1
|
Martínez-Romero R, González-Chávez SA, Urías-Rubí VR, Gómez-Moreno VM, Blanco-Cantero MF, Bernal-Velázquez HM, Luévano-González A, Pacheco-Tena C. Microarray Analysis of Visceral Adipose Tissue in Obese Women Reveals Common Crossroads Among Inflammation, Metabolism, Addictive Behaviors, and Cancer: AKT3 and MAPK1 Cross Point in Obesity. J Obes 2024; 2024:4541071. [PMID: 39484291 PMCID: PMC11527533 DOI: 10.1155/2024/4541071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/16/2024] [Indexed: 11/03/2024] Open
Abstract
Background: Visceral adipose tissue (VAT) abnormalities are directly associated with obesity-associated disorders. The underlying mechanisms that confer increased pathological risk to VAT in obesity have not been fully described. Methods: A case-control study was conducted that included 10 women with obesity (36.80 ± 7.39 years, BMI ≥ 30 kg/m2) and 10 women of normal weight (32.70 ± 9.45 years, BMI < 24.9 kg/m2). RNA was extracted from greater omentum biopsies, and, using a DNA microarray, differential transcriptomic expression of VAT in women with obesity was evaluated taking as a reference that of women with normal weight. The differentially expressed genes (DEGs) were classified into functional biological processes and signaling pathways; moreover, the protein-protein interaction (PPI) networks were integrated for a deeper analysis of the pathways and genes involved in the central obesity-associated disorders. The expression of TNF-α, MAPK, and AKT proteins was also quantified in VAT. Results: The VAT of women with obesity had 3808 DEGs, mainly associated with the cellular process of inflammation and carbohydrates and lipid metabolism. Overexpressed genes were associated with inflammatory, metabolic, hormonal, neuroendocrine, carcinogenic, and infectious pathways. Cellular processes related to addictive behaviors were notable. MAPK and PI3K-AKT pathways were overexpressed, and Mapk1 and Akt3 genes were common crossing points among obesity-associated disorders' pathways. The increased expression of MAPK, AKT, and TNF proteins was confirmed in the VAT of women with obesity. Conclusion: VAT confers a complex and blended pathogenic transcriptomic profile in obese patients, where abnormal processes are mainly controlled by activating intracellular signaling pathways that exhibit a high degree of redundancy. Identifying shared cross points between those pathways could allow specific targeting treatments to exert a widespread effect over multiple pathogenic processes.
Collapse
Affiliation(s)
- Rolando Martínez-Romero
- Laboratorio PABIOM, Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Chihuahua, Mexico
| | - Susana Aideé González-Chávez
- Laboratorio PABIOM, Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Chihuahua, Mexico
| | - Victor Roberto Urías-Rubí
- Laboratorio PABIOM, Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Chihuahua, Mexico
| | | | | | | | - Arturo Luévano-González
- Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Chihuahua, Mexico
| | - César Pacheco-Tena
- Laboratorio PABIOM, Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Chihuahua, Mexico
| |
Collapse
|
2
|
Shinde A, Chandak N, Singh J, Roy M, Mane M, Tang X, Vasiyani H, Currim F, Gohel D, Shukla S, Goyani S, Saranga MV, Brindley DN, Singh R. TNF-α induced NF-κB mediated LYRM7 expression modulates the tumor growth and metastatic ability in breast cancer. Free Radic Biol Med 2024; 211:158-170. [PMID: 38104742 DOI: 10.1016/j.freeradbiomed.2023.12.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/19/2023]
Abstract
Tumor microenvironment (TME) of solid tumors including breast cancer is complex and contains a distinct cytokine pattern including TNF-α, which determines the progression and metastasis of breast tumors. The metastatic potential of triple negative breast cancer subtypes is high as compared to other subtypes of breast cancer. NF-κB is key transcription factor regulating inflammation and mitochondrial bioenergetics including oxidative phosphorylation (OXPHOS) genes which determine its oxidative capacity and generating reducing equivalents for synthesis of key metabolites for proliferating breast cancer cells. The differential metabolic adaptation and OXPHOS function of breast cancer subtypes in inflammatory conditions and its contribution to metastasis is not well understood. Here we demonstrated that different subunits of NF-κB are differentially expressed in subtypes of breast cancer patients. RELA, one of the major subunits in regulation of the NF-κB pathway is positively correlated with high level of TNF-α in breast cancer patients. TNF-α induced NF-κB regulates the expression of LYRM7, an assembly factor for mitochondrial complex III. Downregulation of LYRM7 in MDA-MB-231 cells decreases mitochondrial super complex assembly and enhances ROS levels, which increases the invasion and migration potential of these cells. Further, in vivo studies using Infliximab, a monoclonal antibody against TNF-α showed decreased expression of LYRM7 in tumor tissue. Large scale breast cancer databases and human patient samples revealed that LYRM7 levels decreased in triple negative breast cancer patients compared to other subtypes and is determinant of survival outcome in patients. Our results indicate that TNF-α induced NF-κB is a critical regulator of LYRM7, a major factor for modulating mitochondrial functions under inflammatory conditions, which determines growth and survival of breast cancer cells.
Collapse
Affiliation(s)
- Anjali Shinde
- Department of Biochemistry, Faculty of Science, The MS University of Baroda, Vadodara, 390002, Gujarat, India
| | - Nisha Chandak
- Department of Biochemistry, Faculty of Science, The MS University of Baroda, Vadodara, 390002, Gujarat, India
| | - Jyoti Singh
- Department of Biochemistry, Faculty of Science, The MS University of Baroda, Vadodara, 390002, Gujarat, India
| | - Milton Roy
- Institute for Cell Engineering, John Hopkins University School of Medicine, 733 North Broadway, MRB 731, Baltimore, MD, 21205, USA
| | - Minal Mane
- Department of Biochemistry, Faculty of Science, The MS University of Baroda, Vadodara, 390002, Gujarat, India
| | - Xiaoyun Tang
- Cancer Research Institute of Northern Alberta, Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G2S2, Canada
| | - Hitesh Vasiyani
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA-23284, USA
| | - Fatema Currim
- Department of Biochemistry, Faculty of Science, The MS University of Baroda, Vadodara, 390002, Gujarat, India
| | - Dhruv Gohel
- Department of Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Shatakshi Shukla
- Department of Biochemistry, Faculty of Science, The MS University of Baroda, Vadodara, 390002, Gujarat, India
| | - Shanikumar Goyani
- Department of Biochemistry, Faculty of Science, The MS University of Baroda, Vadodara, 390002, Gujarat, India
| | - M V Saranga
- Department of Biochemistry, Faculty of Science, The MS University of Baroda, Vadodara, 390002, Gujarat, India
| | - David N Brindley
- Cancer Research Institute of Northern Alberta, Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G2S2, Canada
| | - Rajesh Singh
- Department of Biochemistry, Faculty of Science, The MS University of Baroda, Vadodara, 390002, Gujarat, India; Department of Molecular and Human Genetics, Banaras Hindu University (BHU) (IoE), Varanasi, 221005, UP, India.
| |
Collapse
|
3
|
Zhang D, Yu N, Yuan Z, Li W, Sun X, Zou Q, Li X, Liu Z, Zhang W, Gao R. stMMR: accurate and robust spatial domain identification from spatially resolved transcriptomics with multimodal feature representation. Gigascience 2024; 13:giae089. [PMID: 39607984 PMCID: PMC11604062 DOI: 10.1093/gigascience/giae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/03/2024] [Accepted: 10/23/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Deciphering spatial domains using spatially resolved transcriptomics (SRT) is of great value for characterizing and understanding tissue architecture. However, the inherent heterogeneity and varying spatial resolutions present challenges in the joint analysis of multimodal SRT data. RESULTS We introduce a multimodal geometric deep learning method, named stMMR, to effectively integrate gene expression, spatial location, and histological information for accurate identifying spatial domains from SRT data. stMMR uses graph convolutional networks and a self-attention module for deep embedding of features within unimodality and incorporates similarity contrastive learning for integrating features across modalities. CONCLUSIONS Comprehensive benchmark analysis on various types of spatial data shows superior performance of stMMR in multiple analyses, including spatial domain identification, pseudo-spatiotemporal analysis, and domain-specific gene discovery. In chicken heart development, stMMR reconstructed the spatiotemporal lineage structures, indicating an accurate developmental sequence. In breast cancer and lung cancer, stMMR clearly delineated the tumor microenvironment and identified marker genes associated with diagnosis and prognosis. Overall, stMMR is capable of effectively utilizing the multimodal information of various SRT data to explore and characterize tissue architectures of homeostasis, development, and tumor.
Collapse
Affiliation(s)
- Daoliang Zhang
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan 250061, China
| | - Na Yu
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan 250061, China
| | - Zhiyuan Yuan
- Institute of Science and Technology for Brain-Inspired Intelligence, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Fudan University, Shanghai 200433, China
| | - Wenrui Li
- MOE Key Lab of Bioinformatics and Bioinformatics Division of BNRIST, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Xue Sun
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan 250061, China
| | - Qi Zou
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan 250061, China
| | - Xiangyu Li
- School of Software Engineering, Beijing Jiaotong University, Beijing 100044, China
| | - Zhiping Liu
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan 250061, China
| | - Wei Zhang
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan 250061, China
| | - Rui Gao
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan 250061, China
| |
Collapse
|
4
|
Masood M, Masood MBE, Us Subah N, Shabbir M, Paracha RZ, Rafiq M. Investigating isoform switching in RHBDF2 and its role in neoplastic growth in breast cancer. PeerJ 2022; 10:e14124. [PMID: 36452073 PMCID: PMC9703992 DOI: 10.7717/peerj.14124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 09/06/2022] [Indexed: 11/27/2022] Open
Abstract
Background Breast cancer is the second leading cause of cancer-related deaths globally, and its prevalence rates are increasing daily. In the past, studies predicting therapeutic drug targets for cancer therapy focused on the assumption that one gene is responsible for producing one protein. Therefore, there is always an immense need to find promising and novel anti-cancer drug targets. Furthermore, proteases have an integral role in cell proliferation and growth because the proteolysis mechanism is an irreversible process that aids in regulating cellular growth during tumorigenesis. Therefore, an inactive rhomboid protease known as iRhom2 encoded by the gene RHBDF2 can be considered an important target for cancer treatment. Speculatively, previous studies on gene expression analysis of RHBDF2 showed heterogenous behaviour during tumorigenesis. Consistent with this, several studies have reported the antagonistic role of iRhom2 in tumorigenesis, i.e., either they are involved in negative regulation of EGFR ligands via the ERAD pathway or positively regulate EGFR ligands via the EGFR signalling pathway. Additionally, different opinions suggest iRhom2 mediated cleavage of EGFR ligands takes place TACE dependently or TACE independently. However, reconciling these seemingly opposing roles is still unclear and might be attributed to more than one transcript isoform of iRhom2. Methods To observe the differences at isoform resolution, the current strategy identified isoform switching in RHBDF2 via differential transcript usage using RNA-seq data during breast cancer initiation and progression. Furthermore, interacting partners were found via correlation and enriched to explain their antagonistic role. Results Isoform switching was observed at DCIS, grade 2 and grade 3, from canonical to the cub isoform. Neither EGFR nor ERAD was found enriched. However, pathways leading to TACE-dependent EGFR signalling pathways were more observant, specifically MAPK signalling pathways, GPCR signalling pathways, and toll-like receptor pathways. Nevertheless, it was noteworthy that during CTCs, the cub isoform switches back to the canonical isoform, and the proteasomal degradation pathway and cytoplasmic ribosomal protein pathways were significantly enriched. Therefore, it could be inferred that cub isoform functions during cancer initiation in EGFR signalling. In contrast, during metastasis, where invasion is the primary task, the isoform switches back to the canonical isoform.
Collapse
Affiliation(s)
- Mehar Masood
- School of Interdisciplinary Engineering and Sciences, National University of Sciences and Technology, Islamabad, Pakistan,Faculty of Rehabilitation & Allied Health Sciences, Riphah International University, Islamabad, Pakistan
| | - Madahiah Bint E Masood
- School of Interdisciplinary Engineering and Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Noor Us Subah
- School of Interdisciplinary Engineering and Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Maria Shabbir
- Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Rehan Zafar Paracha
- School of Interdisciplinary Engineering and Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Mehak Rafiq
- School of Interdisciplinary Engineering and Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| |
Collapse
|
5
|
Ren YM, Duan YH, Sun YB, Yang T, Hou WY, Liu C, Tian MQ. mRNA and long non-coding RNA expression profiles of rotator cuff tear patients reveal inflammatory features in long head of biceps tendon. BMC Med Genomics 2022; 15:140. [PMID: 35725478 PMCID: PMC9210618 DOI: 10.1186/s12920-022-01292-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 06/15/2022] [Indexed: 02/06/2024] Open
Abstract
Background This study aimed to identify the differentially expressed mRNAs and lncRNAs in inflammatory long head of biceps tendon (LHBT) of rotator cuff tear (RCT) patients and further explore the function and potential targets of differentially expressed lncRNAs in biceps tendon pathology. Methods Human gene expression microarray was made between 3 inflammatory LHBT samples and 3 normal LHBT samples from RCT patients. GO analysis and KEGG pathway analysis were performed to annotate the function of differentially expressed mRNAs. The real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was admitted to verify their expression. LncRNA-mRNA co-expression network, cis-acting element, trans-acting element and transcription factor (TF) regulation analysis were constructed to predict the potential molecular regulatory mechanisms and targets for LHB tendinitis. Results 103 differentially expressed lncRNAs and mRNAs, of which 75 were up-regulated and 28 were down-regulated, were detected to be differentially expressed in LHBT. The expressions of 4 most differentially expressed lncRNAs (A2MP1, LOC100996671, COL6A4P, lnc-LRCH1-5) were confirmed by qRT-PCR. GO functional analysis indicated that related lncRNAs and mRNAs were involved in the biological processes of regulation of innate immune response, neutrophil chemotaxis, interleukin-1 cell response and others. KEGG pathway analysis indicated that related lncRNAs and mRNAs were involved in MAPK signaling pathway, NF-kappa B signaling pathway, cAMP signaling pathway and others. TF regulation analysis revealed that COL6A4P2, A2MP1 and LOC100996671 target NFKB2. Conclusions LlncRNA-COL6A4P2, A2MP1 and LOC100996671 may regulate the inflammation of LHBT in RCT patients through NFKB2/NF-kappa B signaling pathway, and preliminarily revealed the pathological molecular mechanism of tendinitis of LHBT. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01292-y.
Collapse
Affiliation(s)
- Yi-Ming Ren
- Department of Joint and Sport Medicine, Tianjin Union Medical Center, Nankai University Affiliated People's Hospital, Jieyuan Road 190, Hongqiao District, Tianjin, 300121, People's Republic of China
| | - Yuan-Hui Duan
- Department of Joint and Sport Medicine, Tianjin Union Medical Center, Nankai University Affiliated People's Hospital, Jieyuan Road 190, Hongqiao District, Tianjin, 300121, People's Republic of China
| | - Yun-Bo Sun
- Department of Joint and Sport Medicine, Tianjin Union Medical Center, Nankai University Affiliated People's Hospital, Jieyuan Road 190, Hongqiao District, Tianjin, 300121, People's Republic of China
| | - Tao Yang
- Department of Joint and Sport Medicine, Tianjin Union Medical Center, Nankai University Affiliated People's Hospital, Jieyuan Road 190, Hongqiao District, Tianjin, 300121, People's Republic of China
| | - Wei-Yu Hou
- Department of Joint and Sport Medicine, Tianjin Union Medical Center, Nankai University Affiliated People's Hospital, Jieyuan Road 190, Hongqiao District, Tianjin, 300121, People's Republic of China
| | - Chang Liu
- Schoole of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Meng-Qiang Tian
- Department of Joint and Sport Medicine, Tianjin Union Medical Center, Nankai University Affiliated People's Hospital, Jieyuan Road 190, Hongqiao District, Tianjin, 300121, People's Republic of China.
| |
Collapse
|
6
|
Xu R, Yang X, Tao Y, Luo W, Xiong Y, He L, Zhou F, He Y. Analysis of the Molecular Mechanism of Evodia rutaecarpa Fruit in the Treatment of Nasopharyngeal Carcinoma Using Network Pharmacology and Molecular Docking. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:6277139. [PMID: 35463684 PMCID: PMC9020960 DOI: 10.1155/2022/6277139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 12/08/2022]
Abstract
Background Nasopharyngeal carcinoma (NPC), a neoplasm of the head and neck, has high incidence and mortality rates in East and Southeast Asia. Evodia rutaecarpa is a tree native to Korea and China, and its fruit (hereafter referred to as Evodia) exhibits remarkable antitumour properties. However, little is known about its mechanism of action in NPC. In this study, we employed network pharmacology to identify targets of active Evodia compounds in nasopharyngeal carcinoma and generate an interaction network. Methods The active ingredients of Evodia and targets in NPC were obtained from multiple databases, and an interaction network was constructed via the Cytoscape and STRING databases. The key biological processes and signalling pathways were predicted using Gene Ontology and Kyoto Encyclopaedia of Genes and Genomes pathway enrichment analyses. Molecular docking technology was used to identify the affinity and activity of target genes, and The Cancer Genome Atlas and Human Protein Atlas databases were used to analyse differential expression. Cell Counting Kit-8 (CCK-8) and Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) dual-fluorescence staining were used for experimental verification. Results Active Evodia compounds included quercetin, isorhamnetin, and evodiamine, and important NPC targets included MAPK14, AKT1, RELA, MAPK1, JUN, and p53, which were enriched in lipid and atherosclerosis signalling pathways. Additionally, we verified the high affinity and activity of the active compounds through molecular docking, and the target proteins were verified using immunohistochemistry and differential expression analyses. Furthermore, CCK-8 assays and Annexin V-FITC/PI dual-fluorescence staining showed that isorhamnetin inhibited the proliferation of NPC cells and induced apoptosis. Conclusion Our results identified the molecular mechanisms of Evodia and demonstrated its ability to alter the proliferation and apoptosis of NPC cells through multiple targets and pathways, thereby providing evidence for the clinical application of Evodia.
Collapse
Affiliation(s)
- Runshi Xu
- Hunan University of Chinese Medicine, Hanpu Science and Education Park, Changsha, China
| | - Ximing Yang
- Hunan University of Chinese Medicine, Hanpu Science and Education Park, Changsha, China
| | - Yangyang Tao
- Hunan University of Chinese Medicine, Hanpu Science and Education Park, Changsha, China
| | - Wang Luo
- Hunan University of Chinese Medicine, Hanpu Science and Education Park, Changsha, China
| | - Yu Xiong
- Hunan University of Chinese Medicine, Hanpu Science and Education Park, Changsha, China
| | - Lan He
- First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, China
- Hunan Engineering Technology Research Center for Prevention & Treatment of Ophthalmology and Otolaryngology Diseases and Visual Function Protection with Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Fangliang Zhou
- Hunan University of Chinese Medicine, Hanpu Science and Education Park, Changsha, China
- Hunan Key Laboratory for Prevention & Treatment of Ophthalmology and Otolaryngology Diseases with Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yingchun He
- Hunan University of Chinese Medicine, Hanpu Science and Education Park, Changsha, China
- Hunan Engineering Technology Research Center for Prevention & Treatment of Ophthalmology and Otolaryngology Diseases and Visual Function Protection with Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Key Laboratory for Prevention & Treatment of Ophthalmology and Otolaryngology Diseases with Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
7
|
Qazi S, Raza K. In silico approach to understand epigenetics of POTEE in ovarian cancer. J Integr Bioinform 2021; 18:jib-2021-0028. [PMID: 34788504 PMCID: PMC8709732 DOI: 10.1515/jib-2021-0028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/04/2021] [Indexed: 12/20/2022] Open
Abstract
Ovarian cancer is the third leading cause of cancer-related deaths in India. Epigenetics mechanisms seemingly plays an important role in ovarian cancer. This paper highlights the crucial epigenetic changes that occur in POTEE that get hypomethylated in ovarian cancer. We utilized the POTEE paralog mRNA sequence to identify major motifs and also performed its enrichment analysis. We identified 6 motifs of varying lengths, out of which only three motifs, including CTTCCAGCAGATGTGGATCA, GGAACTGCC, and CGCCACATGCAGGC were most likely to be present in the nucleotide sequence of POTEE. By enrichment and occurrences identification analyses, we rectified the best match motif as CTTCCAGCAGATGT. Since there is no experimentally verified structure of POTEE paralog, thus, we predicted the POTEE structure using an automated workflow for template-based modeling using the power of a deep neural network. Additionally, to validate our predicted model we used AlphaFold predicted POTEE structure and observed that the residual stretch starting from 237-958 had a very high confidence per residue. Furthermore, POTEE predicted model stability was evaluated using replica exchange molecular dynamic simulation for 50 ns. Our network-based epigenetic analysis discerns only 10 highly significant, direct, and physical associators of POTEE. Our finding aims to provide new insights about the POTEE paralog.
Collapse
Affiliation(s)
- Sahar Qazi
- Department of Computer Science, Jamia Millia Islamia, New Delhi 110025, India
| | - Khalid Raza
- Department of Computer Science, Jamia Millia Islamia, New Delhi 110025, India
| |
Collapse
|
8
|
Molecular epigenetic dynamics in breast carcinogenesis. Arch Pharm Res 2021; 44:741-763. [PMID: 34392501 DOI: 10.1007/s12272-021-01348-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022]
Abstract
Breast cancer has become one of the most common dreadful diseases that target women across the globe. The most obvious reasons we associate with it are either genetic mutations or dysregulation of pathways. However, there is yet another domain that has a significant role in influencing the genetic mutations and pathways. Epigenetic mechanisms influence these pathways either independently or in association with genetic mutations, thereby expediting the process of breast carcinogenesis. Breast cancer is governed by various transduction pathways such as PI3K/AKT/mTOR, NOTCH, β Catenin, NF-kB, Hedgehog, etc. There are many proteins as well that serve to be tumor suppressors but somehow lose their ability to function. This may be because of either genetic mutation or a process that represses their function. Apart from these, there are a lot of individual factors like puberty, breastfeeding, abortion, parity, circadian rhythm, alcohol consumption, pollutants, and obesity that drive these mutations and hence alter the pathways. Epigenetic mechanisms like DNA methylation, histone modifications, and lncRNAs directly or indirectly bring alterations in the proteins that are involved in the pathways. They do this by either promoting the transcription of genes or by repressing it at the ground genetic level that advances breast carcinogenesis. Epigenetics precedes genetic mutation in driving carcinogenesis and so, it needs to be explored further to diversify the possibilities of target specific treatments. In this review, the general role of DNA methylation, histone modification, and lncRNAs in breast cancer and their role in influencing the oncogenic signaling pathways along with the various factors governing them have been discussed for a better understanding of the role of epigenetics in breast carcinogenesis.
Collapse
|