357 results on '"super-enhancers"'
Search Results
2. CASCADES, a novel SOX2 super‐enhancer‐associated long noncoding RNA, regulates cancer stem cell specification and differentiation in glioblastoma.
- Author
-
Shahzad, Uswa, Nikolopoulos, Marina, Li, Christopher, Johnston, Michael, Wang, Jenny J., Sabha, Nesrin, Varn, Frederick S., Riemenschneider, Alexandra, Krumholtz, Stacey, Krishnamurthy, Pranathi Meda, Smith, Christian A., Karamchandani, Jason, Watts, Jonathan K., Verhaak, Roel G. W., Gallo, Marco, Rutka, James T., and Das, Sunit
- Subjects
- *
LINCRNA , *CANCER stem cells , *BRAIN tumors , *ISOCITRATE dehydrogenase , *CELL differentiation - Abstract
Glioblastoma is the most common primary malignant brain tumor in adults, with a median survival of just over 1 year. The failure of available treatments to achieve remission in patients with glioblastoma (GBM) has been attributed to the presence of cancer stem cells (CSCs), which are thought to play a central role in tumor development and progression and serve as a treatment‐resistant cell repository capable of driving tumor recurrence. In fact, the property of “stemness” itself may be responsible for treatment resistance. In this study, we identify a novel long noncoding RNA (lncRNA), cancer stem cell‐associated distal enhancer of SOX2 (CASCADES), that functions as an epigenetic regulator in glioma CSCs (GSCs). CASCADES is expressed in isocitrate dehydrogenase (IDH)‐wild‐type GBM and is significantly enriched in GSCs. Knockdown of CASCADES in GSCs results in differentiation towards a neuronal lineage in a cell‐ and cancer‐specific manner. Bioinformatics analysis reveals that CASCADES functions as a super‐enhancer‐associated lncRNA epigenetic regulator of SOX2. Our findings identify CASCADES as a critical regulator of stemness in GSCs that represents a novel epigenetic and therapeutic target for disrupting the CSC compartment in glioblastoma. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Effects of super-enhancers in cancer metastasis: mechanisms and therapeutic targets
- Author
-
Shenglan Liu, Wei Dai, Bei Jin, Feng Jiang, Hao Huang, Wen Hou, Jinxia Lan, Yanli Jin, Weijie Peng, and Jingxuan Pan
- Subjects
Super-enhancers ,Metastasis ,Molecular mechanisms ,Therapeutic targets ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Metastasis remains the principal cause of cancer-related lethality despite advancements in cancer treatment. Dysfunctional epigenetic alterations are crucial in the metastatic cascade. Among these, super-enhancers (SEs), emerging as new epigenetic regulators, consist of large clusters of regulatory elements that drive the high-level expression of genes essential for the oncogenic process, upon which cancer cells develop a profound dependency. These SE-driven oncogenes play an important role in regulating various facets of metastasis, including the promotion of tumor proliferation in primary and distal metastatic organs, facilitating cellular migration and invasion into the vasculature, triggering epithelial-mesenchymal transition, enhancing cancer stem cell-like properties, circumventing immune detection, and adapting to the heterogeneity of metastatic niches. This heavy reliance on SE-mediated transcription delineates a vulnerable target for therapeutic intervention in cancer cells. In this article, we review current insights into the characteristics, identification methodologies, formation, and activation mechanisms of SEs. We also elaborate the oncogenic roles and regulatory functions of SEs in the context of cancer metastasis. Ultimately, we discuss the potential of SEs as novel therapeutic targets and their implications in clinical oncology, offering insights into future directions for innovative cancer treatment strategies.
- Published
- 2024
- Full Text
- View/download PDF
4. Mechanism of ERBB2 gene overexpression by the formation of super-enhancer with genomic structural abnormalities in lung adenocarcinoma without clinically actionable genetic alterations
- Author
-
Syuzo Kaneko, Ken Takasawa, Ken Asada, Kouya Shiraishi, Noriko Ikawa, Hidenori Machino, Norio Shinkai, Maiko Matsuda, Mari Masuda, Shungo Adachi, Satoshi Takahashi, Kazuma Kobayashi, Nobuji Kouno, Amina Bolatkan, Masaaki Komatsu, Masayoshi Yamada, Mototaka Miyake, Hirokazu Watanabe, Akiko Tateishi, Takaaki Mizuno, Yu Okubo, Masami Mukai, Tatsuya Yoshida, Yukihiro Yoshida, Hidehito Horinouchi, Shun-Ichi Watanabe, Yuichiro Ohe, Yasushi Yatabe, Vassiliki Saloura, Takashi Kohno, and Ryuji Hamamoto
- Subjects
Lung adenocarcinoma ,Super-enhancers ,Structural variations ,Targeted therapy ,Driver mutations ,Integrated analysis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background In an extensive genomic analysis of lung adenocarcinomas (LUADs), driver mutations have been recognized as potential targets for molecular therapy. However, there remain cases where target genes are not identified. Super-enhancers and structural variants are frequently identified in several hundred loci per case. Despite this, most cancer research has approached the analysis of these data sets separately, without merging and comparing the data, and there are no examples of integrated analysis in LUAD. Methods We performed an integrated analysis of super-enhancers and structural variants in a cohort of 174 LUAD cases that lacked clinically actionable genetic alterations. To achieve this, we conducted both WGS and H3K27Ac ChIP-seq analyses using samples with driver gene mutations and those without, allowing for a comprehensive investigation of the potential roles of super-enhancer in LUAD cases. Results We demonstrate that most genes situated in these overlapped regions were associated with known and previously unknown driver genes and aberrant expression resulting from the formation of super-enhancers accompanied by genomic structural abnormalities. Hi-C and long-read sequencing data further corroborated this insight. When we employed CRISPR-Cas9 to induce structural abnormalities that mimicked cases with outlier ERBB2 gene expression, we observed an elevation in ERBB2 expression. These abnormalities are associated with a higher risk of recurrence after surgery, irrespective of the presence or absence of driver mutations. Conclusions Our findings suggest that aberrant gene expression linked to structural polymorphisms can significantly impact personalized cancer treatment by facilitating the identification of driver mutations and prognostic factors, contributing to a more comprehensive understanding of LUAD pathogenesis.
- Published
- 2024
- Full Text
- View/download PDF
5. Mechanism of ERBB2 gene overexpression by the formation of super-enhancer with genomic structural abnormalities in lung adenocarcinoma without clinically actionable genetic alterations.
- Author
-
Kaneko, Syuzo, Takasawa, Ken, Asada, Ken, Shiraishi, Kouya, Ikawa, Noriko, Machino, Hidenori, Shinkai, Norio, Matsuda, Maiko, Masuda, Mari, Adachi, Shungo, Takahashi, Satoshi, Kobayashi, Kazuma, Kouno, Nobuji, Bolatkan, Amina, Komatsu, Masaaki, Yamada, Masayoshi, Miyake, Mototaka, Watanabe, Hirokazu, Tateishi, Akiko, and Mizuno, Takaaki
- Subjects
- *
GENETIC overexpression , *SUPER enhancers , *GENE expression , *GENE enhancers , *GENOMICS , *LUNGS - Abstract
Background: In an extensive genomic analysis of lung adenocarcinomas (LUADs), driver mutations have been recognized as potential targets for molecular therapy. However, there remain cases where target genes are not identified. Super-enhancers and structural variants are frequently identified in several hundred loci per case. Despite this, most cancer research has approached the analysis of these data sets separately, without merging and comparing the data, and there are no examples of integrated analysis in LUAD. Methods: We performed an integrated analysis of super-enhancers and structural variants in a cohort of 174 LUAD cases that lacked clinically actionable genetic alterations. To achieve this, we conducted both WGS and H3K27Ac ChIP-seq analyses using samples with driver gene mutations and those without, allowing for a comprehensive investigation of the potential roles of super-enhancer in LUAD cases. Results: We demonstrate that most genes situated in these overlapped regions were associated with known and previously unknown driver genes and aberrant expression resulting from the formation of super-enhancers accompanied by genomic structural abnormalities. Hi-C and long-read sequencing data further corroborated this insight. When we employed CRISPR-Cas9 to induce structural abnormalities that mimicked cases with outlier ERBB2 gene expression, we observed an elevation in ERBB2 expression. These abnormalities are associated with a higher risk of recurrence after surgery, irrespective of the presence or absence of driver mutations. Conclusions: Our findings suggest that aberrant gene expression linked to structural polymorphisms can significantly impact personalized cancer treatment by facilitating the identification of driver mutations and prognostic factors, contributing to a more comprehensive understanding of LUAD pathogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Effects of super-enhancers in cancer metastasis: mechanisms and therapeutic targets.
- Author
-
Liu, Shenglan, Dai, Wei, Jin, Bei, Jiang, Feng, Huang, Hao, Hou, Wen, Lan, Jinxia, Jin, Yanli, Peng, Weijie, and Pan, Jingxuan
- Subjects
- *
SUPER enhancers , *DRUG target , *METASTASIS , *ONCOLOGY , *EPITHELIAL-mesenchymal transition , *GENETIC transcription - Abstract
Metastasis remains the principal cause of cancer-related lethality despite advancements in cancer treatment. Dysfunctional epigenetic alterations are crucial in the metastatic cascade. Among these, super-enhancers (SEs), emerging as new epigenetic regulators, consist of large clusters of regulatory elements that drive the high-level expression of genes essential for the oncogenic process, upon which cancer cells develop a profound dependency. These SE-driven oncogenes play an important role in regulating various facets of metastasis, including the promotion of tumor proliferation in primary and distal metastatic organs, facilitating cellular migration and invasion into the vasculature, triggering epithelial-mesenchymal transition, enhancing cancer stem cell-like properties, circumventing immune detection, and adapting to the heterogeneity of metastatic niches. This heavy reliance on SE-mediated transcription delineates a vulnerable target for therapeutic intervention in cancer cells. In this article, we review current insights into the characteristics, identification methodologies, formation, and activation mechanisms of SEs. We also elaborate the oncogenic roles and regulatory functions of SEs in the context of cancer metastasis. Ultimately, we discuss the potential of SEs as novel therapeutic targets and their implications in clinical oncology, offering insights into future directions for innovative cancer treatment strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Histone H3.3 K27M chromatin functions implicate a network of neurodevelopmental factors including ASCL1 and NEUROD1 in DIPG
- Author
-
Lewis, Nichole A, Klein, Rachel Herndon, Kelly, Cailin, Yee, Jennifer, and Knoepfler, Paul S
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Rare Diseases ,Pediatric Cancer ,Cancer Genomics ,Genetics ,Neurosciences ,Pediatric ,Cancer ,Human Genome ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Non-Human ,Brain Disorders ,Brain Cancer ,2.1 Biological and endogenous factors ,Generic health relevance ,Humans ,Basic Helix-Loop-Helix Transcription Factors ,Brain Stem Neoplasms ,Chromatin ,Glioma ,Histones ,Mutation ,ATAC-seq ,ASCL1 ,NEUROD1 ,diffuse midline glioma ,Super-enhancers ,Epigenetics ,DIPG ,H3 ,Open chromatin ,Transcription factor binding motifs ,H3.3K27M - Abstract
BackgroundThe histone variant H3.3 K27M mutation is a defining characteristic of diffuse intrinsic pontine glioma (DIPG)/diffuse midline glioma (DMG). This histone mutation is responsible for major alterations to histone H3 post-translational modification (PTMs) and subsequent aberrant gene expression. However, much less is known about the effect this mutation has on chromatin structure and function, including open versus closed chromatin regions as well as their transcriptomic consequences.ResultsRecently, we developed isogenic CRISPR-edited DIPG cell lines that are wild-type for histone H3.3 that can be compared to their matched K27M lines. Here we show via ATAC-seq analysis that H3.3K27M glioma cells have unique accessible chromatin at regions corresponding to neurogenesis, NOTCH, and neuronal development pathways and associated genes that are overexpressed in H3.3K27M compared to our isogenic wild-type cell line. As to mechanisms, accessible enhancers and super-enhancers corresponding to increased gene expression in H3.3K27M cells were also mapped to genes involved in neurogenesis and NOTCH signaling, suggesting that these pathways are key to DIPG tumor maintenance. Motif analysis implicates specific transcription factors as central to the neuro-oncogenic K27M signaling pathway, in particular, ASCL1 and NEUROD1.ConclusionsAltogether our findings indicate that H3.3K27M causes chromatin to take on a more accessible configuration at key regulatory regions for NOTCH and neurogenesis genes resulting in increased oncogenic gene expression, which is at least partially reversible upon editing K27M back to wild-type.
- Published
- 2022
8. NSMCE2, a novel super-enhancer-regulated gene, is linked to poor prognosis and therapy resistance in breast cancer.
- Author
-
Di Benedetto, Carolina, Oh, Justin, Choudhery, Zainab, Shi, Weiquan, Valdes, Gilmer, and Betancur, Paola
- Subjects
Chromatin ,Humans ,Breast Neoplasms ,Ligases ,RNA ,Prognosis ,Gene Expression Regulation ,Neoplastic ,Female ,Enhancer Elements ,Genetic ,Myelin and Lymphocyte-Associated Proteolipid Proteins ,Breast cancer ,MAL2 ,NSMCE2 ,Super-enhancers ,Therapy resistance ,Breast Cancer ,Cancer ,Biotechnology ,Human Genome ,Genetics ,5.1 Pharmaceuticals ,2.1 Biological and endogenous factors ,Development of treatments and therapeutic interventions ,Aetiology ,Oncology and Carcinogenesis ,Public Health and Health Services ,Oncology & Carcinogenesis - Abstract
BackgroundDespite today's advances in the treatment of cancer, breast cancer-related mortality remains high, in part due to the lack of effective targeted therapies against breast tumor types that do not respond to standard treatments. Therefore, identifying additional breast cancer molecular targets is urgently needed. Super-enhancers are large regions of open chromatin involved in the overactivation of oncogenes. Thus, inhibition of super-enhancers has become a focus in clinical trials for its therapeutic potential. Here, we aimed to identify novel super-enhancer dysregulated genes highly associated with breast cancer patients' poor prognosis and negative response to treatment.MethodsUsing existing datasets containing super-enhancer-associated genes identified in breast tumors and public databases comprising genomic and clinical information for breast cancer patients, we investigated whether highly expressed super-enhancer-associated genes correlate to breast cancer patients' poor prognosis and to patients' poor response to therapy. Our computational findings were experimentally confirmed in breast cancer cells by pharmacological SE disruption and gene silencing techniques.ResultsWe bioinformatically identified two novel super-enhancer-associated genes - NSMCE2 and MAL2 - highly upregulated in breast tumors, for which high RNA levels significantly and specifically correlate with breast cancer patients' poor prognosis. Through in-vitro pharmacological super-enhancer disruption assays, we confirmed that super-enhancers upregulate NSMCE2 and MAL2 transcriptionally, and, through bioinformatics, we found that high levels of NSMCE2 strongly associate with patients' poor response to chemotherapy, especially for patients diagnosed with aggressive triple negative and HER2 positive tumor types. Finally, we showed that decreasing NSMCE2 gene expression increases breast cancer cells' sensitivity to chemotherapy treatment.ConclusionsOur results indicate that moderating the transcript levels of NSMCE2 could improve patients' response to standard chemotherapy consequently improving disease outcome. Our approach offers a new avenue to identify a signature of tumor specific genes that are not frequently mutated but dysregulated by super-enhancers. As a result, this strategy can lead to the discovery of potential and novel pharmacological targets for improving targeted therapy and the treatment of breast cancer.
- Published
- 2022
9. Pathogenic role of super-enhancers as potential therapeutic targets in lung cancer.
- Author
-
Zhiyuan Yao, Peng Song, and Wenjie Jiao
- Subjects
SUPER enhancers ,LUNG cancer ,DRUG target ,CANCER patients - Abstract
Lung cancer is still one of the deadliest malignancies today, and most patients with advanced lung cancer pass away from disease progression that is uncontrollable by medications. Super-enhancers (SEs) are large clusters of enhancers in the genome's non-coding sequences that actively trigger transcription. Although SEs have just been identified over the past 10 years, their intricate structure and crucial role in determining cell identity and promoting tumorigenesis and progression are increasingly coming to light. Here, we review the structural composition of SEs, the auto-regulatory circuits, the control mechanisms of downstream genes and pathways, and the characterization of subgroups classified according to SEs in lung cancer. Additionally, we discuss the therapeutic targets, several small-molecule inhibitors, and available treatment options for SEs in lung cancer. Combination therapies have demonstrated considerable advantages in preclinical models, and we anticipate that these drugs will soon enter clinical studies and benefit patients. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Super-Enhancers and Their Parts: From Prediction Efforts to Pathognomonic Status.
- Author
-
Vasileva, Anastasia V., Gladkova, Marina G., Ashniev, German A., Osintseva, Ekaterina D., Orlov, Alexey V., Kravchuk, Ekaterina V., Boldyreva, Anna V., Burenin, Alexander G., Nikitin, Petr I., and Orlova, Natalia N.
- Subjects
- *
SUPER enhancers , *GENE expression , *GENETIC regulation , *REGULATOR genes , *HUMAN genome - Abstract
Super-enhancers (SEs) are regions of the genome that play a crucial regulatory role in gene expression by promoting large-scale transcriptional responses in various cell types and tissues. Recent research suggests that alterations in super-enhancer activity can contribute to the development and progression of various disorders. The aim of this research is to explore the multifaceted roles of super-enhancers in gene regulation and their significant implications for understanding and treating complex diseases. Here, we study and summarise the classification of super-enhancer constituents, their possible modes of interaction, and cross-regulation, including super-enhancer RNAs (seRNAs). We try to investigate the opportunity of SE dynamics prediction based on the hierarchy of enhancer single elements (enhancers) and their aggregated action. To further our understanding, we conducted an in silico experiment to compare and differentiate between super-enhancers and locus-control regions (LCRs), shedding light on the enigmatic relationship between LCRs and SEs within the human genome. Particular attention is paid to the classification of specific mechanisms and their diversity, exemplified by various oncological, cardiovascular, and immunological diseases, as well as an overview of several anti-SE therapies. Overall, the work presents a comprehensive analysis of super-enhancers across different diseases, aiming to provide insights into their regulatory roles and may act as a rationale for future clinical interventions targeting these regulatory elements. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Transcription factor condensates and signaling driven transcription.
- Author
-
Mann, Rajat and Notani, Dimple
- Subjects
- *
TRANSCRIPTION factors , *TRANSGENIC organisms , *PROTEINS , *RNA , *DNA - Abstract
Transcription Factor (TF) condensates are a heterogenous mix of RNA, DNA, and multiple co-factor proteins capable of modulating the transcriptional response of the cell. The dynamic nature and the spatial location of TF-condensates in the 3D nuclear space is believed to provide a fast response, which is on the same pace as the signaling cascade and yet ever-so-specific in the crowded environment of the nucleus. However, the current understanding of how TF-condensates can achieve these feet so quickly and efficiently is still unclear. In this review, we draw parallels with other protein condensates and share our speculations on how the nucleus uses these TF-condensates to achieve high transcriptional specificity and fidelity. We discuss the various constituents of TF-condensates, their properties, and the known and unknown functions of TF-condensates with a particular focus on steroid signaling-induced transcriptional programs. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
12. Etiology of super-enhancer reprogramming and activation in cancer
- Author
-
Royce W. Zhou and Ramon E. Parsons
- Subjects
Enhancers ,Super-enhancers ,Cancer ,Extrachromosomal DNA ,Topologically associated domain ,Non-coding mutations ,Genetics ,QH426-470 - Abstract
Abstract Super-enhancers are large, densely concentrated swaths of enhancers that regulate genes critical for cell identity. Tumorigenesis is accompanied by changes in the super-enhancer landscape. These aberrant super-enhancers commonly form to activate proto-oncogenes, or other genes upon which cancer cells depend, that initiate tumorigenesis, promote tumor proliferation, and increase the fitness of cancer cells to survive in the tumor microenvironment. These include well-recognized master regulators of proliferation in the setting of cancer, such as the transcription factor MYC which is under the control of numerous super-enhancers gained in cancer compared to normal tissues. This Review will cover the expanding cell-intrinsic and cell-extrinsic etiology of these super-enhancer changes in cancer, including somatic mutations, copy number variation, fusion events, extrachromosomal DNA, and 3D chromatin architecture, as well as those activated by inflammation, extra-cellular signaling, and the tumor microenvironment.
- Published
- 2023
- Full Text
- View/download PDF
13. Role and Mechanism of Super-enhancers in Tumor Metastasis
- Author
-
GUO Caiyao, WANG Yu, DAI Wei, and LIU Shenglan
- Subjects
super-enhancers ,tumor metastasis ,long non-coding rna ,epithelial-mesenchymal transition ,cancer stem cells ,tumor microenvironment ,inhibitor ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Super-enhancers (SEs) are large clusters of enhancers located near the promoter and are necessary to determine the identity of cancer cells. The alterations of super-enhancers can cause dysregulation of the transcriptional program, which resulted in tumor cells being addicted to certain transcriptional programs. Tumor metastasis is the leading cause of death in cancer. Recently, SEs have been demonstrated to facilitate tumor metastasis by regulating lncRNA generation, tumor microenvironment, epithelial-mesenchymal transition, and cancer stem cells. In this review, the characteristics of SEs, the relationship between SEs and tumor metastasis, and inhibitors against SEs are summarized to provide a reference for the relevant mechanism of SEs regulating tumor metastasis and provide new perspectives for the diagnosis and treatment of patients with cancer metastasis.
- Published
- 2023
- Full Text
- View/download PDF
14. Super-enhancer driven SOX2 promotes tumor formation by chromatin re-organization in nasopharyngeal carcinomaResearch in context
- Author
-
Shang-Xin Liu, Chong Wang, Ruo-Bin Lin, Wei-Yue Ding, Gaurab Roy, Hong-Bo Wang, Ting Yang, Qian Liu, Yi-Ling Luo, Shui-Lin Jin, Mu-Sheng Zeng, Bo Zhao, and Qian Zhong
- Subjects
Nasopharyngeal carcinoma ,Enhancer connectomes ,Super-enhancers ,Chromatin re-organization ,Genetic variation ,Medicine ,Medicine (General) ,R5-920 - Abstract
Summary: Background: Nasopharyngeal carcinoma (NPC) is a malignant head and neck cancer with a high incidence in Southern China and Southeast Asia. Patients with remote metastasis and recurrent NPC have poor prognosis. Thus, a better understanding of NPC pathogenesis may identify novel therapies to address the unmet clinical needs. Methods: H3K27ac ChIP-seq and HiChIP was applied to understand the enhancer landscapes and the chromosome interactions. Whole genome sequencing was conducted to analyze the relationship between genomic variations and epigenetic dysregulation. CRISPRi and JQ1 treatment were used to evaluate the transcriptional regulation of SOX2 SEs. Colony formation assay, survival analysis and in vivo subcutaneous patient-derived xenograft assays were applied to explore the function and clinical relevance of SOX2 in NPC. Findings: We globally mapped the enhancer landscapes and generated NPC enhancer connectomes, linking NPC specific enhancers and SEs. We found five overlapped genes, including SOX2, among super-enhancer regulated genes, survival related genes and NPC essential genes. The mRNA expression of SOX2 was repressed when applying CRISPRi targeting different SOX2 SEs or JQ1 treatment. Next, we identified a genetic variation (Chr3:181422197, G > A) in SOX2 SE which is correlated with higher expression of SOX2 and poor survival. In addition, SOX2 was highly expressed in NPC and is correlated with short survival in patients with NPC. Knock-down of SOX2 suppressed tumor growth in vitro and in vivo. Interpretation: Our study demonstrated the super-enhancer landscape with chromosome interactions and identified super-enhancer driven SOX2 promotes tumorigenesis, suggesting that SOX2 is a potential therapeutic target for patients with NPC. Funding: A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.
- Published
- 2023
- Full Text
- View/download PDF
15. Transcription factor condensates and signaling driven transcription
- Author
-
Rajat Mann and Dimple Notani
- Subjects
Transcription ,chromatin ,histones ,super-enhancers ,condensates ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
ABSTRACTTranscription Factor (TF) condensates are a heterogenous mix of RNA, DNA, and multiple co-factor proteins capable of modulating the transcriptional response of the cell. The dynamic nature and the spatial location of TF-condensates in the 3D nuclear space is believed to provide a fast response, which is on the same pace as the signaling cascade and yet ever-so-specific in the crowded environment of the nucleus. However, the current understanding of how TF-condensates can achieve these feet so quickly and efficiently is still unclear. In this review, we draw parallels with other protein condensates and share our speculations on how the nucleus uses these TF-condensates to achieve high transcriptional specificity and fidelity. We discuss the various constituents of TF-condensates, their properties, and the known and unknown functions of TF-condensates with a particular focus on steroid signaling-induced transcriptional programs.
- Published
- 2023
- Full Text
- View/download PDF
16. Residual Neural Network in Genomics
- Author
-
Sara Sabba, Meroua Smara, Mehdi Benhacine, Loubna Terra, and Zine Eddine Terra
- Subjects
deep learning ,genomics ,convolutional neural network ,companion ,residual neural network ,super-enhancers ,viral genomes ,Electronic computers. Computer science ,QA75.5-76.95 - Abstract
Residual neural network (ResNet) is a Deep Learning model introduced by He et al. in 2015 to enhance traditional convolutional neural networks proposed to solve computer vision problems. It uses skip connections over some layer blocks to avoid vanishing gradient problem. Currently, many researches are focused to test and prove the efficiency of the ResNet on different domains such as genomics. In fact, the study of human genomes provides important information on the detection of diseases and their best treatments. Therefore, most of the scientists opted for bioinformatics solutions to get results in a reasonable time. In this paper, our interest is to show the effectiveness of the ResNet model on genomics. For that, we propose two new ResNet models to enhance the results of two genomic problems previously resolved by CNN models. The obtained results are very promising and they proved the performance of our ResNet models compared to the CNN models.
- Published
- 2022
- Full Text
- View/download PDF
17. A body map of super-enhancers and their function in pig
- Author
-
Youbing Yang, Xinyue Li, Zhu Meng, Yongjian Liu, Kaifeng Qian, Mingxing Chu, and Zhangyuan Pan
- Subjects
super-enhancers ,pig ,features ,tissue-specific ,function ,Veterinary medicine ,SF600-1100 - Abstract
IntroductionSuper-enhancers (SEs) are clusters of enhancers that act synergistically to drive the high-level expression of genes involved in cell identity and function. Although SEs have been extensively investigated in humans and mice, they have not been well characterized in pigs.MethodsHere, we identified 42,380 SEs in 14 pig tissues using chromatin immunoprecipitation sequencing, and statistics of its overall situation, studied the composition and characteristics of SE, and explored the influence of SEs characteristics on gene expression.ResultsWe observed that approximately 40% of normal enhancers (NEs) form SEs. Compared to NEs, we found that SEs were more likely to be enriched with an activated enhancer and show activated functions. Interestingly, SEs showed X chromosome depletion and short interspersed nuclear element enrichment, implying that SEs play an important role in sex traits and repeat evolution. Additionally, SE-associated genes exhibited higher expression levels and stronger conservation than NE-associated genes. However, genes with the largest SEs had higher expression levels than those with the smallest SEs, indicating that SE size may influence gene expression. Moreover, we observed a negative correlation between SE gene distance and gene expression, indicating that the proximity of SEs can affect gene activity. Gene ontology enrichment and motif analysis revealed that SEs have strong tissue-specific activity. For example, the CORO2B gene with a brain-specific SE shows strong brain-specific expression, and the phenylalanine hydroxylase gene with liver-specific SEs shows strong liver-specific expression.DiscussionIn this study, we illustrated a body map of SEs and explored their functions in pigs, providing information on the composition and tissue-specific patterns of SEs. This study can serve as a valuable resource of gene regulatory and comparative analyses to the scientific community and provides a theoretical reference for genetic control mechanisms of important traits in pigs.
- Published
- 2023
- Full Text
- View/download PDF
18. Etiology of super-enhancer reprogramming and activation in cancer.
- Author
-
Zhou, Royce W. and Parsons, Ramon E.
- Subjects
- *
EXTRACHROMOSOMAL DNA , *SOMATIC mutation , *GENE enhancers , *ETIOLOGY of diseases , *TUMOR microenvironment , *PROTO-oncogenes - Abstract
Super-enhancers are large, densely concentrated swaths of enhancers that regulate genes critical for cell identity. Tumorigenesis is accompanied by changes in the super-enhancer landscape. These aberrant super-enhancers commonly form to activate proto-oncogenes, or other genes upon which cancer cells depend, that initiate tumorigenesis, promote tumor proliferation, and increase the fitness of cancer cells to survive in the tumor microenvironment. These include well-recognized master regulators of proliferation in the setting of cancer, such as the transcription factor MYC which is under the control of numerous super-enhancers gained in cancer compared to normal tissues. This Review will cover the expanding cell-intrinsic and cell-extrinsic etiology of these super-enhancer changes in cancer, including somatic mutations, copy number variation, fusion events, extrachromosomal DNA, and 3D chromatin architecture, as well as those activated by inflammation, extra-cellular signaling, and the tumor microenvironment. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
19. Oncogenic super-enhancers in cancer: mechanisms and therapeutic targets.
- Author
-
Bacabac, Megan and Xu, Wei
- Abstract
Activation of oncogenes to sustain proliferative signaling and initiate metastasis are important hallmarks of cancer. Oncogenes are amplified or overexpressed in cancer cells and overexpression is often controlled at the level of transcription. Gene expression is tightly controlled by many cis-regulatory elements and trans-acting factors. Large clusters of enhancers known as "super-enhancers" drive robust expression of cell-fate determining transcription factors in cell identity. Cancer cells can take advantage of super-enhancers and become transcriptionally addicted to them leading to tumorigenesis and metastasis. Additionally, the cis-regulatory landscape of cancer includes aberrant super-enhancers that are not present in normal cells. The landscape of super-enhancers in cancer is characterized by high levels of histone H3K27 acetylation and bromodomain-containing protein 4 (BRD4), and Mediator complex. These chromatin features facilitate the identification of cancer type-specific and cell-type-specific super-enhancers that control the expression of important oncogenes to stimulate their growth. Disruption of super-enhancers via inhibiting BRD4 or other epigenetic proteins is a potential therapeutic option. Here, we will describe the discovery of super-enhancers and their unique characteristics compared to typical enhancers. Then, we will highlight how super-enhancer-associated genes contribute to cancer progression in different solid tumor types. Lastly, we will cover therapeutic targets and their epigenetic modulators. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
20. 超级增强子在肿瘤转移中的作用机制研究进展.
- Author
-
郭彩瑶, 王宇, 戴伟, and 刘圣兰
- Abstract
Super-enhancers (SEs) are large clusters of enhancers located near the promoter and are necessary to determine the identity of cancer cells. The alterations of super-enhancers can cause dysregulation of the transcriptional program, which resulted in tumor cells being addicted to certain transcriptional programs. Tumor metastasis is the leading cause of death in cancer. Recently, SEs have been demonstrated to facilitate tumor metastasis by regulating lncRNA generation, tumor microenvironment, epithelial-mesenchymal transition, and cancer stem cells. In this review, the characteristics of SEs, the relationship between SEs and tumor metastasis, and inhibitors against SEs are summarized to provide a reference for the relevant mechanism of SEs regulating tumor metastasis and provide new perspectives for the diagnosis and treatment of patients with cancer metastasis. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
21. Residual Neural Network for Predicting Super-Enhancers on Genome Scale
- Author
-
Sabba, Sara, Smara, Meroua, Benhacine, Mehdi, Hameurlaine, Amina, Kacprzyk, Janusz, Series Editor, Gomide, Fernando, Advisory Editor, Kaynak, Okyay, Advisory Editor, Liu, Derong, Advisory Editor, Pedrycz, Witold, Advisory Editor, Polycarpou, Marios M., Advisory Editor, Rudas, Imre J., Advisory Editor, Wang, Jun, Advisory Editor, Lejdel, Brahim, editor, Clementini, Eliseo, editor, and Alarabi, Louai, editor
- Published
- 2022
- Full Text
- View/download PDF
22. Endodermal Maternal Transcription Factors Establish Super-Enhancers during Zygotic Genome Activation
- Author
-
Paraiso, Kitt D, Blitz, Ira L, Coley, Masani, Cheung, Jessica, Sudou, Norihiro, Taira, Masanori, and Cho, Ken WY
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Human Genome ,Stem Cell Research ,Genetics ,1.1 Normal biological development and functioning ,Animals ,Binding Sites ,Chromatin ,Endoderm ,Enhancer Elements ,Genetic ,Female ,Forkhead Transcription Factors ,Genome ,Histones ,Male ,Morpholinos ,Otx Transcription Factors ,RNA Polymerase II ,T-Box Domain Proteins ,Transcriptome ,Xenopus ,Xenopus Proteins ,Zygote ,Foxh1 ,Otx1 ,Vegt ,Xenopus tropicalis ,endoderm ,enhancer RNAs ,germ layer specification ,histone marks ,super-enhancers ,zygotic genome activation ,Biochemistry and Cell Biology ,Medical Physiology ,Biological sciences - Abstract
Elucidation of the sequence of events underlying the dynamic interaction between transcription factors and chromatin states is essential. Maternal transcription factors function at the top of the regulatory hierarchy to specify the primary germ layers at the onset of zygotic genome activation (ZGA). We focus on the formation of endoderm progenitor cells and examine the interactions between maternal transcription factors and chromatin state changes underlying the cell specification process. Endoderm-specific factors Otx1 and Vegt together with Foxh1 orchestrate endoderm formation by coordinated binding to select regulatory regions. These interactions occur before the deposition of enhancer histone marks around the regulatory regions, and these TFs recruit RNA polymerase II, regulate enhancer activity, and establish super-enhancers associated with important endodermal genes. Therefore, maternal transcription factors Otx1, Vegt, and Foxh1 combinatorially regulate the activity of super-enhancers, which in turn activate key lineage-specifying genes during ZGA.
- Published
- 2019
23. NSMCE2, a novel super-enhancer-regulated gene, is linked to poor prognosis and therapy resistance in breast cancer
- Author
-
Carolina Di Benedetto, Justin Oh, Zainab Choudhery, Weiquan Shi, Gilmer Valdes, and Paola Betancur
- Subjects
Breast cancer ,Super-enhancers ,NSMCE2 ,MAL2 ,Therapy resistance ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Despite today’s advances in the treatment of cancer, breast cancer-related mortality remains high, in part due to the lack of effective targeted therapies against breast tumor types that do not respond to standard treatments. Therefore, identifying additional breast cancer molecular targets is urgently needed. Super-enhancers are large regions of open chromatin involved in the overactivation of oncogenes. Thus, inhibition of super-enhancers has become a focus in clinical trials for its therapeutic potential. Here, we aimed to identify novel super-enhancer dysregulated genes highly associated with breast cancer patients’ poor prognosis and negative response to treatment. Methods Using existing datasets containing super-enhancer-associated genes identified in breast tumors and public databases comprising genomic and clinical information for breast cancer patients, we investigated whether highly expressed super-enhancer-associated genes correlate to breast cancer patients’ poor prognosis and to patients’ poor response to therapy. Our computational findings were experimentally confirmed in breast cancer cells by pharmacological SE disruption and gene silencing techniques. Results We bioinformatically identified two novel super-enhancer-associated genes – NSMCE2 and MAL2 – highly upregulated in breast tumors, for which high RNA levels significantly and specifically correlate with breast cancer patients’ poor prognosis. Through in-vitro pharmacological super-enhancer disruption assays, we confirmed that super-enhancers upregulate NSMCE2 and MAL2 transcriptionally, and, through bioinformatics, we found that high levels of NSMCE2 strongly associate with patients’ poor response to chemotherapy, especially for patients diagnosed with aggressive triple negative and HER2 positive tumor types. Finally, we showed that decreasing NSMCE2 gene expression increases breast cancer cells’ sensitivity to chemotherapy treatment. Conclusions Our results indicate that moderating the transcript levels of NSMCE2 could improve patients’ response to standard chemotherapy consequently improving disease outcome. Our approach offers a new avenue to identify a signature of tumor specific genes that are not frequently mutated but dysregulated by super-enhancers. As a result, this strategy can lead to the discovery of potential and novel pharmacological targets for improving targeted therapy and the treatment of breast cancer.
- Published
- 2022
- Full Text
- View/download PDF
24. Somatic Super-Enhancer Epigenetic Signature for Overall Survival Prediction in Patients with Breast Invasive Carcinoma.
- Author
-
Yang, Xu, Zheng, Wenzhong, Li, Mengqiang, and Zhang, Shiqiang
- Subjects
- *
BREAST , *OVERALL survival , *EPIGENETICS , *FEATURE selection , *BREAST implants , *INDIVIDUALIZED medicine , *GENE expression - Abstract
To analyze genome-wide super-enhancers (SEs) methylation signature of breast invasive carcinoma (BRCA) and its clinical value. Differential methylation sites (DMS) between BRCA and adjacent tissues from The Cancer Genome Atlas (TCGA) database were identified by using ChAMP package in R software. Super-enhancers were identified sing ROSE software. Overlap analysis was used to assess the potential DMS in SEs region. Feature selection was performed by Cox regression and least absolute shrinkage and selection operator (LASSO) algorithm based on TCGA training cohort. Prognosis model validation was performed in TCGA training cohort, TCGA validation cohort, and gene expression omnibus (GEO) test cohort. The gene ontology and KEGG analysis revealed that SEs target genes were significantly enriched in cell-migration-associated processes and pathways. A total of 83 654 DMS were identified between BRCA and adjacent tissues. Around 2397 DMS in SEs region were identified by overlap study and used to feature selection. By using Cox regression and LASSO algorithm, 42 features were selected to develop a clinical prediction model (CPM). Both training (TCGA) and validation cohorts (TCGA and GEO) show that the CPM has ideal discrimination and calibration. The CPM based on DMS at SE regions has ideal discrimination and calibration, which combined with tumor node metastasis (TNM) stage could improve prognostication, and thus contribute to individualized medicine. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
25. Histone H3.3 K27M chromatin functions implicate a network of neurodevelopmental factors including ASCL1 and NEUROD1 in DIPG
- Author
-
Nichole A. Lewis, Rachel Herndon Klein, Cailin Kelly, Jennifer Yee, and Paul S. Knoepfler
- Subjects
ATAC-seq ,ASCL1 ,NEUROD1 ,diffuse midline glioma ,Super-enhancers ,Epigenetics ,Genetics ,QH426-470 - Abstract
Abstract Background The histone variant H3.3 K27M mutation is a defining characteristic of diffuse intrinsic pontine glioma (DIPG)/diffuse midline glioma (DMG). This histone mutation is responsible for major alterations to histone H3 post-translational modification (PTMs) and subsequent aberrant gene expression. However, much less is known about the effect this mutation has on chromatin structure and function, including open versus closed chromatin regions as well as their transcriptomic consequences. Results Recently, we developed isogenic CRISPR-edited DIPG cell lines that are wild-type for histone H3.3 that can be compared to their matched K27M lines. Here we show via ATAC-seq analysis that H3.3K27M glioma cells have unique accessible chromatin at regions corresponding to neurogenesis, NOTCH, and neuronal development pathways and associated genes that are overexpressed in H3.3K27M compared to our isogenic wild-type cell line. As to mechanisms, accessible enhancers and super-enhancers corresponding to increased gene expression in H3.3K27M cells were also mapped to genes involved in neurogenesis and NOTCH signaling, suggesting that these pathways are key to DIPG tumor maintenance. Motif analysis implicates specific transcription factors as central to the neuro-oncogenic K27M signaling pathway, in particular, ASCL1 and NEUROD1. Conclusions Altogether our findings indicate that H3.3K27M causes chromatin to take on a more accessible configuration at key regulatory regions for NOTCH and neurogenesis genes resulting in increased oncogenic gene expression, which is at least partially reversible upon editing K27M back to wild-type.
- Published
- 2022
- Full Text
- View/download PDF
26. Complexity of enhancer networks predicts cell identity and disease genes revealed by single-cell multi-omics analysis.
- Author
-
Hong, Danni, Lin, Hongli, Liu, Lifang, Shu, Muya, Dai, Jianwu, Lu, Falong, Tong, Mengsha, and Huang, Jialiang
- Subjects
- *
GENE regulatory networks , *MULTIOMICS , *GENE expression profiling , *GENES , *NETWORK hubs , *GENE expression - Abstract
Many enhancers exist as clusters in the genome and control cell identity and disease genes; however, the underlying mechanism remains largely unknown. Here, we introduce an algorithm, eNet, to build enhancer networks by integrating single-cell chromatin accessibility and gene expression profiles. The complexity of enhancer networks is assessed by two metrics: the number of enhancers and the frequency of predicted enhancer interactions (PEIs) based on chromatin co-accessibility. We apply eNet algorithm to a human blood dataset and find cell identity and disease genes tend to be regulated by complex enhancer networks. The network hub enhancers (enhancers with frequent PEIs) are the most functionally important. Compared with super-enhancers, enhancer networks show better performance in predicting cell identity and disease genes. eNet is robust and widely applicable in various human or mouse tissues datasets. Thus, we propose a model of enhancer networks containing three modes: Simple, Multiple and Complex, which are distinguished by their complexity in regulating gene expression. Taken together, our work provides an unsupervised approach to simultaneously identify key cell identity and disease genes and explore the underlying regulatory relationships among enhancers in single cells. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
27. Somatic Super-Enhancer Epigenetic Signature for Overall Survival Prediction in Patients with Breast Invasive Carcinoma.
- Author
-
Xu Yang, Wenzhong Zheng, Mengqiang Li, and Shiqiang Zhang
- Subjects
- *
BREAST , *OVERALL survival , *EPIGENETICS , *FEATURE selection , *BREAST implants , *INDIVIDUALIZED medicine , *GENE expression - Abstract
To analyze genome-wide super-enhancers (SEs) methylation signature of breast invasive carcinoma (BRCA) and its clinical value. Differential methylation sites (DMS) between BRCA and adjacent tissues from The Cancer Genome Atlas (TCGA) database were identified by using ChAMP package in R software. Super-enhancers were identified sing ROSE software. Overlap analysis was used to assess the potential DMS in SEs region. Feature selection was performed by Cox regression and least absolute shrinkage and selection operator (LASSO) algorithm based on TCGA training cohort. Prognosis model validation was performed in TCGA training cohort, TCGA validation cohort, and gene expression omnibus (GEO) test cohort. The gene ontology and KEGG analysis revealed that SEs target genes were significantly enriched in cell-migration-associated processes and pathways. A total of 83 654 DMS were identified between BRCA and adjacent tissues. Around 2397 DMS in SEs region were identified by overlap study and used to feature selection. By using Cox regression and LASSO algorithm, 42 features were selected to develop a clinical prediction model (CPM). Both training (TCGA) and validation cohorts (TCGA and GEO) show that the CPM has ideal discrimination and calibration. The CPM based on DMS at SE regions has ideal discrimination and calibration, which combined with tumor node metastasis (TNM) stage could improve prognostication, and thus contribute to individualized medicine. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
28. Expression of risk genes linked to vitamin D receptor super-enhancer regions and their association with phenotype severity in multiple sclerosis.
- Author
-
Orton, Sarah M., Sangha, Amarpreet, Gupta, Mehul, Martens, Kristina, Metz, Luanne M., de Koning, A. P. J., and Pfeffer, Gerald
- Subjects
VITAMIN D receptors ,GENE enhancers ,MULTIPLE sclerosis ,VITAMIN D deficiency ,GENE expression ,PHENOTYPIC plasticity - Abstract
Multiple sclerosis (MS) is a chronic debilitating neurological condition with a wide range of phenotype variability. A complex interplay of genetic and environmental factors contributes to disease onset and progression in MS patients. Vitamin D deficiency is a known susceptibility factor for MS, however the underlyingmechanismof vitamin D-gene interactions in MS etiology is still poorly understood. Vitamin D receptor super-enhancers (VSEs) are enriched in MS risk variants and may modulate these environment-gene interactions. mRNA expression in total of 64 patients with contrasting MS severity was quantified in select genes. First, RNA-seq was performed on a discovery cohort (10 mild, 10 severe MS phenotype) and ten genes regulated by VSEs that have been linked to MS risk were analyzed. Four candidates showed a significant positive association (GRINA, PLEC, PARP10, and LRG1) in the discovery cohort and were then quantified using digital droplet PCR (ddPCR) in a validation cohort (33 mild, 11 severe MS phenotype). A significant differential expression persisted in the validation cohort for three of the VSE-MS genes: GRINA (p = 0.0138), LRG1 (p = 0.0157), and PLEC (p = 0.0391). In summary, genes regulated by VSE regions that contain known MS risk variants were shown to have differential expression based on disease severity (p<0.05). The findings implicate a role for vitamin D super-enhancers in modulating disease activity. In addition, expression levels may have some utility as prognostic biomarkers in the future. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
29. Interplay Between the Histone Variant H2A.Z and the Epigenome in Pancreatic Cancer.
- Author
-
Ávila-López, Pedro A., Nuñez-Martínez, Hober N., Peralta-Alvarez, Carlos A., Martinez-Calvillo, Santiago, Recillas-Targa, Félix, and Hernández-Rivas, Rosaura
- Subjects
- *
PANCREATIC cancer , *RNA polymerase II , *DEVELOPMENTAL biology , *POST-translational modification , *TRANSCRIPTION factors - Abstract
The oncogenic process is orchestrated by a complex network of chromatin remodeling elements that shape the cancer epigenome. Histone variant H2A.Z regulates DNA control elements such as promoters and enhancers in different types of cancer; however, the interplay between H2A.Z and the pancreatic cancer epigenome is unknown. This study analyzed the role of H2A.Z in different DNA regulatory elements. We performed Chromatin Immunoprecipitation Sequencing assays (ChiP-seq) with total H2A.Z and acetylated H2A.Z (acH2A.Z) antibodies and analyzed published data from ChIP-seq, RNA-seq, bromouridine labeling-UV and sequencing (BruUV-seq), Hi-C and ATAC-seq (Assay for Transposase-Accessible Chromatin using sequencing) in the pancreatic cancer cell line PANC-1. The results indicate that total H2A.Z facilitates the recruitment of RNA polymerase II and transcription factors at promoters and enhancers allowing the expression of pro-oncogenic genes. Interestingly, we demonstrated that H2A.Z is enriched in super-enhancers (SEs) contributing to the transcriptional activation of key genes implicated in tumor development. Importantly, we established that H2A.Z contributes to the three-dimensional (3D) genome organization of pancreatic cancer and that it is a component of the Topological Associated Domains (TADs) boundaries in PANC-1 and that total H2A.Z and acH2A.Z are associated with A and B compartments, respectively. H2A.Z participates in the biology and development of pancreatic cancer by generating a pro-oncogenic transcriptome through its posttranslational modifications, interactions with different partners, and regulatory elements, contributing to the oncogenic 3D genome organization. These data allow us to understand the molecular mechanisms that promote an oncogenic transcriptome in pancreatic cancer mediated by H2A.Z. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
30. MCIBox: a toolkit for single-molecule multi-way chromatin interaction visualization and micro-domains identification.
- Author
-
Tian, Simon Zhongyuan, Li, Guoliang, Ning, Duo, Jing, Kai, Xu, Yewen, Yang, Yang, Fullwood, Melissa J, Yin, Pengfei, Huang, Guangyu, Plewczynski, Dariusz, Zhai, Jixian, Dai, Ziwei, Chen, Wei, and Zheng, Meizhen
- Subjects
- *
DIMENSIONAL reduction algorithms , *PROBABILITY density function , *GENE mapping , *FUZZY algorithms , *GENETIC regulation , *VISUALIZATION - Abstract
The emerging ligation-free three-dimensional (3D) genome mapping technologies can identify multiplex chromatin interactions with single-molecule precision. These technologies not only offer new insight into high-dimensional chromatin organization and gene regulation, but also introduce new challenges in data visualization and analysis. To overcome these challenges, we developed MCIBox, a toolkit for multi-way chromatin interaction (MCI) analysis, including a visualization tool and a platform for identifying micro-domains with clustered single-molecule chromatin complexes. MCIBox is based on various clustering algorithms integrated with dimensionality reduction methods that can display multiplex chromatin interactions at single-molecule level, allowing users to explore chromatin extrusion patterns and super-enhancers regulation modes in transcription, and to identify single-molecule chromatin complexes that are clustered into micro-domains. Furthermore, MCIBox incorporates a two-dimensional kernel density estimation algorithm to identify micro-domains boundaries automatically. These micro-domains were stratified with distinctive signatures of transcription activity and contained different cell-cycle-associated genes. Taken together, MCIBox represents an invaluable tool for the study of multiple chromatin interactions and inaugurates a previously unappreciated view of 3D genome structure. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
31. Effective Quality Breeding Directions—Comparison and Conservative Analysis of Hepatic Super-Enhancers between Chinese and Western Pig Breeds.
- Author
-
Zhang, Yi, Zhang, Jinbi, Wang, Caixia, Qin, Xinxin, Zhang, Yuge, Liu, Jingge, and Pan, Zengxiang
- Subjects
- *
SWINE , *MICE , *BINDING sites , *SUPER enhancers - Abstract
Simple Summary: In this study, we identify the promoters, typical enhancers (TEs), and super-enhancers (SEs) in the livers of Chinese local and Western commercial pig breeds. Western breeds included fewer SEs in number, while more QTLs of growth-related economic traits were associated with these SEs. Comparison among different porcine tissues and liver tissues from pigs, humans, and mice suggested a high tissue specificity of SE. We concluded that intense selection could concentrate functional SEs; thus, SEs could be applied as effective detection regions in genomic selection breeding. The transcriptional initiation of genes is closely bound to the functions of cis-regulatory elements, including promoters, typical enhancers (TEs), and recently-identified super-enhancers (SEs). In this study, we identified these cis-regulatory elements in the livers of two Chinese (Meishan and Enshi Black) and two Western (Duroc and Large White) pig breeds using ChIP-seq data, then explored their similarities and differences. In addition, we analyzed the conservation of SEs among different tissues and species (pig, human, and mouse). We observed that SEs were more significantly enriched by transcriptional initiation regions, TF binding sites, and SNPs than other cis-elements. Western breeds included fewer SEs in number, while more growth-related QTLs were associated with these SEs. Additionally, the SEs were highly tissue-specific, and were conserved in the liver among humans, pigs, and mice. We concluded that intense selection could concentrate functional SEs; thus, SEs could be applied as effective detection regions in genomic selection breeding. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
32. The role of super-enhancers in head and neck cancer and its potential therapeutic targets.
- Author
-
Zhou C, Guo G, and Li Q
- Abstract
Head and neck squamous cell carcinoma (HNSCC) poses a significant global health challenge, with over 660,000 new cases diagnosed and more than 325,000 deaths each year. Despite advances in treatment, long-term survival rates for HNSCC patients remain disappointingly low, underscoring the critical need for innovative therapeutic strategies. This review delves into the role of super-enhancers in HNSCC, highlighting their pivotal function in regulating key oncogenes such as KLF4, FOSL1, and ΔNp63, which are crucial for tumor progression and metastasis. Moreover, it explores how super-enhancers contribute to the maintenance of cancer stem cell characteristics by controlling genes like SOX2, BRD4. thereby promoting self-renewal and pluripotency.The study also underscores the potential of BET bromodomain inhibitors, exemplified by JQ1, and CDK7 inhibitors like THZ1, which demonstrate substantial therapeutic promise by effectively disrupting the function of super-enhancers in HNSCC. Overall, this research provides a comprehensive overview of the importance of super-enhancers in HNSCC., Competing Interests: Declaration of competing interest The authors declare no conflicts of interests., (Copyright © 2024 Elsevier Masson SAS. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
33. Expression of risk genes linked to vitamin D receptor super-enhancer regions and their association with phenotype severity in multiple sclerosis
- Author
-
Sarah M. Orton, Amarpreet Sangha, Mehul Gupta, Kristina Martens, Luanne M. Metz, A. P. J. de Koning, and Gerald Pfeffer
- Subjects
multiple sclerosis ,vitamin D deficiency ,super-enhancers ,vitamin D receptor (VDR) ,vitamin D ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Multiple sclerosis (MS) is a chronic debilitating neurological condition with a wide range of phenotype variability. A complex interplay of genetic and environmental factors contributes to disease onset and progression in MS patients. Vitamin D deficiency is a known susceptibility factor for MS, however the underlying mechanism of vitamin D-gene interactions in MS etiology is still poorly understood. Vitamin D receptor super-enhancers (VSEs) are enriched in MS risk variants and may modulate these environment-gene interactions. mRNA expression in total of 64 patients with contrasting MS severity was quantified in select genes. First, RNA-seq was performed on a discovery cohort (10 mild, 10 severe MS phenotype) and ten genes regulated by VSEs that have been linked to MS risk were analyzed. Four candidates showed a significant positive association (GRINA, PLEC, PARP10, and LRG1) in the discovery cohort and were then quantified using digital droplet PCR (ddPCR) in a validation cohort (33 mild, 11 severe MS phenotype). A significant differential expression persisted in the validation cohort for three of the VSE-MS genes: GRINA (p = 0.0138), LRG1 (p = 0.0157), and PLEC (p = 0.0391). In summary, genes regulated by VSE regions that contain known MS risk variants were shown to have differential expression based on disease severity (p
- Published
- 2022
- Full Text
- View/download PDF
34. Super-enhancers in esophageal carcinoma: Transcriptional addictions and therapeutic strategies.
- Author
-
Yang Shi, Meiqi Wang, Dan Liu, Ullah, Saif, Huiyu Yang, Bingrong Liu, and Xing Ma
- Subjects
CARCINOMA ,ADDICTIONS ,ESOPHAGEAL cancer ,CANCER cells ,GENE expression - Abstract
The tumorigenesis of esophageal carcinoma arises from transcriptional dysregulation would become exceptionally dependent on specific regulators of gene expression, which could be preferentially attributed to the larger noncoding cis-regulatory elements, i.e. super-enhancers (SEs). SEs, large genomic regulatory entity in close genomic proximity, are underpinned by control cancer cell identity. As a consequence, the transcriptional addictions driven by SEs could offer an Achilles' heel for molecular treatments on patients of esophageal carcinoma and other types of cancer as well. In this review, we summarize the recent findings about the oncogenic SEs upon which esophageal cancer cells depend, and discuss why SEs could be seen as the hallmark of cancer, how transcriptional dependencies driven by SEs, and what opportunities could be supplied based on this cancer-specific SEs. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
35. CDK12 orchestrates super‐enhancer‐associated CCDC137 transcription to direct hepatic metastasis in colorectal cancer.
- Author
-
Dai, Wei, Wu, Junhong, Peng, Xiaopeng, Hou, Wen, Huang, Hao, Cheng, Qilai, Liu, Zhiping, Luyten, Walter, Schoofs, Liliane, Zhou, Jingfeng, and Liu, Shenglan
- Subjects
- *
CYCLIN-dependent kinases , *LIVER metastasis , *COLORECTAL cancer , *RNA sequencing , *METASTASIS , *SOMATIC embryogenesis - Abstract
Background: Hepatic metastasis is the primary and direct cause of death in individuals with colorectal cancer (CRC) attribute to lack of effective therapeutic targets. The present study aimed to identify potential druggable candidate targets for patients with liver metastatic CRC. Methods: The transcriptional profiles of super‐enhancers (SEs) in primary and liver metastatic CRC were evaluated in publicly accessible CRC datasets. Immunohistochemistry of human CRC tissues was conducted to determine the expression level of CDK12. Cellular proliferation, survival and stemness were examined upon CDK12 inhibition by shCDK12 or a selective CDK12 inhibitor named SR‐4835 with multiple in vitro and in vivo assays. RNA sequencing and bioinformatics analyses were carried out to investigate the mechanisms of CDK12 inhibition in CRC cells. Results: We identified CDK12 as a driver gene for direct hepatic metastasis in CRC. Suppression of CDK12 led to robust inhibition of proliferation, survival and stemness. Mechanistically, CDK12 intervention preferentially repressed the transcription of SE‐associated genes. Integration of the SE landscape and RNA sequencing, BCL2L1 and CCDC137 were identified as SE‐associated oncogenic genes to strengthen the abilities of cellular survival, proliferation and stemness, eventually increasing liver metastasis of CRC. Conclusions: Our data highlight the potential of CDK12 and SE‐associated oncogenic transcripts as therapeutic targets for patients with liver metastatic CRC. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
36. Residual Neural Network in Genomics.
- Author
-
Sabba, Sara, Smara, Meroua, Benhacine, Mehdi, Terra, Loubna, and Terra, Zine Eddine
- Subjects
- *
COMPUTER vision , *CONVOLUTIONAL neural networks , *DEEP learning , *GENOMICS - Abstract
Residual neural network (ResNet) is a Deep Learning model introduced by He et al. in 2015 to enhance traditional convolutional neural networks proposed to solve computer vision problems. It uses skip connections over some layer blocks to avoid vanishing gradient problem. Currently, many researches are focused to test and prove the efficiency of the ResNet on different domains such as genomics. In fact, the study of human genomes provides important information on the detection of diseases and their best treatments. Therefore, most of the scientists opted for bioinformatics solutions to get results in a reasonable time. In this paper, our interest is to show the effectiveness of the ResNet model on genomics. For that, we propose two new ResNet models to enhance the results of two genomic problems previously resolved by CNN models. The obtained results are very promising and they proved the performance of our ResNet models compared to the CNN models. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
37. Genome-Wide Super-Enhancer-Based Analysis: Identification of Prognostic Genes in Oral Squamous Cell Carcinoma.
- Author
-
Saito, Tomoaki, Asai, Shunichi, Tanaka, Nozomi, Nohata, Nijiro, Minemura, Chikashi, Koma, Ayaka, Kikkawa, Naoko, Kasamatsu, Atsushi, Hanazawa, Toyoyuki, Uzawa, Katsuhiro, and Seki, Naohiko
- Subjects
- *
SQUAMOUS cell carcinoma , *CETUXIMAB , *GENES , *THERAPEUTICS , *CANCER relapse , *DRUG resistance , *PHARMACOGENOMICS , *GENE expression profiling - Abstract
Advanced-stage oral squamous cell carcinoma (OSCC) patients are treated with combination therapies, such as surgery, radiation, chemotherapy, and immunotherapy. However, OSCC cells acquire resistance to these treatments, resulting in local recurrence and distant metastasis. The identification of genes involved in drug resistance is essential for improving the treatment of this disease. In this study, we applied chromatin immunoprecipitation sequencing (ChIP-Seq) to profile active enhancers. For that purpose, we used OSCC cell lines that had been exposed to cetuximab for a prolonged period. In total, 64 chromosomal loci were identified as active super-enhancers (SE) according to active enhancer marker histone H3 lysine 27 acetylation (H3K27ac) ChIP-Seq. In addition, a total of 131 genes were located in SE regions, and 34 genes were upregulated in OSCC tissues by TCGA-OSCC analysis. Moreover, high expression of four genes (C9orf89; p = 0.035, CENPA; p = 0.020, PISD; p = 0.0051, and TRAF2; p = 0.0075) closely predicted a poorer prognosis for OSCC patients according to log-rank tests. Increased expression of the four genes (mRNA Z-score ≥ 0) frequently co-occurred in TCGA-OSCC analyses. The high and low expression groups of the four genes showed significant differences in prognosis, suggesting that there are clear differences in the pathways based on the underlying gene expression profiles. These data indicate that potential stratified therapeutic strategies could be used to overcome resistance to drugs (including cetuximab) and further improve responses in drug-sensitive patients. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
38. Functional analysis of the Vsx2 super-enhancer uncovers distinct cis-regulatory circuits controlling Vsx2 expression during retinogenesis.
- Author
-
Bian, Fuyun, Daghsni, Marwa, Fangfang Lu, Liu, Silvia, Gross, Jeffrey M., and Aldiri, Issam
- Subjects
- *
FUNCTIONAL analysis , *BIPOLAR cells , *CELL differentiation , *TRANSCRIPTION factors , *CELL proliferation , *HOMEOSTASIS - Abstract
Vsx2 is a transcription factor essential for retinal proliferation and bipolar cell differentiation, but the molecular mechanisms underlying its developmental roles are unclear. Here, we have profiled VSX2 genomic occupancy during mouse retinogenesis, revealing extensive retinal genetic programs associated with VSX2 during development. VSX2 binds and transactivates its enhancer in association with the transcription factor PAX6. Mice harboring deletions in the Vsx2 regulatory landscape exhibit specific abnormalities in retinal proliferation and in bipolar cell differentiation. In one of those deletions, a complete loss of bipolar cells is associated with a bias towards photoreceptor production. VSX2 occupies cis-regulatory elements nearby genes associated with photoreceptor differentiation and homeostasis in the adult mouse and human retina, including a conserved region nearby Prdm1, a factor implicated in the specification of rod photoreceptors and suppression of bipolar cell fate. VSX2 interacts with the transcription factor OTX2 and can act to suppress OTX2-dependent enhancer transactivation of the Prdm1 enhancer. Taken together, our analyses indicate that Vsx2 expression can be temporally and spatially uncoupled at the enhancer level, and they illuminate important mechanistic insights into how VSX2 is engaged with gene regulatory networks that are essential for retinal proliferation and cell fate acquisition. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
39. Cis-acting super-enhancer lncRNAs as biomarkers to early-stage breast cancer
- Author
-
Ali S. Ropri, Rebecca S. DeVaux, Jonah Eng, Sridar V. Chittur, and Jason I. Herschkowitz
- Subjects
Breast cancer progression ,Ductal carcinoma in situ ,Super-enhancer long non-coding RNAs ,Super-enhancers ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Increased breast cancer screening over the past four decades has led to a substantial rise in the diagnosis of ductal carcinoma in situ (DCIS). Although DCIS lesions precede invasive ductal carcinoma (IDC), they do not always transform into cancer. The current standard-of-care for DCIS is an aggressive course of therapy to prevent invasive and metastatic disease resulting in over-diagnosis and over-treatment. Thus, there is a critical need to identify functional determinants of progression of DCIS to IDC to allow discrimination between indolent and aggressive disease. Recent studies show that super-enhancers, in addition to promoting other gene transcription, are themselves transcribed producing super-enhancer associated long noncoding RNAs (SE-lncRNAs). These SE-lncRNAs can interact with their associated enhancer regions in cis and influence activities and expression of neighboring genes. Furthermore, they represent a novel, untapped group of therapeutic targets. Methods With an integrative analysis of enhancer loci with global expression of SE-lncRNAs in the MCF10A progression series, we have identified differentially expressed SE-lncRNAs which can identify mechanisms for DCIS to IDC progression. Furthermore, cross-referencing these SE-lncRNAs with patient samples in the The Cancer Genome Atlas (TCGA) database, we have unveiled 27 clinically relevant SE-lncRNAs that potentially interact with their enhancer to regulate nearby gene expression. To complement SE-lncRNA expression studies, we conducted an unbiased global analysis of super-enhancers that are acquired or lost in progression. Results Here we designate SE-lncRNAs RP11-379F4.4 and RP11-465B22.8 as potential markers of progression of DCIS to IDC through regulation of the expression of their neighboring genes (RARRES1 and miR-200b, respectively). Moreover, we classified 403 super-enhancer regions in MCF10A normal cells, 627 in AT1, 1053 in DCIS, and 320 in CA1 cells. Comparison analysis of acquired/lost super-enhancer regions with super-enhancer regions classified in 47 ER positive patients, 10 triple negative breast cancer (TNBC) patients, and 11 TNBC cell lines reveal critically acquired pathways including STAT signaling and NF-kB signaling. In contrast, protein folding, and local estrogen production are identified as major pathways lost in progression. Conclusion Collectively, these analyses identify differentially expressed SE-lncRNAs and acquired/lost super-enhancers in progression of breast cancer important for promoting DCIS lesions to IDC.
- Published
- 2021
- Full Text
- View/download PDF
40. Mapping the evolving landscape of super-enhancers during cell differentiation
- Author
-
Yan Kai, Bin E. Li, Ming Zhu, Grace Y. Li, Fei Chen, Yingli Han, Hye Ji Cha, Stuart H. Orkin, Wenqing Cai, Jialiang Huang, and Guo-Cheng Yuan
- Subjects
Super-enhancers ,Enhancer ,Differentiation ,Dynamics ,3D genome ,Hierarchy ,Biology (General) ,QH301-705.5 ,Genetics ,QH426-470 - Abstract
Abstract Background Super-enhancers are clusters of enhancer elements that play critical roles in the maintenance of cell identity. Current investigations on super-enhancers are centered on the established ones in static cell types. How super-enhancers are established during cell differentiation remains obscure. Results Here, by developing an unbiased approach to systematically analyze the evolving landscape of super-enhancers during cell differentiation in multiple lineages, we discover a general trend where super-enhancers emerge through three distinct temporal patterns: conserved, temporally hierarchical, and de novo. The three types of super-enhancers differ further in association patterns in target gene expression, functional enrichment, and 3D chromatin organization, suggesting they may represent distinct structural and functional subtypes. Furthermore, we dissect the enhancer repertoire within temporally hierarchical super-enhancers, and find enhancers that emerge at early and late stages are enriched with distinct transcription factors, suggesting that the temporal order of establishment of elements within super-enhancers may be directed by underlying DNA sequence. CRISPR-mediated deletion of individual enhancers in differentiated cells shows that both the early- and late-emerged enhancers are indispensable for target gene expression, while in undifferentiated cells early enhancers are involved in the regulation of target genes. Conclusions In summary, our analysis highlights the heterogeneity of the super-enhancer population and provides new insights to enhancer functions within super-enhancers.
- Published
- 2021
- Full Text
- View/download PDF
41. CDK12 orchestrates super‐enhancer‐associated CCDC137 transcription to direct hepatic metastasis in colorectal cancer
- Author
-
Wei Dai, Junhong Wu, Xiaopeng Peng, Wen Hou, Hao Huang, Qilai Cheng, Zhiping Liu, Walter Luyten, Liliane Schoofs, Jingfeng Zhou, and Shenglan Liu
- Subjects
CCDC137 ,CDK12 ,colorectal cancer ,liver metastasis ,super‐enhancers ,Medicine (General) ,R5-920 - Abstract
Abstract Background Hepatic metastasis is the primary and direct cause of death in individuals with colorectal cancer (CRC) attribute to lack of effective therapeutic targets. The present study aimed to identify potential druggable candidate targets for patients with liver metastatic CRC. Methods The transcriptional profiles of super‐enhancers (SEs) in primary and liver metastatic CRC were evaluated in publicly accessible CRC datasets. Immunohistochemistry of human CRC tissues was conducted to determine the expression level of CDK12. Cellular proliferation, survival and stemness were examined upon CDK12 inhibition by shCDK12 or a selective CDK12 inhibitor named SR‐4835 with multiple in vitro and in vivo assays. RNA sequencing and bioinformatics analyses were carried out to investigate the mechanisms of CDK12 inhibition in CRC cells. Results We identified CDK12 as a driver gene for direct hepatic metastasis in CRC. Suppression of CDK12 led to robust inhibition of proliferation, survival and stemness. Mechanistically, CDK12 intervention preferentially repressed the transcription of SE‐associated genes. Integration of the SE landscape and RNA sequencing, BCL2L1 and CCDC137 were identified as SE‐associated oncogenic genes to strengthen the abilities of cellular survival, proliferation and stemness, eventually increasing liver metastasis of CRC. Conclusions Our data highlight the potential of CDK12 and SE‐associated oncogenic transcripts as therapeutic targets for patients with liver metastatic CRC.
- Published
- 2022
- Full Text
- View/download PDF
42. Super-enhancer associated core regulatory circuits mediate susceptibility to retinoic acid in neuroblastoma cells
- Author
-
Roshna Lawrence Gomez, Laura M. Woods, Revathy Ramachandran, Ahmad N. Abou Tayoun, Anna Philpott, and Fahad R. Ali
- Subjects
neuroblastoma ,super-enhancers ,retinoic acid ,differentiation ,treatment-resistance ,Biology (General) ,QH301-705.5 - Abstract
Neuroblastoma is a pediatric tumour that accounts for more than 15% of cancer-related deaths in children. High-risk tumours are often difficult to treat, and patients’ survival chances are less than 50%. Retinoic acid treatment is part of the maintenance therapy given to neuroblastoma patients; however, not all tumours differentiate in response to retinoic acid. Within neuroblastoma tumors, two phenotypically distinct cell types have been identified based on their super-enhancer landscape and transcriptional core regulatory circuitries: adrenergic (ADRN) and mesenchymal (MES). We hypothesized that the distinct super-enhancers in these different tumour cells mediate differential response to retinoic acid. To this end, three different neuroblastoma cell lines, ADRN (MYCN amplified and non-amplified) and MES cells, were treated with retinoic acid, and changes in the super-enhancer landscape upon treatment and after subsequent removal of retinoic acid was studied. Using ChIP-seq for the active histone mark H3K27ac, paired with RNA-seq, we compared the super-enhancer landscape in cells that undergo neuronal differentiation in response to retinoic acid versus those that fail to differentiate and identified unique super-enhancers associated with neuronal differentiation. Among the ADRN cells that respond to treatment, MYCN-amplified cells remain differentiated upon removal of retinoic acid, whereas MYCN non-amplified cells revert to an undifferentiated state, allowing for the identification of super-enhancers responsible for maintaining differentiation. This study identifies key super-enhancers that are crucial for retinoic acid-mediated differentiation.
- Published
- 2022
- Full Text
- View/download PDF
43. HDAC4 degradation during senescence unleashes an epigenetic program driven by AP-1/p300 at selected enhancers and super-enhancers
- Author
-
Eros Di Giorgio, Harikrishnareddy Paluvai, Emiliano Dalla, Liliana Ranzino, Alessandra Renzini, Viviana Moresi, Martina Minisini, Raffaella Picco, and Claudio Brancolini
- Subjects
OIS ,Senescence ,Class IIa HDACs ,H3K27 ,Super-enhancers ,BRD4 ,Biology (General) ,QH301-705.5 ,Genetics ,QH426-470 - Abstract
Abstract Background Cellular senescence is a permanent state of replicative arrest defined by a specific pattern of gene expression. The epigenome in senescent cells is sculptured in order to sustain the new transcriptional requirements, particularly at enhancers and super-enhancers. How these distal regulatory elements are dynamically modulated is not completely defined. Results Enhancer regions are defined by the presence of H3K27 acetylation marks, which can be modulated by class IIa HDACs, as part of multi-protein complexes. Here, we explore the regulation of class IIa HDACs in different models of senescence. We find that HDAC4 is polyubiquitylated and degraded during all types of senescence and it selectively binds and monitors H3K27ac levels at specific enhancers and super-enhancers that supervise the senescent transcriptome. Frequently, these HDAC4-modulated elements are also monitored by AP-1/p300. The deletion of HDAC4 in transformed cells which have bypassed oncogene-induced senescence is coupled to the re-appearance of senescence and the execution of the AP-1/p300 epigenetic program. Conclusions Overall, our manuscript highlights a role of HDAC4 as an epigenetic reader and controller of enhancers and super-enhancers that supervise the senescence program. More generally, we unveil an epigenetic checkpoint that has important consequences in aging and cancer.
- Published
- 2021
- Full Text
- View/download PDF
44. A systematic dissection of the epigenomic heterogeneity of lung adenocarcinoma reveals two different subclasses with distinct prognosis and core regulatory networks
- Author
-
Chongze Yuan, Haojie Chen, Shiqi Tu, Hsin-Yi Huang, Yunjian Pan, Xiuqi Gui, Muyu Kuang, Xuxia Shen, Qiang Zheng, Yang Zhang, Chao Cheng, Hui Hong, Xiaoting Tao, Yizhou Peng, Xingxin Yao, Feilong Meng, Hongbin Ji, Zhen Shao, and Yihua Sun
- Subjects
Lung adenocarcinoma ,Classification model ,Epigenome ,Super-enhancers ,Core regulators ,Biology (General) ,QH301-705.5 ,Genetics ,QH426-470 - Abstract
Abstract Background Lung adenocarcinoma (LUAD) is a highly malignant and heterogeneous tumor that involves various oncogenic genetic alterations. Epigenetic processes play important roles in lung cancer development. However, the variation in enhancer and super-enhancer landscapes of LUAD patients remains largely unknown. To provide an in-depth understanding of the epigenomic heterogeneity of LUAD, we investigate the H3K27ac histone modification profiles of tumors and adjacent normal lung tissues from 42 LUAD patients and explore the role of epigenetic alterations in LUAD progression. Results A high intertumoral epigenetic heterogeneity is observed across the LUAD H3K27ac profiles. We quantitatively model the intertumoral variability of H3K27ac levels at proximal gene promoters and distal enhancers and propose a new epigenetic classification of LUAD patients. Our classification defines two LUAD subgroups which are highly related to histological subtypes. Group II patients have significantly worse prognosis than group I, which is further confirmed in the public TCGA-LUAD cohort. Differential RNA-seq analysis between group I and group II groups reveals that those genes upregulated in group II group tend to promote cell proliferation and induce cell de-differentiation. We construct the gene co-expression networks and identify group-specific core regulators. Most of these core regulators are linked with group-specific regulatory elements, such as super-enhancers. We further show that CLU is regulated by 3 group I-specific core regulators and works as a novel tumor suppressor in LUAD. Conclusions Our study systematically characterizes the epigenetic alterations during LUAD progression and provides a new classification model that is helpful for predicting patient prognosis.
- Published
- 2021
- Full Text
- View/download PDF
45. Super-Enhancers, Phase-Separated Condensates, and 3D Genome Organization in Cancer.
- Author
-
Tang, Seng Chuan, Vijayakumar, Udhaya, Zhang, Ying, and Fullwood, Melissa Jane
- Subjects
- *
SUPER enhancers , *TUMOR treatment , *CHROMOSOMES , *GENETICS , *GENE expression , *CELL proliferation , *TRANSCRIPTION factors - Abstract
Simple Summary: Gene expression is primarily controlled at the level of transcription, largely due to binding of transcription factors to specific sites on DNA, namely super-enhancers and promoters. Super-enhancers and enhancers can regulate distal target genes via chromatin looping mechanisms. These long-range enhancer–promoter interactions are often mediated by phase-separated condensates. In this review, we summarize recent insights into super-enhancers and phase-separated condensates and their roles in the transcriptional activation of genes involved in development and diseases such as cancer. In addition, we survey a broad array of drugs that target super-enhancers, three-dimensional genome organization, and phase-separated condensates that could be potentially used for the treatment of cancer. 3D chromatin organization plays an important role in transcription regulation and gene expression. The 3D genome is highly maintained by several architectural proteins, such as CTCF, Yin Yang 1, and cohesin complex. This structural organization brings regulatory DNA elements in close proximity to their target promoters. In this review, we discuss the 3D chromatin organization of super-enhancers and their relationship to phase-separated condensates. Super-enhancers are large clusters of DNA elements. They can physically contact with their target promoters by chromatin looping during transcription. Multiple transcription factors can bind to enhancer and promoter sequences and recruit a complex array of transcriptional co-activators and RNA polymerase II to effect transcriptional activation. Phase-separated condensates of transcription factors and transcriptional co-activators have been implicated in assembling the transcription machinery at particular enhancers. Cancer cells can hijack super-enhancers to drive oncogenic transcription to promote cell survival and proliferation. These dysregulated transcriptional programs can cause cancer cells to become highly dependent on transcriptional regulators, such as Mediator and BRD4. Moreover, the expression of oncogenes that are driven by super-enhancers is sensitive to transcriptional perturbation and often occurs in phase-separated condensates, supporting therapeutic rationales of targeting SE components, 3D genome organization, or dysregulated condensates in cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
46. Super-Enhancer-Associated Long Non-Coding RNA LINC01485 Promotes Osteogenic Differentiation of Human Bone Marrow Mesenchymal Stem Cells by Regulating MiR-619-5p/RUNX2 Axis.
- Author
-
Gu, Wenli, Jiang, Xiao, Wang, Wei, Mujagond, Prabhakar, Liu, Jingpeng, Mai, Zhaoyi, Tang, Hai, li, Simin, Xiao, Hui, and Zhao, Jianjiang
- Subjects
MESENCHYMAL stem cells ,LINCRNA ,RUNX proteins - Abstract
Objective: To investigate the mechanisms of super-enhancer-associated LINC01485/miR-619-5p/RUNX2 signaling axis involvement in osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs). Methods: Osteogenic differentiation of hBMSCs was induced in vitro. The expression levels of LINC01485 and miR-619-5p during osteogenesis were measured using quantitative real-time polymerase chain reaction (qRT-PCR). Osteogenic differentiation was examined by qRT-PCR, western blot, alkaline phosphatase (ALP) staining, ALP activity measurement, and Alizarin Red S (ARS) staining assays. Thereafter, the effects of LINC01485 and miR-619-5p on osteogenic differentiation of hBMSCs were evaluated by performing loss- and gain-of-function experiments. Subsequently, a fluorescence in situ hybridization (FISH) assay was employed to determine the cellular localization of LINC01485. Bioinformatics analysis, RNA antisense purification (RAP) assay, and dual-luciferase reporter assays were conducted to analyze the interactions of LINC01485, miR-619-5p, and RUNX2. Rescue experiments were performed to further delineate the role of the competitive endogenous RNA (ceRNA) signaling axis consisting of LINC01485/miR-619-5p/RUNX2 in osteogenic differentiation of hBMSCs. Results: The expression of LINC01485 was up-regulated during osteogenic differentiation of hBMSCs. The overexpression of LINC01485 promoted osteogenic differentiation of hBMSCs by up-regulating the expression of osteogenesis-related genes [e.g., runt-related transcription factor 2 (RUNX2), osterix (OSX), collagen type 1 alpha 1 (COL1A1), osteocalcin (OCN), and osteopontin (OPN)], and increasing the activity of ALP. ALP staining and ARS staining were also found to be increased upon overexpression of LINC01485. The opposing results were obtained upon LINC01485 interference in hBMSCs. miR-619-5p was found to inhibit osteogenic differentiation. FISH assay displayed that LINC01485 was mainly localized in the cytoplasm. RAP assay results showed that LINC01485 bound to miR-619-5p, and dual-luciferase reporter assay verified that LINC01485 bound to miR-619-5p, while miR-619-5p and RUNX2 bound to each other. Rescue experiments illustrated that LINC01485 could promote osteogenesis by increasing RUNX2 expression by sponging miR-619-5p. Conclusion: LINC01485 could influence RUNX2 expression by acting as a ceRNA of miR-619-5p, thereby promoting osteogenic differentiation of hBMSCs. The LINC01485/miR-619-5p/RUNX2 axis might comprise a novel target in the bone tissue engineering field. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
47. Chromatin profiling reveals relocalization of lysine-specific demethylase 1 by an oncogenic fusion protein
- Author
-
Emily R. Theisen, Julia Selich-Anderson, Kyle R. Miller, Jason M. Tanner, Cenny Taslim, Kathleen I. Pishas, Sunil Sharma, and Stephen L. Lessnick
- Subjects
chromatin ,lsd1 ,epigenetics ,ews/fli ,super-enhancers ,ewing sarcoma ,Genetics ,QH426-470 - Abstract
Paediatric cancers commonly harbour quiet mutational landscapes and are instead characterized by single driver events such as the mutation of critical chromatin regulators, expression of oncohistones, or expression of oncogenic fusion proteins. These events ultimately promote malignancy through disruption of normal gene regulation and development. The driver protein in Ewing sarcoma, EWS/FLI, is an oncogenic fusion and transcription factor that reshapes the enhancer landscape, resulting in widespread transcriptional dysregulation. Lysine-specific demethylase 1 (LSD1) is a critical functional partner for EWS/FLI as inhibition of LSD1 reverses the transcriptional activity of EWS/FLI. However, how LSD1 participates in fusion-directed epigenomic regulation and aberrant gene activation is unknown. We now show EWS/FLI causes dynamic rearrangement of LSD1 and we uncover a role for LSD1 in gene activation through colocalization at EWS/FLI binding sites throughout the genome. LSD1 is integral to the establishment of Ewing sarcoma super-enhancers at GGAA-microsatellites, which ubiquitously overlap non-microsatellite loci bound by EWS/FLI. Together, we show that EWS/FLI induces widespread changes to LSD1 distribution in a process that impacts the enhancer landscape throughout the genome.
- Published
- 2021
- Full Text
- View/download PDF
48. An intriguing characteristic of enhancer-promoter interactions
- Author
-
Amlan Talukder, Haiyan Hu, and Xiaoman Li
- Subjects
Enhancers ,Promoters ,Enhancer clusters ,Super-enhancers ,Biotechnology ,TP248.13-248.65 ,Genetics ,QH426-470 - Abstract
Abstract Background It is still challenging to predict interacting enhancer-promoter pairs (IEPs), partially because of our limited understanding of their characteristics. To understand IEPs better, here we studied the IEPs in nine cell lines and nine primary cell types. Results By measuring the bipartite clustering coefficient of the graphs constructed from these experimentally supported IEPs, we observed that one enhancer is likely to interact with either none or all of the target genes of another enhancer. This observation implies that enhancers form clusters, and every enhancer in the same cluster synchronously interact with almost every member of a set of genes and only this set of genes. We perceived that an enhancer can be up to two megabase pairs away from other enhancers in the same cluster. We also noticed that although a fraction of these clusters of enhancers do overlap with super-enhancers, the majority of the enhancer clusters are different from the known super-enhancers. Conclusions Our study showed a new characteristic of IEPs, which may shed new light on distal gene regulation and the identification of IEPs.
- Published
- 2021
- Full Text
- View/download PDF
49. Super-Enhancer-Associated Long Non-Coding RNA LINC01485 Promotes Osteogenic Differentiation of Human Bone Marrow Mesenchymal Stem Cells by Regulating MiR-619-5p/RUNX2 Axis
- Author
-
Wenli Gu, Xiao Jiang, Wei Wang, Prabhakar Mujagond, Jingpeng Liu, Zhaoyi Mai, Hai Tang, Simin li, Hui Xiao, and Jianjiang Zhao
- Subjects
LINC01485 ,miR-619-5p ,RUNX2 ,super-enhancers ,bone marrow mesenchymal stem cells ,osteogenesis ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
ObjectiveTo investigate the mechanisms of super-enhancer-associated LINC01485/miR-619-5p/RUNX2 signaling axis involvement in osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs).MethodsOsteogenic differentiation of hBMSCs was induced in vitro. The expression levels of LINC01485 and miR-619-5p during osteogenesis were measured using quantitative real-time polymerase chain reaction (qRT-PCR). Osteogenic differentiation was examined by qRT-PCR, western blot, alkaline phosphatase (ALP) staining, ALP activity measurement, and Alizarin Red S (ARS) staining assays. Thereafter, the effects of LINC01485 and miR-619-5p on osteogenic differentiation of hBMSCs were evaluated by performing loss- and gain-of-function experiments. Subsequently, a fluorescence in situ hybridization (FISH) assay was employed to determine the cellular localization of LINC01485. Bioinformatics analysis, RNA antisense purification (RAP) assay, and dual-luciferase reporter assays were conducted to analyze the interactions of LINC01485, miR-619-5p, and RUNX2. Rescue experiments were performed to further delineate the role of the competitive endogenous RNA (ceRNA) signaling axis consisting of LINC01485/miR-619-5p/RUNX2 in osteogenic differentiation of hBMSCs.ResultsThe expression of LINC01485 was up-regulated during osteogenic differentiation of hBMSCs. The overexpression of LINC01485 promoted osteogenic differentiation of hBMSCs by up-regulating the expression of osteogenesis-related genes [e.g., runt-related transcription factor 2 (RUNX2), osterix (OSX), collagen type 1 alpha 1 (COL1A1), osteocalcin (OCN), and osteopontin (OPN)], and increasing the activity of ALP. ALP staining and ARS staining were also found to be increased upon overexpression of LINC01485. The opposing results were obtained upon LINC01485 interference in hBMSCs. miR-619-5p was found to inhibit osteogenic differentiation. FISH assay displayed that LINC01485 was mainly localized in the cytoplasm. RAP assay results showed that LINC01485 bound to miR-619-5p, and dual-luciferase reporter assay verified that LINC01485 bound to miR-619-5p, while miR-619-5p and RUNX2 bound to each other. Rescue experiments illustrated that LINC01485 could promote osteogenesis by increasing RUNX2 expression by sponging miR-619-5p.ConclusionLINC01485 could influence RUNX2 expression by acting as a ceRNA of miR-619-5p, thereby promoting osteogenic differentiation of hBMSCs. The LINC01485/miR-619-5p/RUNX2 axis might comprise a novel target in the bone tissue engineering field.
- Published
- 2022
- Full Text
- View/download PDF
50. A holistic view of mouse enhancer architectures reveals analogous pleiotropic effects and correlation with human disease
- Author
-
Siddharth Sethi, Ilya E. Vorontsov, Ivan V. Kulakovskiy, Simon Greenaway, John Williams, Vsevolod J. Makeev, Steve D. M. Brown, Michelle M. Simon, and Ann-Marie Mallon
- Subjects
Super-enhancers ,Typical-enhancers ,Tissue-specificity ,Expression ,Phenotypes ,Protein-protein interactions ,Biotechnology ,TP248.13-248.65 ,Genetics ,QH426-470 - Abstract
Abstract Background Efforts to elucidate the function of enhancers in vivo are underway but their vast numbers alongside differing enhancer architectures make it difficult to determine their impact on gene activity. By systematically annotating multiple mouse tissues with super- and typical-enhancers, we have explored their relationship with gene function and phenotype. Results Though super-enhancers drive high total- and tissue-specific expression of their associated genes, we find that typical-enhancers also contribute heavily to the tissue-specific expression landscape on account of their large numbers in the genome. Unexpectedly, we demonstrate that both enhancer types are preferentially associated with relevant ‘tissue-type’ phenotypes and exhibit no difference in phenotype effect size or pleiotropy. Modelling regulatory data alongside molecular data, we built a predictive model to infer gene-phenotype associations and use this model to predict potentially novel disease-associated genes. Conclusion Overall our findings reveal that differing enhancer architectures have a similar impact on mammalian phenotypes whilst harbouring differing cellular and expression effects. Together, our results systematically characterise enhancers with predicted phenotypic traits endorsing the role for both types of enhancers in human disease and disorders.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.