC, Jhang JF, Kuo HC, Huang HH, Chan MWY, Lin GL, Cheng WC, Lin SC, Wang HJ. Hypermethylation Loci of ZNF671, IRF8, and OTX1 as Potential Urine-Based Predictive Biomarkers for Bladder Cancer. Diagnostics (Basel) 2024; 14:468. [PMID: 38472940 DOI: 10.3390/diagnostics14050468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 03/14/2024] Open
Abstract
Bladder cancer (BCa) is a significant health issue and poses a healthcare burden on patients, highlighting the importance of an effective detection method. Here, we developed a urine DNA methylation diagnostic panel for distinguishing between BCa and non-BCa. In the discovery stage, an analysis of the TCGA database was conducted to identify BCa-specific DNA hypermethylation markers. In the validation phase, DNA methylation levels of urine samples were measured with real-time quantitative methylation-specific PCR (qMSP). Comparative analysis of the methylation levels between BCa and non-BCa, along with the receiver operating characteristic (ROC) analyses with machine learning algorithms (logistic regression and decision tree methods) were conducted to develop practical diagnostic panels. The performance evaluation of the panel shows that the individual biomarkers of ZNF671, OTX1, and IRF8 achieved AUCs of 0.86, 0.82, and 0.81, respectively, while the combined yielded an AUC of 0.91. The diagnostic panel using the decision tree algorithm attained an accuracy, sensitivity, and specificity of 82.6%, 75.0%, and 90.9%, respectively. Our results show that the urine-based DNA methylation diagnostic panel provides a sensitive and specific method for detecting and stratifying BCa, showing promise as a standard test that could enhance the diagnosis and prognosis of BCa in clinical settings.
Collapse
Affiliation(s)
- Yuan-Hong Jiang
- Department of Urology, Hualien Tzu Chi Hospital, Tzu Chi University, Hualien 970374, Taiwan
| | - Yu-Shu Liu
- Guzip Biomarkers Corporation, Hsinchu City 302041, Taiwan
- Phalanx Biotech, Hsinchu City 302041, Taiwan
| | - Yu-Chung Wei
- Graduate Institute of Statistics and Information Science, National Changhua University of Education, Changhua City 500207, Taiwan
| | - Jia-Fong Jhang
- Department of Urology, Hualien Tzu Chi Hospital, Tzu Chi University, Hualien 970374, Taiwan
| | - Hann-Chorng Kuo
- Department of Urology, Hualien Tzu Chi Hospital, Tzu Chi University, Hualien 970374, Taiwan
- Guzip Biomarkers Corporation, Hsinchu City 302041, Taiwan
| | - Hsin-Hui Huang
- Guzip Biomarkers Corporation, Hsinchu City 302041, Taiwan
- Phalanx Biotech, Hsinchu City 302041, Taiwan
| | - Michael W Y Chan
- Department of Biomedical Sciences, National Chung Cheng University, Minhsiung, Chiayi 621301, Taiwan
- Epigenomics and Human Disease Research Center, National Chung Cheng University, Minhsiung, Chiayi 621301, Taiwan
- Center for Innovative Research on Aging Society (CIRAS), National Chung Cheng University, Min-Hsiung, Chiayi 621301, Taiwan
| | - Guan-Ling Lin
- Department of Biomedical Sciences, National Chung Cheng University, Minhsiung, Chiayi 621301, Taiwan
- Epigenomics and Human Disease Research Center, National Chung Cheng University, Minhsiung, Chiayi 621301, Taiwan
- Center for Innovative Research on Aging Society (CIRAS), National Chung Cheng University, Min-Hsiung, Chiayi 621301, Taiwan
| | - Wen-Chi Cheng
- Institute of Medical Sciences, Tzu Chi University, Hualien 970374, Taiwan
| | - Shu-Chuan Lin
- Guzip Biomarkers Corporation, Hsinchu City 302041, Taiwan
- Phalanx Biotech, Hsinchu City 302041, Taiwan
| | - Hung-Jung Wang
- Institute of Medical Sciences, Tzu Chi University, Hualien 970374, Taiwan
- Doctoral Degree Program in Translational Medicine, Tzu Chi University and Academia Sinica, Hualien 97004, Taiwan
- Department of Biomedical Sciences and Engineering, Tzu Chi University, Hualien 970374, Taiwan
| |
Collapse
|
6
|
Song K, Artibani M. The role of DNA methylation in ovarian cancer chemoresistance: A narrative review. Health Sci Rep 2023; 6:e1235. [PMID: 37123549 PMCID: PMC10140645 DOI: 10.1002/hsr2.1235] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 05/02/2023] Open
Abstract
Background and Aims Ovarian cancer (OC) is the most lethal gynecological cancer. In 2018, it was responsible for over 180,000 deaths worldwide. The high mortality rate is the culmination of a lack of early diagnosis and high rates of chemotherapy resistance, which is synonymous with disease recurrence. Over the last two decades, an increasingly significant role of epigenetic mechanisms, in particular DNA methylation, has emerged. This review will discuss several of the most significant genes whose hypo/hypermethylation profiles are associated with chemoresistance. Aside from functionally elucidating and evaluating these epimutations, this review will discuss recent trials of DNA methyltransferase inhibitors (DNMTi). Finally, we will propose future directions that could enhance the feasibility of utilizing these candidate epimutations as clinical biomarkers. Methods To perform this review, a comprehensive literature search based on our keywords was conducted across the online databases PubMed and Google Scholar for identifying relevant studies published up until August 2022. Results Epimutations affecting MLH1, MSH2, and Ras-association domain family 1 isoform A (DNA damage repair and apoptosis); ATP-binding cassette subfamily B member 1 and methylation-controlled J (drug export); secreted frizzled-related proteins (Wnt/β-catenin signaling), neurocalcin delta (calcium and G protein-coupled receptor signaling), and zinc finger protein 671 all have potential as biomarkers for chemoresistance. However, specific uncertainties relating to these epimutations include histotype-specific differences, intrinsic versus acquired chemoresistance, and the interplay with complete surgical debulking. DNMTi for chemoresistant OC patients has shown some promise; however, issues surrounding their efficacy and dose-limiting toxicities remain; a personalized approach is required to maximize their effectiveness. Conclusion Establishing a panel of aberrantly methylated chemoresistance-related genes to predict chemoresponsiveness and patients' suitability to DNMTi could significantly reduce OC recurrence, while improving DNMTi therapy viability. To achieve this, a large-scale prospective genome-wide DNA methylation profile study that spans different histotypes, includes paired samples (before and after chemotherapy), and integrates transcriptomic and methylomic analysis, is warranted.
Collapse
Affiliation(s)
- Kaiyang Song
- Green Templeton CollegeUniversity of OxfordOxfordUK
| | - Mara Artibani
- Ovarian Cancer Cell Laboratory, Medical Research Council (MRC) Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
- Nuffield Department of Women's & Reproductive HealthUniversity of OxfordOxfordUK
| |
Collapse
|
7
|
Wang Y, Chen FR, Wei CC, Sun LL, Liu CY, Yang LB, Guo XY. Zinc finger protein 671 has a cancer-inhibiting function in colorectal carcinoma via the deactivation of Notch signaling. Toxicol Appl Pharmacol 2023; 458:116326. [PMID: 36436566 DOI: 10.1016/j.taap.2022.116326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
Zinc finger protein 671 (ZNF671) has been described as a vital cancer inhibitor in multiple neoplasms, yet the functional roles of ZNF671 in colorectal carcinoma (CRC) remain unresolved. This project examined the possible link between ZNF671 and CRC. Lower levels of ZNF671 were observed in CRC tissue compared with noncancerous tissue, which were related to a worse survival rate in CRC patients. High methylation levels at the ZNF671 gene promoter region were shown in CRC tissue, which were inversely correlated with ZNF671 expression. Treatment with demethylation agents restored ZNF671 levels in CRC cell lines. Up-regulation of ZNF671 resulted in suppressive effects on the proliferative ability and metastatic potency of CRC cells. Moreover, the up-regulation of ZNF671 reinforced the chemosensitivity of CRC cells. A mechanism study determined ZNF671 to be a vital mediator of Notch signaling. The up-regulation of ZNF671 decreased the expression of Notch1 and lowered the levels of NICD, HES1, and HEY1. The overexpression of NICD1 diminished ZNF671-mediated antitumor effects. ZNF671 depletion reinforced Notch signaling, and Notch suppression reversed ZNF671-depletion-elicited protumor effects. Moreover, the overexpression of ZNF671 weakened the tumorigenicity of CRC cells in a xenograft model in vivo. In summary, ZNF671 exerts a cancer-inhibiting function in CRC via the deactivation of Notch signaling. Low ZNF671 levels caused by gene promoter hypermethylation contribute to the malignant transformation of CRC. This work underlines the interest of ZNF671 as a target candidate for exploiting novel anti-CRC therapies.
Collapse
Affiliation(s)
- Yan Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Fen-Rong Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Chong-Cao Wei
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Lin-Lang Sun
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Chen-Yu Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Long-Bao Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| | - Xiao-Yan Guo
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| |
Collapse
|
8
|
Tulsyan S, Aftab M, Sisodiya S, Khan A, Chikara A, Tanwar P, Hussain S. Molecular basis of epigenetic regulation in cancer diagnosis and treatment. Front Genet 2022; 13:885635. [PMID: 36092905 PMCID: PMC9449878 DOI: 10.3389/fgene.2022.885635] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 07/19/2022] [Indexed: 02/01/2023] Open
Abstract
The global cancer cases and mortality rates are increasing and demand efficient biomarkers for accurate screening, detection, diagnosis, and prognosis. Recent studies have demonstrated that variations in epigenetic mechanisms like aberrant promoter methylation, altered histone modification and mutations in ATP-dependent chromatin remodelling complexes play an important role in the development of carcinogenic events. However, the influence of other epigenetic alterations in various cancers was confirmed with evolving research and the emergence of high throughput technologies. Therefore, alterations in epigenetic marks may have clinical utility as potential biomarkers for early cancer detection and diagnosis. In this review, an outline of the key epigenetic mechanism(s), and their deregulation in cancer etiology have been discussed to decipher the future prospects in cancer therapeutics including precision medicine. Also, this review attempts to highlight the gaps in epigenetic drug development with emphasis on integrative analysis of epigenetic biomarkers to establish minimally non-invasive biomarkers with clinical applications.
Collapse
Affiliation(s)
- Sonam Tulsyan
- Division of Cellular and Molecular Diagnostics (Molecular Biology Group), ICMR- National Institute of Cancer Prevention and Research, Noida, India
| | - Mehreen Aftab
- Division of Cellular and Molecular Diagnostics (Molecular Biology Group), ICMR- National Institute of Cancer Prevention and Research, Noida, India
| | - Sandeep Sisodiya
- Division of Cellular and Molecular Diagnostics (Molecular Biology Group), ICMR- National Institute of Cancer Prevention and Research, Noida, India
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Asiya Khan
- Laboratory Oncology Unit, Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Atul Chikara
- Division of Cellular and Molecular Diagnostics (Molecular Biology Group), ICMR- National Institute of Cancer Prevention and Research, Noida, India
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India
| | - Pranay Tanwar
- Laboratory Oncology Unit, Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
- *Correspondence: Showket Hussain, ; Pranay Tanwar,
| | - Showket Hussain
- Division of Cellular and Molecular Diagnostics (Molecular Biology Group), ICMR- National Institute of Cancer Prevention and Research, Noida, India
- *Correspondence: Showket Hussain, ; Pranay Tanwar,
| |
Collapse
|
9
|
Li J, Xu X, Liu C, Xi X, Wang Y, Wu X, Li H. miR-181a-2-3p Stimulates Gastric Cancer Progression via Targeting MYLK. Front Bioeng Biotechnol 2021; 9:687915. [PMID: 34733825 PMCID: PMC8558245 DOI: 10.3389/fbioe.2021.687915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/01/2021] [Indexed: 12/26/2022] Open
Abstract
Background: The abnormal expression of miRNAs facilitates tumorigenesis and development. miR-181a-2-3p is up-regulated in various cancers, yet its mechanism in gastric cancer (GC) remains elusive. Objective: To understand mechanism of miR-181a-2-3p stimulating GC cell progression via targeting Myosin Light Chain Kinase (MYLK) expression. Methods: Downstream genes of miRNA of interest were predicted in TargetScan and miRTarBase. qRT-PCR and western blot were applied to assess miR-181a-2-3p and MYLK expression in GC cells and normal cells. Dual-luciferase and RIP assays were completed to assess binding of miR-181a-2-3p and MYLK. Cell Counting Kit-8 (CCK-8) assay was conducted for detecting viability of AGS and SNU-1 cells, while Transwell tested migratory and invasive abilities of cells. Nude mouse transplantation tumor experiment was performed to assay tumor growth in vivo. Results: miR-181a-2-3p was notably increased in human GC cell lines, while MYLK was remarkably down-regulated. RIP and dual-luciferase assay disclosed that miR-181a-2-3p targeted MYLK and repressed MYLK. Forced miR-181a-2-3p expression fostered GC cell proliferation, invasion, migration, and fostered tumor growth in vivo. Promoting effect of miR-181a-2-3p on GC cells was reversed when miR-181a-2-3p and MYLK were simultaneously overexpressed. Conclusion: miR-181a-2-3p facilitated GC cell progression by targeting MYLK, and it may be a pivotal prognostic biomarker in investigating molecular mechanism of GC.
Collapse
Affiliation(s)
- Jianjie Li
- Department of Gastrointestinal Surgery, Tangshan Central Hospital, Tangshan, China
| | - Xiaoyue Xu
- Department of Gastrointestinal Surgery, Tangshan Gongren Hospital, Tangshan, China
| | - Chunhui Liu
- Department of General Surgery, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Xiaoxue Xi
- Department of Gastrointestinal Surgery, Tangshan Central Hospital, Tangshan, China
| | - Yang Wang
- Department of Gastrointestinal Surgery, Tangshan Central Hospital, Tangshan, China
| | - Xiaotang Wu
- Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, China
| | - Hua Li
- Department of Gastrointestinal Surgery, Tangshan Gongren Hospital, Tangshan, China
| |
Collapse
|
10
|
The Identification of Zinc-Finger Protein 433 as a Possible Prognostic Biomarker for Clear-Cell Renal Cell Carcinoma. Biomolecules 2021; 11:biom11081193. [PMID: 34439859 PMCID: PMC8392881 DOI: 10.3390/biom11081193] [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: 06/21/2021] [Revised: 07/31/2021] [Accepted: 08/06/2021] [Indexed: 11/18/2022] Open
Abstract
Clear-cell renal cell carcinoma (ccRCC) is the most common and aggressive form of all urological cancers, with poor prognosis and high mortality. At late stages, ccRCC is known to be mainly resistant to chemotherapy and radiotherapy. Therefore, it is urgent and necessary to identify biomarkers that can facilitate the early detection of ccRCC in patients. In this study, the levels of transcripts of ccRCC from The Cancer Genome Atlas (TCGA) dataset were used to identify prognostic biomarkers in this disease. Analyzing the data obtained indicated that the KRAB-ZNF protein is significantly suppressed in clear-cell carcinomas. Furthermore, ZNF433 is differentially expressed in ccRCC in a stage- and histological-grade-specific manner. In addition, ZNF433 expression was correlated with metastasis, with greater node involvement associated with lower ZNF433 expression (p < 0.01) and with a more unsatisfactory overall survival outcome (HR, 0.45; 95% CI, 0.33–0.6; p = 8.5 × 10−8). Since ccRCC is characterized by mutations in proteins that alter epigenetic modifications and /or chromatin remodeling, we examined the expression of ZNF433 transcripts in ccRCC with wildtype and mutated forms of BAP1, KDMC5, MTOR, PBRM1, SETD2, and VHL. Analysis revealed that ZNF433 expression was significantly reduced in ccRCC with mutations in the BAP1, SETD2, and KDM5C genes (p < 0.05). In addition, the ZNF433 promoter region was highly methylated, and hypermethylation was significantly associated with mRNA suppression (p < 2.2 × 10−16). In silico analysis of potential ZNF target genes found that the largest group of target genes are involved in cellular metabolic processes, which incidentally are particularly impaired in ccRCC. It was concluded from this study that gene expression of ZNF433 is associated with cancer progression and poorer prognosis, and that ZNF433 behaves in a manner that suggests that it is a prognostic marker and a possible tumor-suppressor gene in clear-cell renal cell carcinoma.
Collapse
|
11
|
Song X, Yang T, Zhang X, Yuan Y, Yan X, Wei Y, Zhang J, Zhou C. Comparison of the Microsatellite Distribution Patterns in the Genomes of Euarchontoglires at the Taxonomic Level. Front Genet 2021; 12:622724. [PMID: 33719337 PMCID: PMC7953163 DOI: 10.3389/fgene.2021.622724] [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: 10/29/2020] [Accepted: 02/05/2021] [Indexed: 02/05/2023] Open
Abstract
Microsatellite or simple sequence repeat (SSR) instability within genes can induce genetic variation. The SSR signatures remain largely unknown in different clades within Euarchontoglires, one of the most successful mammalian radiations. Here, we conducted a genome-wide characterization of microsatellite distribution patterns at different taxonomic levels in 153 Euarchontoglires genomes. Our results showed that the abundance and density of the SSRs were significantly positively correlated with primate genome size, but no significant relationship with the genome size of rodents was found. Furthermore, a higher level of complexity for perfect SSR (P-SSR) attributes was observed in rodents than in primates. The most frequent type of P-SSR was the mononucleotide P-SSR in the genomes of primates, tree shrews, and colugos, while mononucleotide or dinucleotide motif types were dominant in the genomes of rodents and lagomorphs. Furthermore, (A)n was the most abundant motif in primate genomes, but (A)n, (AC)n, or (AG)n was the most abundant motif in rodent genomes which even varied within the same genus. The GC content and the repeat copy numbers of P-SSRs varied in different species when compared at different taxonomic levels, reflecting underlying differences in SSR mutation processes. Notably, the CDSs containing P-SSRs were categorized by functions and pathways using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes annotations, highlighting their roles in transcription regulation. Generally, this work will aid future studies of the functional roles of the taxonomic features of microsatellites during the evolution of mammals in Euarchontoglires.
Collapse
Affiliation(s)
- Xuhao Song
- Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, Nanchong, China.,Institute of Ecology, China West Normal University, Nanchong, China
| | - Tingbang Yang
- Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, Nanchong, China.,Institute of Ecology, China West Normal University, Nanchong, China
| | - Xinyi Zhang
- Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, Nanchong, China
| | - Ying Yuan
- Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, Nanchong, China
| | - Xianghui Yan
- Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, Nanchong, China
| | - Yi Wei
- Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, Nanchong, China.,Institute of Ecology, China West Normal University, Nanchong, China
| | - Jun Zhang
- Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, Nanchong, China.,Institute of Ecology, China West Normal University, Nanchong, China
| | - Caiquan Zhou
- Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, Nanchong, China.,Institute of Ecology, China West Normal University, Nanchong, China
| |
Collapse
|
12
|
Balogh A, Reiniger L, Hetey S, Kiraly P, Toth E, Karaszi K, Juhasz K, Gelencser Z, Zvara A, Szilagyi A, Puskas LG, Matko J, Papp Z, Kovalszky I, Juhasz C, Than NG. Decreased Expression of ZNF554 in Gliomas is Associated with the Activation of Tumor Pathways and Shorter Patient Survival. Int J Mol Sci 2020; 21:E5762. [PMID: 32796700 PMCID: PMC7461028 DOI: 10.3390/ijms21165762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 01/01/2023] Open
Abstract
Zinc finger protein 554 (ZNF554), a member of the Krüppel-associated box domain zinc finger protein subfamily, is predominantly expressed in the brain and placenta in humans. Recently, we unveiled that ZNF554 regulates trophoblast invasion during placentation and its decreased expression leads to the early pathogenesis of preeclampsia. Since ZNF proteins are immensely implicated in the development of several tumors including malignant tumors of the brain, here we explored the pathological role of ZNF554 in gliomas. We examined the expression of ZNF554 at mRNA and protein levels in normal brain and gliomas, and then we searched for genome-wide transcriptomic changes in U87 glioblastoma cells transiently overexpressing ZNF554. Immunohistochemistry of brain tissues in our cohort (n = 62) and analysis of large TCGA RNA-Seq data (n = 687) of control, oligodendroglioma, and astrocytoma tissues both revealed decreased expression of ZNF554 towards higher glioma grades. Furthermore, low ZNF554 expression was associated with shorter survival of grade III and IV astrocytoma patients. Overexpression of ZNF554 in U87 cells resulted in differential expression, mostly downregulation of 899 genes. The "PI3K-Akt signaling pathway", known to be activated during glioma development, was the most impacted among 116 dysregulated pathways. Most affected pathways were cancer-related and/or immune-related. Congruently, cell proliferation was decreased and cell cycle was arrested in ZNF554-transfected glioma cells. These data collectively suggest that ZNF554 is a potential tumor suppressor and its decreased expression may lead to the loss of oncogene suppression, activation of tumor pathways, and shorter survival of patients with malignant glioma.
Collapse
Affiliation(s)
- Andrea Balogh
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.B.); (S.H.); (P.K.); (E.T.); (K.K.); (K.J.); (Z.G.); (A.S.)
| | - Lilla Reiniger
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary; (L.R.); (I.K.)
| | - Szabolcs Hetey
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.B.); (S.H.); (P.K.); (E.T.); (K.K.); (K.J.); (Z.G.); (A.S.)
| | - Peter Kiraly
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.B.); (S.H.); (P.K.); (E.T.); (K.K.); (K.J.); (Z.G.); (A.S.)
| | - Eszter Toth
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.B.); (S.H.); (P.K.); (E.T.); (K.K.); (K.J.); (Z.G.); (A.S.)
| | - Katalin Karaszi
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.B.); (S.H.); (P.K.); (E.T.); (K.K.); (K.J.); (Z.G.); (A.S.)
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary; (L.R.); (I.K.)
| | - Kata Juhasz
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.B.); (S.H.); (P.K.); (E.T.); (K.K.); (K.J.); (Z.G.); (A.S.)
| | - Zsolt Gelencser
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.B.); (S.H.); (P.K.); (E.T.); (K.K.); (K.J.); (Z.G.); (A.S.)
| | - Agnes Zvara
- Laboratory of Functional Genomics, Department of Genetics, Biological Research Centre, H-6726 Szeged, Hungary; (A.Z.); (L.G.P.)
| | - Andras Szilagyi
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.B.); (S.H.); (P.K.); (E.T.); (K.K.); (K.J.); (Z.G.); (A.S.)
| | - Laszlo G. Puskas
- Laboratory of Functional Genomics, Department of Genetics, Biological Research Centre, H-6726 Szeged, Hungary; (A.Z.); (L.G.P.)
| | - Janos Matko
- Department of Immunology, Eotvos Lorand University, H-1117 Budapest, Hungary;
| | - Zoltan Papp
- Maternity Private Clinic, H-1126 Budapest, Hungary;
- Department of Obstetrics and Gynecology, Semmelweis University, H-1088 Budapest, Hungary
| | - Ilona Kovalszky
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary; (L.R.); (I.K.)
| | - Csaba Juhasz
- Department of Pediatrics, Neurology, Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Barbara Ann Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Nandor Gabor Than
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.B.); (S.H.); (P.K.); (E.T.); (K.K.); (K.J.); (Z.G.); (A.S.)
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary; (L.R.); (I.K.)
- Maternity Private Clinic, H-1126 Budapest, Hungary;
| |
Collapse
|