1
|
Prabhakaran R, Thamarai R, Sivasamy S, Dhandayuthapani S, Batra J, Kamaraj C, Karthik K, Shah MA, Mallik S. Epigenetic frontiers: miRNAs, long non-coding RNAs and nanomaterials are pioneering to cancer therapy. Epigenetics Chromatin 2024; 17:31. [PMID: 39415281 PMCID: PMC11484394 DOI: 10.1186/s13072-024-00554-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024] Open
Abstract
Cancer has arisen from both genetic mutations and epigenetic changes, making epigenetics a crucial area of research for innovative cancer prevention and treatment strategies. This dual perspective has propelled epigenetics into the forefront of cancer research. This review highlights the important roles of DNA methylation, histone modifications and non-coding RNAs (ncRNAs), particularly microRNAs (miRNAs) and long non-coding RNAs, which are key regulators of cancer-related gene expression. It explores the potential of epigenetic-based therapies to revolutionize patient outcomes by selectively modulating specific epigenetic markers involved in tumorigenesis. The review examines promising epigenetic biomarkers for early cancer detection and prognosis. It also highlights recent progress in oligonucleotide-based therapies, including antisense oligonucleotides (ASOs) and antimiRs, to precisely modulate epigenetic processes. Furthermore, the concept of epigenetic editing is discussed, providing insight into the future role of precision medicine for cancer patients. The integration of nanomedicine into cancer therapy has been explored and offers innovative approaches to improve therapeutic efficacy. This comprehensive review of recent advances in epigenetic-based cancer therapy seeks to advance the field of precision oncology, ultimately culminating in improved patient outcomes in the fight against cancer.
Collapse
Affiliation(s)
- Rajkumar Prabhakaran
- Central Research Facility, Santosh Deemed to be University, Ghaziabad, UP, India
| | - Rajkumar Thamarai
- UGC Dr. D.S. Kothari Postdoctoral Fellow, Department of Animal Science, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu, 627012, India
| | - Sivabalan Sivasamy
- Central Research Facility, Santosh Deemed to be University, Ghaziabad, UP, India
| | | | - Jyoti Batra
- Central Research Facility, Santosh Deemed to be University, Ghaziabad, UP, India.
| | - Chinnaperumal Kamaraj
- Interdisciplinary Institute of Indian System of Medicine, Directorate of Research, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India.
| | - Krishnasamy Karthik
- Department of Mechanical Engineering, Vel Tech Rangarajan Dr. Sagunthala R&D Institute of Science and Technology, Chennai, India
| | - Mohd Asif Shah
- Department of Economics, Kardan University, Parwane Du, 1001, Kabul, Afghanistan.
- Division of Research and Development, Lovely Professional University, Phagwara, Punjab, 144001, India.
- Centre of Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India.
| | - Saurav Mallik
- Department of Environmental Health, Harvard T H Chan School of Public Health, Boston, Massachusetts, 02115, United States.
- Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
2
|
Cheng Y, Liang X, Bi X, Liu C, Yang Y. Identification ATP5F1D as a Biomarker Linked to Diagnosis, Prognosis, and Immune Infiltration in Endometrial Cancer Based on Data-Independent Acquisition (DIA) Analysis. Biochem Genet 2024; 62:4215-4236. [PMID: 38265620 DOI: 10.1007/s10528-023-10646-9] [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: 07/12/2023] [Accepted: 12/19/2023] [Indexed: 01/25/2024]
Abstract
In developed countries, endometrial cancer (EC) is the most prevalent gynecological cancer. ATP5F1D is a subunit of ATP synthase, as well as an important component of the mitochondrial electron transport chain (ETC). ETC plays a compelling role in carcinogenesis. To date, little is known about the role of ATP5F1D in EC. We undertook data-independent acquisition mass spectrometry (DIA-MS) of 20 EC patients, comprising 10 high-grade and 10 low-grade cancer tissues. Biological functions of differentially expressed genes (DEGs) were analyzed by GO and KEGG. The expression level, clinicopathological features, diagnostic potency, prognostic value, RNA modifications, immune characteristics, and therapy response of ATP5F1D were investigated. In total, 77 DEGs were acquired by DIA analysis, which were closely related to regulating immune response and metabolic pathways. Among the five genes (NDUFB8, SLC26A2, RAF1, ATP5F1D, and GSTM5) involving in reactive oxygen species pathway, ATP5F1D showed the most significant differential expression (2.903-fold change). We found ATP5F1D had a high diagnostic value and was associated with a favorable prognosis in EC patients. After analyzing the RNA modifications of ATP5F1D, revealing a negative regulation between them. Additionally, ATP5F1D was closely related to tumor immune infiltration. Our results suggested T-cell dysfunction and TAM-M2 polarization might be the important mechanisms of ATP5F1D to facilitate tumor immune escape. Noticeably, EC patients with ATP5F1D-high expression had better immune treatment responses and were more sensitive to chemotherapy drugs. ATP5F1D can be used as a biomarker for diagnosis, prognosis, and immune infiltration of EC, and offers a crucial reference for personalized treatment of EC patients.
Collapse
Affiliation(s)
- Yuemei Cheng
- The First Clinical Medical College of Lanzhou University, Department of Obstetrics and Gynecology, Gansu Provincial Clinical Research Center for Gynecological Oncology, Lanzhou, 730000, Gansu, China
| | - Xiaolei Liang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Gansu Provincial Clinical Research Center for Gynecological Oncology, Lanzhou, 730000, Gansu, China
| | - Xuehan Bi
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Gansu Provincial Clinical Research Center for Gynecological Oncology, Lanzhou, 730000, Gansu, China
| | - Chang Liu
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Gansu Provincial Clinical Research Center for Gynecological Oncology, Lanzhou, 730000, Gansu, China
| | - Yongxiu Yang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Gansu Provincial Clinical Research Center for Gynecological Oncology, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
3
|
Lin Y, Liu M, Deng P, Zhang J. TET1 mediated m5C modification of RelB aggravates cerebral ischemia/reperfusion-induced neuroinflammation through regulating microglia polarization. Cell Signal 2024; 120:111210. [PMID: 38705503 DOI: 10.1016/j.cellsig.2024.111210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/11/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Microglia mediated neuroinflammation is one of the major contributors to brain damage in cerebral ischemia reperfusion injury (CI/RI). Recently, RNA modification was found to contribute to the regulation of microglia polarization and the subsequent development of cerebral I/R neuroinflammation. Herein, we investigated the effect and mechanism of m5C RNA modification in the microglia induced CI/RI neuroinflammation. We found that the m5C RNA modification levels decreased in the primary microglia isolated from a mouse model of intraluminal middle cerebral artery occlusion/reperfusion (MCAO/R) and the BV2 microglial cells subjected to oxygen-glucose deprivation and reoxygenation (OGD/R), and this change was accompanied by an increase in the M1/M2 polarization ratio. Furthermore, the expression of m5C demethylase TET1 in microglia increased, which promoted M1 polarization but impeded M2 polarization. Mechanistically, the higher TET1 expression decreased the m5C modification level of RelB and enhanced its mRNA stability, which subsequently increased the M1/M2 polarization ratio. In conclusion, this study provides insight into the role of m5C RNA modification in the pathogenesis of cerebral I/R neuroinflammation and may deepen our understanding on clinical therapy targeting the TET1-RelB axis.
Collapse
Affiliation(s)
- Yan Lin
- Department of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Elctronic Science and Technology of China, 32 2rd Setion of Yihuan Road West, Qingyang District, Chengdu, 610072, Sichuan Province, China
| | - Mei Liu
- Department of Neurology, The Six People's Hospital of Chengdu, Chengdu, Sichuan Province 610072, China
| | - Pinghuan Deng
- Department of Encephalopathy, Dechang County Hospital of Traditional Chinese Medicine, Dechang, Sichuan Province 615500, China
| | - Jinzhi Zhang
- Department of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Elctronic Science and Technology of China, 32 2rd Setion of Yihuan Road West, Qingyang District, Chengdu, 610072, Sichuan Province, China..
| |
Collapse
|
4
|
Xie H, Deng YM, Li JY, Xie KH, Tao T, Zhang JF. Predicting the risk of primary Sjögren's syndrome with key N7-methylguanosine-related genes: A novel XGBoost model. Heliyon 2024; 10:e31307. [PMID: 38803884 PMCID: PMC11128997 DOI: 10.1016/j.heliyon.2024.e31307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024] Open
Abstract
Objectives N7-methylguanosine (m7G) plays a crucial role in mRNA metabolism and other biological processes. However, its regulators' function in Primary Sjögren's Syndrome (PSS) remains enigmatic. Methods We screened five key m7G-related genes across multiple datasets, leveraging statistical and machine learning computations. Based on these genes, we developed a prediction model employing the extreme gradient boosting decision tree (XGBoost) method to assess PSS risk. Immune infiltration in PSS samples was analyzed using the ssGSEA method, revealing the immune landscape of PSS patients. Results The XGBoost model exhibited high accuracy, AUC, sensitivity, and specificity in both training, test sets and extra-test set. The decision curve confirmed its clinical utility. Our findings suggest that m7G methylation might contribute to PSS pathogenesis through immune modulation. Conclusions m7G regulators play an important role in the development of PSS. Our study of m7G-realted genes may inform future immunotherapy strategies for PSS.
Collapse
Affiliation(s)
- Hui Xie
- Department of Radiotherapy, Affiliated Hospital (Clinical College) of Xiangnan University, Chenzhou, 423000, PR China
- Faulty of Applied Sciences, Macao Polytechnic University, Macao, 999078, PR China
| | - Yin-mei Deng
- Department of Nursing, Affiliated Hospital (Clinical College) of Xiangnan University, Chenzhou, 423000, PR China
| | - Jiao-yan Li
- Department of Rheumatology and Clinical Immunology, The First Hospital of Changsha, 410005, Changsha, PR China
| | - Kai-hong Xie
- Department of Oncology, Affiliated Hospital (Clinical College) of Xiangnan University, Chenzhou, 423000, PR China
| | - Tan Tao
- Faulty of Applied Sciences, Macao Polytechnic University, Macao, 999078, PR China
| | - Jian-fang Zhang
- Department of Physical Examination, Center for Disease Control and Prevention of Beihu District, Chenzhou, 423000, PR China
| |
Collapse
|
5
|
Wang R, Xue W, Kan F, Zhang H, Wang D, Wang L, Wang J. NSUN2 affects diabetic retinopathy progression by regulating MUC1 expression through RNA m 5C methylation. J Transl Med 2024; 22:476. [PMID: 38764010 PMCID: PMC11103891 DOI: 10.1186/s12967-024-05287-4] [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: 02/22/2024] [Accepted: 05/10/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND Diabetic retinopathy (DR) is the leading cause of blinding eye disease among working adults and is primarily attributed to the excessive proliferation of microvessels, which leads to vitreous hemorrhage and retinal traction, thereby significantly impairing patient vision. NSUN2-mediated RNA m5C methylation is implicated in various diseases, and in this investigation, we focused on elucidating the impact of NSUN2 on the regulation of the expression of the downstream gene MUC1, specifically through RNA m5C methylation, on the progression of DR. METHOD Utilizing Microarray analysis, we examined patient vitreous fluid to pinpoint potential therapeutic targets for DR. Differential expression of NSUN2 was validated through qRT-PCR, Western blot, and immunofluorescence in human tissue, animal tissue, and cell model of DR. The relationship between NSUN2 and DR was explored in vitro and in vivo through gene knockdown and overexpression. Various techniques, such as MeRIP-qPCR and dot blot, were applied to reveal the downstream targets and mechanism of action of NSUN2. RESULTS The levels of both NSUN2 and RNA m5C methylation were significantly elevated in the DR model. Knockdown of NSUN2 mitigated DR lesion formation both in vitro and in vivo. Mechanistically, NSUN2 promoted MUC1 expression by binding to the RNA m5C reader ALYREF. Knockdown of ALYREF resulted in DR lesion alterations similar to those observed with NSUN2 knockdown. Moreover, MUC1 overexpression successfully reversed a series of DR alterations induced by NSUN2 silencing. CONCLUSIONS NSUN2 regulates the expression of MUC1 through ALYREF-mediated RNA m5C methylation, thereby regulating the progression of DR and providing a new option for the treatment of DR in the future.
Collapse
Affiliation(s)
- Runze Wang
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Wei Xue
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Feifei Kan
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Huiying Zhang
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Di Wang
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Lei Wang
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, 150000, China.
| | - Jianwen Wang
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, 150000, China.
| |
Collapse
|
6
|
Ge Y, Weng H, Sun Y, Wu M. Integrated single-cell and spatial transcriptomic analysis reveals YBX1 drives immune regulation in GBM progression. Heliyon 2024; 10:e29451. [PMID: 38628755 PMCID: PMC11019236 DOI: 10.1016/j.heliyon.2024.e29451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
The RNA modification 5-methylcytosine (m5C) is widespread across various RNA types, significantly impacting RNA stability and translational efficiency. Accumulating evidence highlights its significant role within the tumorigenesis and progression of multiple malignancies. Nevertheless, the specific process through m5C is implicated in Glioblastoma (GBM) remains unclear. We conducted acomprehensive analysis of m5C expression distribution in single-cell GBM data. Our findings revealed elevated m5C scores in GBM single-cell data compared to the normal group. Additionally, multiple tumors exhibited significantly higher m5C scores than the normal group. Moreover, there was a positive correlation observed between the m5C score and inflammation score. m5C regulatory factor YBX1 exhibited a heightened expression in GBM, correlating closely with metastatic tendencies and an unfavorable prognosis across various cancer types. YBX1 has different biological functions in myeloid cells 1 and myeloid cells 2. YBX1 may act as immunosuppressive regulator by inhibiting the NF-κB pathway and inflammatory response in myeloid cells 1. YBX1 is essential for immune infiltrates, which creates a highly immunosuppressive tumor microenvironment by TNF signaling pathway in myeloid cells 2. YBX1+ neoplastic cells promote cell proliferation by NF-κB pathway. APOE mediates the interaction of YBX1+ myeloid cells and neoplastic cells by NF-κB.
Collapse
Affiliation(s)
- Yanshan Ge
- Hunan Provincial Tumor Hospital / the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410008, Hunan, China
| | - Huiting Weng
- Department of Clinical Nursing, The Second Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Yingnan Sun
- Hunan Provincial Tumor Hospital / the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
| | - Minghua Wu
- Hunan Provincial Tumor Hospital / the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410008, Hunan, China
| |
Collapse
|
7
|
Liu HT, Rau CS, Liu YW, Hsieh TM, Huang CY, Chien PC, Lin HP, Wu CJ, Chuang PC, Hsieh CH. Deciphering the Divergent Gene Expression Landscapes of m6A/m5C/m1A Methylation Regulators in Hepatocellular Carcinoma Through Single-Cell and Bulk RNA Transcriptomic Analysis. J Hepatocell Carcinoma 2023; 10:2383-2395. [PMID: 38164510 PMCID: PMC10758181 DOI: 10.2147/jhc.s448047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction RNA modifications mediated by the m6A, m1A, and m5C regulatory genes are crucial for the progression of malignancy. This study aimed to explore the expression of regulator genes for m6A/m5C/m1A methylation at the single-cell level and to validate their expression in cancerous and adjacent para-cancerous liver tissues of adult patients with HCC who underwent tumor resection. Methods The bulk sequencing from The Cancer Genome Atlas (TCGA) database and the single-cell RNA sequencing (scRNA-seq) data obtained from the Gene Expression Omnibus (GEO) database were used to identify the dysregulated m6A/m5C/m1A genes for hepatocellular carcinoma (HCC). A real-time polymerase chain reaction (real-time PCR) was used to measure the expression of dysregulated m6A/m5C/m1A genes in collected human HCC tissues and compared with adjacent para-cancerous liver tissues. Immune cell infiltration with these significantly expressed methylation-related genes was evaluated using Timer2.0. Results A discrepancy in m6A/m5C/m1A gene expression was observed between bulk sequencing and scRNA-seq. The clustered heatmap of the scRNA-seq-identified dysregulated m6A/m5C/m1A genes in TCGA cohort revealed heterogeneous expression of these methylation regulators within the cancer, whereas their expression in the adjacent liver tissues was more homogeneous. The real-time PCR validated the significant overexpression of DNMT1, NSUN5, TRMT6, IGF2BP1, and IGFBP3, which were identified using scRNA-seq, and IGFBP2, which was identified using bulk sequencing. These dysregulated methylation genes are mainly correlated with the infiltration of natural killer cells. Discussion This study suggests that cellular diversity inside tumors contributes to the discrepancy in the expression of methylation regulator genes between traditional bulk sequencing and scRNA-seq. This study identified five regulatory genes that will be the focus of further studies regarding the function of m6A/m5C/m1A in HCC.
Collapse
Affiliation(s)
- Hang-Tsung Liu
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Cheng-Shyuan Rau
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Yueh-Wei Liu
- Department of General Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Ting-Min Hsieh
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Chun-Ying Huang
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Peng-Chen Chien
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Hui-Ping Lin
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Chia-Jung Wu
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Pei-Chin Chuang
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301, Taiwan
| | - Ching-Hua Hsieh
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| |
Collapse
|