Background: The objective was to elucidate the potential epigenetic regulatory mechanism in HMOX1 expression in preeclampsia. Materials & methods:HMOX1 promoter DNA methylation was evaluated in the placental tissue and blood of preeclamptic and normotensive pregnant women. HMOX1 and miR-153-3p gene expression were assessed in placental tissue and peripheral blood mononuclear cells (PBMCs). Related microarray datasets in the Gene Expression Omnibus database were also analyzed. Results: In placental tissue, despite HMOX1 expression downregulation, there was no significant change in HMOX1 methylation. In PBMCs, there was no significant alteration in HMOX1 expression, while hypomethylation was observed in blood. The miR-153-3p expression increased in the placental tissue and in the PBMCs of preeclampsia. Conclusion: DNA methylation does not affect HMOX1 expression, while miR-153-3p might be a biomarker for preeclampsia.
{"title":"The study of <i>HMOX1</i> DNA methylation and gene expression and the diagnostic potential of <i>miR-153-3p</i> in preeclampsia.","authors":"Somayeh Rahimi, Nayebali Rezvani, Saeed Khazayel, Nazanin Jalilian, Ebrahim Shakiba, Fatemeh Khadir, Kheirollah Yari, Zohreh Rahimi","doi":"10.2217/epi-2023-0377","DOIUrl":"10.2217/epi-2023-0377","url":null,"abstract":"<p><p><b>Background:</b> The objective was to elucidate the potential epigenetic regulatory mechanism in <i>HMOX1</i> expression in preeclampsia. <b>Materials & methods:</b> <i>HMOX1</i> promoter DNA methylation was evaluated in the placental tissue and blood of preeclamptic and normotensive pregnant women. <i>HMOX1</i> and <i>miR-153-3p</i> gene expression were assessed in placental tissue and peripheral blood mononuclear cells (PBMCs). Related microarray datasets in the Gene Expression Omnibus database were also analyzed. <b>Results:</b> In placental tissue, despite <i>HMOX1</i> expression downregulation, there was no significant change in <i>HMOX1</i> methylation. In PBMCs, there was no significant alteration in <i>HMOX1</i> expression, while hypomethylation was observed in blood. The <i>miR-153-3p</i> expression increased in the placental tissue and in the PBMCs of preeclampsia. <b>Conclusion:</b> DNA methylation does not affect <i>HMOX1</i> expression, while <i>miR-153-3p</i> might be a biomarker for preeclampsia.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"389-401"},"PeriodicalIF":3.8,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139971501","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: The objective of this research was to determine whether HDAC2 function is associated with gastric cancer progression. Methods: HDAC2 was knocked out in EPG85.257 cells using CRISPR/Cas9 and tumorigenesis pathways were evaluated. Results: Cell proliferation, colony formation, wound healing and transwell invasion were inhibited in ΔHDAC2:EPG85.257 cells. Quantitative analyses revealed a significant downregulation of MMP1, p53, Bax, MAPK1, MAPK3, pro-Caspase3, ERK1/2, p-ERK1/2, AKT1/2/3, p-AKT1/2/3, p-NF-κB (p65), Twist, Snail and p-FAK transcripts/proteins, while SIRT1, PTEN, p21 and Caspase3 were upregulated in ΔHDAC2:EPG85.257 cells. Conclusion: These results indicated that HDAC2 enhanced migration, colony formation and transmigration ability. HDAC2 inhibition may improve gastric cancer chemotherapy pathways.
{"title":"Epigenetic disruption of histone deacetylase-2 accelerated apoptotic signaling and retarded malignancy in gastric cells.","authors":"Sayedeh Azimeh Hosseini, Mahdi Ghatrehsamani, Hajar Yaghoobi, Fatemeh Elahian, Seyed Abbas Mirzaei","doi":"10.2217/epi-2023-0350","DOIUrl":"10.2217/epi-2023-0350","url":null,"abstract":"<p><p><b>Background:</b> The objective of this research was to determine whether HDAC2 function is associated with gastric cancer progression. <b>Methods:</b> HDAC2 was knocked out in EPG85.257 cells using CRISPR/Cas9 and tumorigenesis pathways were evaluated. <b>Results:</b> Cell proliferation, colony formation, wound healing and transwell invasion were inhibited in ΔHDAC2:EPG85.257 cells. Quantitative analyses revealed a significant downregulation of MMP1, p53, Bax, MAPK1, MAPK3, pro-Caspase3, ERK1/2, p-ERK1/2, AKT1/2/3, p-AKT1/2/3, p-NF-κB (p65), Twist, Snail and p-FAK transcripts/proteins, while SIRT1, PTEN, p21 and Caspase3 were upregulated in ΔHDAC2:EPG85.257 cells. <b>Conclusion:</b> These results indicated that HDAC2 enhanced migration, colony formation and transmigration ability. HDAC2 inhibition may improve gastric cancer chemotherapy pathways.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"277-292"},"PeriodicalIF":3.8,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139734834","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-03-01Epub Date: 2024-02-15DOI: 10.2217/epi-2023-0339
Meisam Bagheri, Min Kyung Lee, Kristen E Muller, Todd W Miller, Diwakar R Pattabiraman, Brock C Christensen
Background: Triple-negative breast cancer (TNBC) is an aggressive disease with limited treatment options. Eribulin, a chemotherapeutic drug, induces epigenetic changes in cancer cells, suggesting a unique mechanism of action. Materials & methods: MDA-MB 231 cells were treated with eribulin and paclitaxel, and the samples from 53 patients treated with neoadjuvant eribulin were compared with those from 14 patients who received the standard-of-care treatment using immunohistochemistry. Results: Eribulin treatment caused significant DNA methylation changes in drug-tolerant persister TNBC cells, and it also elicited changes in the expression levels of epigenetic modifiers (DNMT1, TET1, DNMT3A/B) in vitro and in primary TNBC tumors. Conclusion: These findings provide new insights into eribulin's mechanism of action and potential biomarkers for predicting TNBC treatment response.
{"title":"Alteration of DNA methyltransferases by eribulin elicits broad DNA methylation changes with potential therapeutic implications for triple-negative breast cancer.","authors":"Meisam Bagheri, Min Kyung Lee, Kristen E Muller, Todd W Miller, Diwakar R Pattabiraman, Brock C Christensen","doi":"10.2217/epi-2023-0339","DOIUrl":"10.2217/epi-2023-0339","url":null,"abstract":"<p><p><b>Background:</b> Triple-negative breast cancer (TNBC) is an aggressive disease with limited treatment options. Eribulin, a chemotherapeutic drug, induces epigenetic changes in cancer cells, suggesting a unique mechanism of action. <b>Materials & methods:</b> MDA-MB 231 cells were treated with eribulin and paclitaxel, and the samples from 53 patients treated with neoadjuvant eribulin were compared with those from 14 patients who received the standard-of-care treatment using immunohistochemistry. <b>Results:</b> Eribulin treatment caused significant DNA methylation changes in drug-tolerant persister TNBC cells, and it also elicited changes in the expression levels of epigenetic modifiers (DNMT1, TET1, DNMT3A/B) <i>in vitro</i> and in primary TNBC tumors. <b>Conclusion:</b> These findings provide new insights into eribulin's mechanism of action and potential biomarkers for predicting TNBC treatment response.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"293-308"},"PeriodicalIF":3.8,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10910603/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139734833","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-03-01Epub Date: 2024-02-15DOI: 10.2217/epi-2023-0388
Lele Ye, Zhuofeng Jiang, Mengxia Zheng, Kan Pan, Jingru Lian, Bing Ju, Xuefei Liu, Sangsang Tang, Gangqiang Guo, Songfa Zhang, Xin Hong, Weiguo Lu
Background: To explore the role of fatty acid metabolism (FAM)-related lncRNAs in the prognosis and antitumor immunity of serous ovarian cancer (SOC). Materials & methods: A SOC FAM-related lncRNA risk model was developed and evaluated by a series of analyses. Additional immune-related analyses were performed to further assess the associations between immune state, tumor microenvironment and the prognostic risk model. Results: Five lncRNAs associated with the FAM genes were found and used to create a predictive risk model. The patients with a low-risk profile exhibited favorable prognostic outcomes. Conclusion: The established prognostic risk model exhibits better predictive capabilities for the prognosis of patients with SOC and offers novel potential therapy targets for SOC.
{"title":"Fatty acid metabolism-related lncRNA prognostic signature for serous ovarian carcinoma.","authors":"Lele Ye, Zhuofeng Jiang, Mengxia Zheng, Kan Pan, Jingru Lian, Bing Ju, Xuefei Liu, Sangsang Tang, Gangqiang Guo, Songfa Zhang, Xin Hong, Weiguo Lu","doi":"10.2217/epi-2023-0388","DOIUrl":"10.2217/epi-2023-0388","url":null,"abstract":"<p><p><b>Background:</b> To explore the role of fatty acid metabolism (FAM)-related lncRNAs in the prognosis and antitumor immunity of serous ovarian cancer (SOC). <b>Materials & methods:</b> A SOC FAM-related lncRNA risk model was developed and evaluated by a series of analyses. Additional immune-related analyses were performed to further assess the associations between immune state, tumor microenvironment and the prognostic risk model. <b>Results:</b> Five lncRNAs associated with the FAM genes were found and used to create a predictive risk model. The patients with a low-risk profile exhibited favorable prognostic outcomes. <b>Conclusion:</b> The established prognostic risk model exhibits better predictive capabilities for the prognosis of patients with SOC and offers novel potential therapy targets for SOC.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"309-329"},"PeriodicalIF":3.8,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139734835","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-03-01Epub Date: 2024-03-05DOI: 10.2217/epi-2023-0405
Ramisetti Rajeev, Rakesh K Mishra, Sanjeev Khosla
Aim: To explore the role of Piwi protein and piRNAs in DNMT3L-mediated epigenetic inheritance. Materials & methods: Transgenic Drosophila were used to examine the effect of ectopically expressed DNMT3L on the profile of piRNAs by sequencing of small RNAs. Results & conclusion: Our previous work showed accumulation and inheritance of epimutations across multiple generations in transgenic DNMT3L Drosophila. Here, we show interaction of DNMT3L with Piwi and a significant alteration in the piRNA profile across multiple generations in transgenic Drosophila. In the light of its interaction with histone H1, we propose that in addition to its role of modulating core histone modifications, DNMT3L allows for inheritance of epigenetic information through its collaboration with Piwi, piRNAs and histone H1.
{"title":"DNMT3L interacts with Piwi and modulates the expression of piRNAs in transgenic <i>Drosophila</i>.","authors":"Ramisetti Rajeev, Rakesh K Mishra, Sanjeev Khosla","doi":"10.2217/epi-2023-0405","DOIUrl":"10.2217/epi-2023-0405","url":null,"abstract":"<p><p><b>Aim:</b> To explore the role of Piwi protein and piRNAs in DNMT3L-mediated epigenetic inheritance. <b>Materials & methods:</b> Transgenic <i>Drosophila</i> were used to examine the effect of ectopically expressed DNMT3L on the profile of piRNAs by sequencing of small RNAs. <b>Results & conclusion:</b> Our previous work showed accumulation and inheritance of epimutations across multiple generations in transgenic DNMT3L <i>Drosophila</i>. Here, we show interaction of DNMT3L with Piwi and a significant alteration in the piRNA profile across multiple generations in transgenic <i>Drosophila</i>. In the light of its interaction with histone H1, we propose that in addition to its role of modulating core histone modifications, DNMT3L allows for inheritance of epigenetic information through its collaboration with Piwi, piRNAs and histone H1.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"375-388"},"PeriodicalIF":3.8,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140027808","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-03-01Epub Date: 2024-02-07DOI: 10.2217/epi-2023-0428
Balapal S Basavarajappa, Shivakumar Subbanna
Recent genomics and epigenetic advances have empowered the exploration of DNA/RNA methylation and histone modifications crucial for gene expression in response to stress, aging and disease. Interest in understanding neuronal plasticity's epigenetic mechanisms, influencing brain rewiring amid development, aging and neurodegenerative disorders, continues to grow. Histone acetylation dysregulation, a commonality in diverse brain disorders, has become a therapeutic focus. Histone acetyltransferases and histone deacetylases have emerged as promising targets for neurodegenerative disorder treatment. This review delves into histone acetylation regulation, potential therapies and future perspectives for disorders like Alzheimer's, Parkinson's and Huntington's. Exploring genetic-environmental interplay through models and studies reveals molecular changes, behavioral insights and early intervention possibilities targeting the epigenome in at-risk individuals.
{"title":"Unlocking the epigenetic symphony: histone acetylation's impact on neurobehavioral change in neurodegenerative disorders.","authors":"Balapal S Basavarajappa, Shivakumar Subbanna","doi":"10.2217/epi-2023-0428","DOIUrl":"10.2217/epi-2023-0428","url":null,"abstract":"<p><p>Recent genomics and epigenetic advances have empowered the exploration of DNA/RNA methylation and histone modifications crucial for gene expression in response to stress, aging and disease. Interest in understanding neuronal plasticity's epigenetic mechanisms, influencing brain rewiring amid development, aging and neurodegenerative disorders, continues to grow. Histone acetylation dysregulation, a commonality in diverse brain disorders, has become a therapeutic focus. Histone acetyltransferases and histone deacetylases have emerged as promising targets for neurodegenerative disorder treatment. This review delves into histone acetylation regulation, potential therapies and future perspectives for disorders like Alzheimer's, Parkinson's and Huntington's. Exploring genetic-environmental interplay through models and studies reveals molecular changes, behavioral insights and early intervention possibilities targeting the epigenome in at-risk individuals.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"331-358"},"PeriodicalIF":3.8,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10910622/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139697212","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-03-01Epub Date: 2024-02-27DOI: 10.2217/epi-2023-0382
Monu Pandey, Jayanta Bhattacharyya
Colorectal cancer (CRC) is a leading cause of cancer-related deaths worldwide. The occurrence of CRC is associated with various genetic and epigenetic mutations in intestinal epithelial cells that transform them into adenocarcinomas. There is increasing evidence indicating the gut microbiota plays a crucial role in the regulation of host physiological processes. Alterations in gut microbiota composition are responsible for initiating carcinogenesis through diverse epigenetic modifications, including histone modifications, ncRNAs and DNA methylation. This work was designed to comprehensively review recent findings to provide insight into the associations between the gut microbiota and CRC at an epigenetic level. These scientific insights can be used in the future to develop effective strategies for early detection and treatment of CRC.
结肠直肠癌(CRC)是全球癌症相关死亡的主要原因。CRC 的发生与肠道上皮细胞的各种基因和表观遗传突变有关,这些突变会使其转化为腺癌。越来越多的证据表明,肠道微生物群在调节宿主生理过程中发挥着至关重要的作用。肠道微生物群组成的改变可通过组蛋白修饰、ncRNA 和 DNA 甲基化等多种表观遗传修饰引发癌变。这项工作旨在全面回顾最近的研究结果,从表观遗传学层面深入探讨肠道微生物群与 CRC 之间的关联。这些科学见解可用于未来制定早期检测和治疗 CRC 的有效策略。
{"title":"Gut microbiota and epigenetics in colorectal cancer: implications for carcinogenesis and therapeutic intervention.","authors":"Monu Pandey, Jayanta Bhattacharyya","doi":"10.2217/epi-2023-0382","DOIUrl":"10.2217/epi-2023-0382","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is a leading cause of cancer-related deaths worldwide. The occurrence of CRC is associated with various genetic and epigenetic mutations in intestinal epithelial cells that transform them into adenocarcinomas. There is increasing evidence indicating the gut microbiota plays a crucial role in the regulation of host physiological processes. Alterations in gut microbiota composition are responsible for initiating carcinogenesis through diverse epigenetic modifications, including histone modifications, ncRNAs and DNA methylation. This work was designed to comprehensively review recent findings to provide insight into the associations between the gut microbiota and CRC at an epigenetic level. These scientific insights can be used in the future to develop effective strategies for early detection and treatment of CRC.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"403-418"},"PeriodicalIF":3.8,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139971499","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-03-01Epub Date: 2024-03-05DOI: 10.2217/epi-2023-0381
Annunziata G Cicatiello, Annarita Nappi, Fabiana Franchini, Immacolata C Nettore, Maddalena Raia, Carmine Rocca, Tommaso Angelone, Monica Dentice, Paola Ungaro, Paolo E Macchia
Aim: To study the expression of histone methyltransferase SMYD1 in white adipose tissue (WAT) and brown adipose tissue and during differentiation of preadipocytes to white and beige phenotypes. Methods: C57BL/6J mice fed a high-fat diet (and exposed to cold) and 3T3-L1 cells stimulated to differentiate into white and beige adipocytes were used. Results: SMYD1 expression increased in WAT of high-fat diet fed mice and in WAT and brown adipose tissue of cold-exposed mice, suggesting its role in thermogenesis. SMYD1 expression was higher in beige adipocytes than in white adipocytes, and its silencing leads to a decrease in mitochondrial content and in Pgc-1α expression. Conclusion: These data suggest a novel role for SMYD1 as a positive regulator of energy control in adipose tissue.
{"title":"The histone methyltransferase SMYD1 is induced by thermogenic stimuli in adipose tissue.","authors":"Annunziata G Cicatiello, Annarita Nappi, Fabiana Franchini, Immacolata C Nettore, Maddalena Raia, Carmine Rocca, Tommaso Angelone, Monica Dentice, Paola Ungaro, Paolo E Macchia","doi":"10.2217/epi-2023-0381","DOIUrl":"10.2217/epi-2023-0381","url":null,"abstract":"<p><p><b>Aim:</b> To study the expression of histone methyltransferase SMYD1 in white adipose tissue (WAT) and brown adipose tissue and during differentiation of preadipocytes to white and beige phenotypes. <b>Methods:</b> C57BL/6J mice fed a high-fat diet (and exposed to cold) and 3T3-L1 cells stimulated to differentiate into white and beige adipocytes were used. <b>Results:</b> SMYD1 expression increased in WAT of high-fat diet fed mice and in WAT and brown adipose tissue of cold-exposed mice, suggesting its role in thermogenesis. SMYD1 expression was higher in beige adipocytes than in white adipocytes, and its silencing leads to a decrease in mitochondrial content and in <i>Pgc-1α</i> expression. <b>Conclusion:</b> These data suggest a novel role for SMYD1 as a positive regulator of energy control in adipose tissue.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"359-374"},"PeriodicalIF":3.8,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140027809","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-03-01Epub Date: 2024-02-05DOI: 10.2217/epi-2023-0321
Shota Nishitani, Alicia K Smith, Akemi Tomoda, Takashi X Fujisawa
Tweetable abstract This article reviews machine learning models that leverages epigenomic data for predicting multifactorial diseases and symptoms as well as how such models can be utilized to explore new research questions.
{"title":"Data science using the human epigenome for predicting multifactorial diseases and symptoms.","authors":"Shota Nishitani, Alicia K Smith, Akemi Tomoda, Takashi X Fujisawa","doi":"10.2217/epi-2023-0321","DOIUrl":"10.2217/epi-2023-0321","url":null,"abstract":"<p><p>Tweetable abstract This article reviews machine learning models that leverages epigenomic data for predicting multifactorial diseases and symptoms as well as how such models can be utilized to explore new research questions.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"273-276"},"PeriodicalIF":3.8,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139680903","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-02-01Epub Date: 2024-01-29DOI: 10.2217/epi-2023-0213
Basireh Bahrami, Markus Wolfien, Parvaneh Nikpour
Background: Enhancer RNAs (eRNAs) are involved in gene expression regulation. Although functional roles of eRNAs in the pathophysiology of neoplasms have been reported, their involvement in gastric cancer (GC) is less known. Materials & methods: A network-based integrative approach was utilized for analyzing transcriptome and epigenome alterations in GC, and an eRNA was selected for experimental validation. Survival analysis and clinicopathological associations were also performed. Results: A hub eRNA, ENSR00000272060, showed significantly increased expression in tumor versus nontumor tissues, as well as an association with clinicopathological features. A seven-gene prognostic model was also constructed. Conclusion: The constructed network provides a comprehensive understanding of the underlying processes implicated in the progression of GC, along with a starting point from which to derive potential diagnostic/prognostic biomarkers.
{"title":"Integrated analysis of transcriptome and epigenome reveals <i>ENSR00000272060</i> as a potential biomarker in gastric cancer.","authors":"Basireh Bahrami, Markus Wolfien, Parvaneh Nikpour","doi":"10.2217/epi-2023-0213","DOIUrl":"10.2217/epi-2023-0213","url":null,"abstract":"<p><p><b>Background:</b> Enhancer RNAs (eRNAs) are involved in gene expression regulation. Although functional roles of eRNAs in the pathophysiology of neoplasms have been reported, their involvement in gastric cancer (GC) is less known. <b>Materials & methods:</b> A network-based integrative approach was utilized for analyzing transcriptome and epigenome alterations in GC, and an eRNA was selected for experimental validation. Survival analysis and clinicopathological associations were also performed. <b>Results:</b> A hub eRNA, <i>ENSR00000272060</i>, showed significantly increased expression in tumor versus nontumor tissues, as well as an association with clinicopathological features. A seven-gene prognostic model was also constructed. <b>Conclusion:</b> The constructed network provides a comprehensive understanding of the underlying processes implicated in the progression of GC, along with a starting point from which to derive potential diagnostic/prognostic biomarkers.</p>","PeriodicalId":11959,"journal":{"name":"Epigenomics","volume":" ","pages":"159-173"},"PeriodicalIF":3.8,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139570148","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}