1
|
Alluri A, Saxena P, Mishra A, Gutti RK. Association of long non-coding RNA in lipid metabolism: Implications in leukemia. Int J Biochem Cell Biol 2025; 184:106785. [PMID: 40246061 DOI: 10.1016/j.biocel.2025.106785] [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: 05/10/2024] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
Cancer has high mortality rate and occupies second position among major diseases. Despite extensive research and therapies, in every nook and corner of the world, death rate is increasing exponentially. Hallmarks of cancer are benchmarks of cancer cells describing the fundamental principle and capabilities of the cells transforming from normal to malignant tumour. One of the major ones among them is the deregulation of cellular metabolism or metabolic reprogramming, involving alterations in glucose and lipid metabolism. Progressive research in this area has visualized the vital role of lncRNAs in lipid metabolism with respect to AML. lncRNAs involve in various cellular processes and also contribute for significant functions of the cell like chromatin remodelling, transcriptional activation and repression, gene regulation, immune response, cell differentiation, and cell cycle regulation, in addition to oncogenic processes such as proliferation, angiogenesis, migration, and apoptosis. Structural similarities are observed among mRNAs and lncRNAs in terms of poly A-tail and 5' cap however protein-coding regions are lacking. A large body of evidence has shown that lncRNAs directly or indirectly mediate lipid metabolism by activating downstream genes. Considering their potential involvement in leukemia, these lncRNAs can be explored and considered as biomarkers for therapeutics, prognosis, and diagnosis. The present review is planned to summarize the functional classification of lncRNAs, the role of lipid metabolism in cancer, different lncRNAs involved in leukemia, and different cancer types related to lipid metabolism.
Collapse
Affiliation(s)
- Anjani Alluri
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, (PO) Gachibowli, Hyderabad, TS 500046, India
| | - Pallavi Saxena
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, (PO) Gachibowli, Hyderabad, TS 500046, India
| | - Amit Mishra
- Department of Bioscience & Bioengineering, Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, RJ 342037, India
| | - Ravi Kumar Gutti
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, (PO) Gachibowli, Hyderabad, TS 500046, India.
| |
Collapse
|
2
|
Alkhathami AG, Altalbawy FMA, Rizaev JA, H M, Jaswinder Kaur, Ali Jeddoa ZM, Jabir MS, Jawad SF, Yumashev A, Zwamel AH. An overview of lncRNA GAPLINC's role in human cancer growth and metastasis. Arch Biochem Biophys 2025; 771:110506. [PMID: 40513977 DOI: 10.1016/j.abb.2025.110506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2025] [Revised: 06/05/2025] [Accepted: 06/11/2025] [Indexed: 06/16/2025]
Abstract
Long non-coding RNAs (lncRNAs) have emerged as critical regulators in cancer biology, influencing tumor initiation, progression, and metastasis. Among them, GAPLINC (Gastric Adenocarcinoma Predictive Long Intergenic Non-coding RNA) has garnered attention due to its aberrant expression across various malignancies and its association with poor clinical outcomes. While several studies have highlighted GAPLINC's oncogenic roles through interactions with microRNAs and involvement in epithelial-mesenchymal transition (EMT), a comprehensive synthesis of its mechanistic functions and clinical implications remains limited. This review addresses this limitation by systematically analyzing current findings on GAPLINC's molecular mechanisms, regulatory networks, and functional roles in different cancer types, including gastric, colorectal, non-small cell lung cancer (NSCLC), glioma, renal cell carcinoma (RCC), and esophageal squamous cell carcinoma (ESCC). We further evaluate the potential of GAPLINC as a diagnostic, prognostic, and therapeutic target, providing an integrated perspective that consolidates existing knowledge and identifies directions for future research. By highlighting both common and cancer-specific mechanisms involving GAPLINC, this review adds new insights into its significance as a versatile biomarker and therapeutic target in oncology.
Collapse
Affiliation(s)
- Ali G Alkhathami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
| | - Jasur Alimdjanovich Rizaev
- Department of Public health and Healthcare management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan
| | - Malathi H
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Jaswinder Kaur
- Chandigarh pharmacy college, Chandigarh Group of colleges, jhanjeri, Mohali 140307, punjab, India
| | | | - Majid S Jabir
- Department of applied sciences/ university of technology- Iraq
| | - Sabrean F Jawad
- Department of Pharmacy, Al-Mustaqbal University College, 51001 Hillah, Babylon, Iraq
| | | | - Ahmed Hussein Zwamel
- Medical laboratory technique college, the Islamic University, Najaf, Iraq*; Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
3
|
Zhang M, Ju B, Wang X, Zhou S, Zhao H, Wang ZL, Li X. Global knowledge mapping and emerging research trends in non-coding RNAs related to animal and plant male sterility: A visual analysis of CiteSpace maps. Medicine (Baltimore) 2025; 104:e42612. [PMID: 40489841 DOI: 10.1097/md.0000000000042612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/11/2025] Open
Abstract
Animal and plant male sterility is a complex and closely studied phenomenon that significantly impacts species survival and reproduction. Advances in biotechnology and molecular biology have deepened our understanding of gene expression regulation, particularly the role of noncoding RNAs (ncRNAs). This study aims to systematically review and analyze the published literature on ncRNAs in relation to both animal and plant sterility using bibliometric methods. A bibliometric analysis was conducted with CiteSpace 6.2.R6, Scimago Graphica, VOSviewer 1.6.18, and Microsoft Excel 2016 to identify research hotspots, key developments, and emerging trends. Data were retrieved from the Web of Science Core Collection on March 3, 2024, covering publications from 2005 to 2023. The analysis revealed a consistent increase in annual publications on ncRNA research in both plant and animal fields, with China and the United States leading in publication volume. Notable scholars include Professor Abu-Halima, a prominent figure in ncRNA research related to animal male sterility, and Professor Meyers, a key contributor to plant male sterility research. Journals such as PLoS ONE serve as major platforms for disseminating findings on animal male sterility, while The Plant Cell plays a similar role for plant male sterility. Analysis of cited literature and keyword trends highlighted significant themes, including gene regulation and the application of novel technologies. At present, new technologies, model organisms, and gene regulation remain major research hotspots. Meanwhile, disease diagnosis, disease treatment, and crop improvement are emerging as important directions for future research.
Collapse
Affiliation(s)
- Mingzhao Zhang
- Department of Andrology, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Department of Clinical Medicine, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Baojun Ju
- Department of Andrology, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Department of Clinical Medicine, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xiangyu Wang
- Department of Andrology, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Department of Clinical Medicine, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Shuxi Zhou
- Department of Andrology, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Department of Clinical Medicine, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Haobin Zhao
- Department of Clinical Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Zu Long Wang
- Department of Andrology, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xiao Li
- Department of Andrology, The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
4
|
Temaj G, Saha S, Chichiarelli S, Telkoparan-Akillilar P, Nuhii N, Hadziselimovic R, Saso L. Polyploid giant cancer cells: Underlying mechanisms, signaling pathways, and therapeutic strategies. Crit Rev Oncol Hematol 2025; 213:104802. [PMID: 40484156 DOI: 10.1016/j.critrevonc.2025.104802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2025] [Revised: 05/26/2025] [Accepted: 06/04/2025] [Indexed: 06/19/2025] Open
Abstract
Polyploid giant cancer cells (PGCCs) are characterized by enlarged nuclei, association with tumors, and resistance to treatment, contributing significantly to cellular heterogeneity. These cells arise from endoreplication and cell fusion, often triggered by stressors such as radiation and chemotherapy. PGCCs exhibit chromosomal instability and aneuploidy, leading to poor prognosis in various cancers. Key features include the ability to produce progeny cells via amitotic division and the expression of cancer stem cell markers. PGCC formation and function involve signaling pathways like cell fusion (GCM1/syncytin-1), cell cycle control, stress response, and EMT. Understanding these pathways is crucial for identifying therapeutic targets. Current therapeutic strategies targeting PGCCs involve drugs like azacitidine, decitabine, and zoledronic acid, as well as DNMT inhibitors in combination therapies. These approaches aim to reverse drug resistance and enhance antitumor efficacy. Furthermore, microRNAs (miRNAs) are pivotal in regulating gene expression and influencing the cell cycle, proliferation, and apoptosis. Cataloging miRNAs and understanding their function is critical for developing potential cancer treatments. Researchers are exploring miRNA-based modulation of signaling pathways to block tumor growth. This review highlights the complex biology of PGCCs and emphasizes the need for targeted therapies to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Gazmend Temaj
- Faculty of Pharmacy, College UBT, Prishtina 10000, Kosovo
| | - Sarmistha Saha
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, Uttar Pradesh 00185, India.
| | - Silvia Chichiarelli
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome 00185, Italy
| | | | - Nexhibe Nuhii
- Department of Pharmacy, Faculty of Medical Sciences, State University of Tetovo, Tetovo 1200, North Macedonia
| | - Rifat Hadziselimovic
- Faculty of Science, University of Sarajevo, Sarajevo 71000, Bosnia and Herzegovina
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", La Sapienza University, Rome 00185, Italy
| |
Collapse
|
5
|
Saadh MJ, Bishoyi AK, Rekha MM, Verma A, Nanda A, Panigrahi R, Verma R, Gabble BC. Dual roles of long non-coding RNAs in thyroid cancer: regulation of programmed cell death pathways. Med Oncol 2025; 42:217. [PMID: 40407962 DOI: 10.1007/s12032-025-02750-0] [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: 02/14/2025] [Accepted: 04/28/2025] [Indexed: 06/01/2025]
Abstract
Thyroid cancer (TC) represents the most common endocrine malignancy; however, the intricacies of its carcinogenesis pose significant challenges to therapeutic interventions. A comprehensive understanding of the molecular mechanisms that drive TC progression is crucial for the development of effective treatment strategies, especially considering the increasingly recognized role of non-coding RNAs (ncRNAs) in oncogenesis. Notwithstanding recent advancements, the regulatory functions of long non-coding RNAs (lncRNAs) and their interactions with microRNAs (miRNAs) in the context of TC are not yet fully elucidated. This review aims to address this knowledge deficiency by investigating the dual roles of lncRNAs in the pathogenesis of TC, specifically their regulation of programmed cell death (PCD) pathways. Current literature indicates that disrupted competitive endogenous RNA (ceRNA) networks are involved in drug resistance, epithelial-mesenchymal transition (EMT), as well as tumor proliferation, angiogenesis, invasion, and metastasis in TC. The basis of cancer therapy-induced tumor cell elimination is programmed cell death (PCD), which includes well-studied processes such as apoptosis, autophagy, and ferroptosis as well as novel pathways, such as cuproptosis, immunogenic cell death (ICD), and PANoptosis. Recent research has shown the critical function of long non-coding RNAs (lncRNAs) in modifying these several PCD pathways, impacting TC growth and therapy response. This review synthesizes evidence on how lncRNAs regulate PCD to influence TC progression and therapeutic outcomes. Additionally, we examine the clinical relevance of lncRNAs in TC, highlighting their potential as biomarkers for diagnosis and prognosis, therapeutic targets, and contributors to drug resistance, while emphasizing recent advancements in this field.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan.
| | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat, 360003, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Ashish Verma
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - Anima Nanda
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Rajashree Panigrahi
- Department of Microbiology, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Rajni Verma
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab, 140307, India
| | - Baneen C Gabble
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
| |
Collapse
|
6
|
Yang R, Xu Y, Zhu F, Ma X, Fan T, Wang HL. Gut microbiome, a potential modulator of neuroepigenome. J Nutr Biochem 2025; 144:109961. [PMID: 40412567 DOI: 10.1016/j.jnutbio.2025.109961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 04/01/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025]
Abstract
Gut microbiome has a considerable impact on the central nervous system via the "gut-brain axis." Neuroepigenome emerges as the interface between environment and genes, potentially help conveying the signals derived from the microbiome to the brain tissue. While only a limited number of studies have implicated epigenetic roles in the gut-brain axis, this review explores how gut microbiome might impact various brain-based epigenetic mechanisms, including DNA methylation, histone modification, ncRNA and RNA methylation, notably in the context of the specific neural complications. Among the epigenetic mechanisms, histone acetylation was most well-studied with respect to its relationships with gut microbiome, exerting a dynamic influence on gene expression in the brain. Furthermore, the pathways connecting gut bacteria to neuroepigenome were summarized, highlighting the roles of metabolites such as butyrate, propionate, acetate, lactate, and folate. Of particular interest, the roles of butyrate are emphasized due to their outstanding inhibitory activity towards histone deacetylases (HDACs), among other mechanisms. It is worth noting that some indirect gut-brain pathways may also be associated with the interplay between microbiome and neuroepigenome, while IL-6 has been found to effectively transmit microbe-derived signals to histone methylation in brains. Finally, we recapitulate the future perspectives critical to understanding this gut-brain crosstalk, such as clarifying the cause-and-effect relationship, bacterial cross-feeding within the gut, and the mechanisms underlying the site-specific histone modification in the brain. Together, this review attempts to consolidate our current knowledge about the "microbiome-neuroepigenome interplay" and propose a conceptual pathway to decipher the gut-brain axis in various neurological conditions.
Collapse
Affiliation(s)
- Ruili Yang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yi Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| | - Feng Zhu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaojing Ma
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Tingting Fan
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Hui-Li Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| |
Collapse
|
7
|
Alimohammadi M, Fooladi AAI, Mafi A, Alavioun SM, Cho WC, Reiter RJ, Khormizi FZ, Yousefi T, Farahani N, Khoshnazar SM, Hushmandi K. Long noncoding RNAs and HPV-related cervical cancer: Uncovering molecular mechanisms and clinical applications. Transl Oncol 2025; 55:102363. [PMID: 40121995 PMCID: PMC11982485 DOI: 10.1016/j.tranon.2025.102363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 03/08/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025] Open
Abstract
Cervical cancer (CC) is the primary cause of cancer-related mortality among women in developing countries and is the most prevalent disease linked to human papillomavirus (HPV). Over 70 % of CC cases result from persistent infections with high-risk HPV types. The virus typically targets the mucocutaneous epithelium, generating viral particles in mature epithelial cells, which leads to disruptions in normal cell-cycle regulation and promotes uncontrolled cellular proliferation. This unchecked cell division results in the accumulation of genetic damage, contributing to the pathogenesis of CC. While HPV infection is a key etiological factor, the disease's progression also necessitates the involvement of genetic and epigenetic influences. One of the epigenetic regulators, long noncoding RNAs (lncRNAs), are characterized by transcripts exceeding 200 nucleotides. These molecules play crucial roles in various cellular processes, including transcription regulation, RNA metaboli35 per 100,000sm, and apoptosis. Investigating the specific roles of lncRNAs in modulating gene expression related to the oncogenic mechanisms of CC, particularly in the context of high-risk HPV infections, may provide valuable insights for diagnostic and therapeutic advancements. Herein, we first review key molecular mechanisms by which lncRNAs interfere with CC-related HPV development. Then, diagnostic, prognostic, and therapeutic potentials of these lncRNA molecules will be highlighted in depth. The focus of this article is on the role of lncRNAs associated with HPV-related CC, emphasizing the investigation of signaling pathways and their underlying molecular mechanisms. Furthermore, we explore the therapeutic potential and diagnostic relevance of the most significant lncRNAs in the context of CC, thereby highlighting their importance in advancing treatment strategies and improving patient outcomes.
Collapse
Affiliation(s)
- Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Mafi
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyedeh Mana Alavioun
- Department of Basic sciences, Faculty of Veterinary Medicine, Urmia university, Urmia, Iran
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA
| | | | - Tooba Yousefi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran.
| | - Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Luo J, Sun T, Jiang S, Yang Z, Xiao C, Deng J, Zhou B, Yang X. Comprehensive analysis of non-coding RNAs in the ovaries of high and low egg production hens. Anim Reprod Sci 2025; 276:107836. [PMID: 40220592 DOI: 10.1016/j.anireprosci.2025.107836] [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: 12/11/2024] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Egg production performance a critical economic trait in the poultry industry. The regulatory mechanisms underlying egg production performance mediated by non-coding RNAs remain to be characterized. To systematically investigate ovarian lncRNAs, circRNAs, and miRNAs associated with laying efficiency, we conducted comparative transcriptomic analyses using RNA sequencing (RNA-seq) of ovarian tissues from phenotypically divergent groups - high egg production (HEP) and low egg production (LEP) hens. In our study, we identified 675 lncRNAs, 140 circRNAs, and 10 miRNAs that were significantly differentially expressed (DE) between HEP and LEP. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that target genes of DE lncRNAs, DE miRNAs, and the source genes of DE circRNAs are involved in the MAPK signaling pathway, endocytosis, notch signaling pathway, among others. Furthermore, we identified five miRNA-mRNA interactions related to egg production including gga-miR-449c-3p, and five genes (GLI2, TAC1, EML6, THOC3, MMP9). These findings establish the first comprehensive ncRNA interactome driving ovarian efficiency, offering both biomarkers for breeding selection and mechanistic targets for reproductive enhancement.
Collapse
Affiliation(s)
- Jintang Luo
- From the College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Tiantian Sun
- From the College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Siyi Jiang
- From the Beijing Royal School, Beijing 102209, China
| | - Zhuliang Yang
- From the College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Cong Xiao
- From the College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Jixian Deng
- From the College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Biyan Zhou
- From the College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Xiurong Yang
- From the College of Animal Science and Technology, Guangxi University, Nanning 530004, China; From the Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Naning 530004, China.
| |
Collapse
|
9
|
Feng H, Nie Q, Yang S. SORFPP: Enhancing rich sequence-driven information to identify SEPs based on fused framework on validation datasets. PLoS One 2025; 20:e0320314. [PMID: 40294059 PMCID: PMC12036913 DOI: 10.1371/journal.pone.0320314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 02/17/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Genome sequencing has enabled us to find functional peptides encoded by short open read frames (sORFs) in long non-coding RNAs (lncRNAs). sORFs-encoded peptides (SEPs) regulate gene expression, signaling, and so on and have significant roles, unlike common peptides. Various computational methods have been proposed. However, there is a lack of contributive features and effective models. Therefore, a high-throughput computational method to predict SEPs is needed. RESULTS We propose a computational method, SORFPP, to predict SEPs by mining feature information from multiple perspectives in an experimentally validated dataset from TranLnc. SORFPP fully extracts SEP sequence information using the protein language model ESM-2 and curated traditional encoding, including QSOrder, k-mer, etc. SORFPP uses CatBoost to solve the sparsity problem of traditional encoding. SORFPP also analyzes ESM-2 pre-training characterization information with the Self-attention model. Finally, an ensemble learning framework combines the two models and their results are fed into Logistic Regression model for accurate and robust predictions. For comparison, SORFPP outperforms other state-of-the-art models in Matthew correlation coefficient by 12.2%-24.2% on three benchmark datasets. CONCLUSION Integrating the ensemble learning strategy with contributive traditional features and the protein language encoding methods shows better performance. Datasets and codes are accessible at https://doi.org/10.6084/m9.figshare.28079897 and http://111.229.198.94:5000/.
Collapse
Affiliation(s)
- Hongqi Feng
- School of Computer Science and Artificial Intelligence Aliyun School of Big Data School of Software, Changzhou University, Changzhou, China
| | - Qi Nie
- School of Computer Science and Artificial Intelligence Aliyun School of Big Data School of Software, Changzhou University, Changzhou, China
| | - Sen Yang
- School of Computer Science and Artificial Intelligence Aliyun School of Big Data School of Software, Changzhou University, Changzhou, China
- The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
10
|
Wang Y, Wang B, Zou J, Wu A, Liu Y, Wan Y, Luo J, Wu J. Capsule neural network and its applications in drug discovery. iScience 2025; 28:112217. [PMID: 40241764 PMCID: PMC12002614 DOI: 10.1016/j.isci.2025.112217] [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] [Indexed: 04/18/2025] Open
Abstract
Deep learning holds great promise in drug discovery, yet its application is hindered by high labeling costs and limited datasets. Developing algorithms that effectively learn from sparsely labeled data is crucial. Capsule networks (CapsNet), introduced in 2017, solve the spatial information loss in traditional neural networks and excel in handling small datasets by capturing spatial hierarchical relationships among features. This capability makes CapsNet particularly promising for drug discovery, where data scarcity is a common challenge. Various modified CapsNet architectures have been successfully applied to drug design and discovery tasks. This review provides a comprehensive analysis of CapsNet's theoretical foundations, its current applications in drug discovery, and its performance in addressing key challenges in the field. Additionally, the study highlights the limitations of CapsNet and outlines potential future research directions to further enhance its utility in drug discovery, offering valuable insights for researchers in both computational and pharmaceutical sciences.
Collapse
Affiliation(s)
- Yiwei Wang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Binyou Wang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Jun Zou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Yuan Liu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Ying Wan
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Jiesi Luo
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Jianming Wu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
11
|
Wang D, Gao Y, Tan Y, Li N, Li X, Li J, Pan Y, Zhao X, Yan M, Wang Y. lncRNA Ubr5 promotes BMSCs apoptosis and inhibits their proliferation and osteogenic differentiation in weightless bone loss. Front Cell Dev Biol 2025; 13:1543929. [PMID: 40241795 PMCID: PMC11999945 DOI: 10.3389/fcell.2025.1543929] [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: 12/12/2024] [Accepted: 03/10/2025] [Indexed: 04/18/2025] Open
Abstract
Background Weightless bone loss is a common pathological phenomenon in weightless environments, yet its specific molecular mechanism remain incompletely elucidated. The aim of this study was to systematically investigate the differential expression profiles of mRNAs and long noncoding RNAs (lncRNAs) to explore the molecular pathogenesis underlying weightless bone loss. Methods Transcriptome sequencing was performed on bone marrow mesenchymal stem cell (BMSCs) samples from the Ground control group and simulated microgravity (SMG) group using Illumina technology. Using the DESeq2 algorithm, we accurately identify and analyzed the differentially expressed genes (DEGs). Subsequently, the molecular functions and signaling pathways enriched by DEG were comprehensively analyzed by GO and KEGG. In addition, by constructing lncRNA-mRNA coexpression network, this study screened and verified key lncRNAs as potential genes to further explore their role in the occurrence and development of weightless bone loss. Results A total of 215 differentially expressed lncRNAs (DElncRNAs) and 381 differentially expressed mRNAs (DEmRNAs) were identified, in the SMG group. DEmRNAs were primarily involved in the cell response to mechanical stimulation, microtubule motility and TNF signaling pathway. Meanwhile, DElncRNAs are significantly enriched in cell differentiation, fatty acid metabolic process and biosynthesis of amino acids. In addition, the expression levels of related lncRNAs and mRNAs in weightless bone loss were verified via qRT-PCR. lncRNA-mRNA coexpression network found that lncRNA Ubr5 closely related to osteoblast proliferation and differentiation. Further experimental results revealed that knocking down lncRNA Ubr5 can promote the apoptosis of BMSCs and inhibit their proliferation and osteogenic differentiation. Conclusion This study revealed the molecular pathogenesis of weightless bone loss, identified lncRNA Ubr5 as a potential intervention target, and provided an important scientific basis and strategic guidance for the prevention and treatment of weightless bone loss.
Collapse
Affiliation(s)
- Dong Wang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Air Force Medical University, Xi’an, China
| | - Yuan Gao
- Department of Aerospace Medical Training, School of Aerospace Medicine, Air Force Medical University, Xi’an, China
| | - Yingjun Tan
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Na Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Air Force Medical University, Xi’an, China
| | - Xi Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Air Force Medical University, Xi’an, China
| | - Jiaxiang Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Air Force Medical University, Xi’an, China
| | - Yikai Pan
- Department of Aerospace Medical Training, School of Aerospace Medicine, Air Force Medical University, Xi’an, China
| | - Xingcheng Zhao
- Department of Aerospace Medical Training, School of Aerospace Medicine, Air Force Medical University, Xi’an, China
| | - Ming Yan
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yongchun Wang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Air Force Medical University, Xi’an, China
| |
Collapse
|
12
|
Solaimani M, Hosseinzadeh S, Abasi M. Non-coding RNAs, a double-edged sword in breast cancer prognosis. Cancer Cell Int 2025; 25:123. [PMID: 40170036 PMCID: PMC11959806 DOI: 10.1186/s12935-025-03679-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 02/06/2025] [Indexed: 04/03/2025] Open
Abstract
Cancer is a rising issue worldwide, and numerous studies have focused on understanding the underlying reasons for its occurrence and finding proper ways to defeat it. By applying technological advances, researchers are continuously uncovering and updating treatments in cancer therapy. Their vast functions in the regulation of cell growth and proliferation and their significant role in the progression of diseases, including cancer. This review provides a comprehensive analysis of ncRNAs in breast cancer, focusing on long non-coding RNAs such as HOTAIR, MALAT1, and NEAT1, as well as microRNAs such as miR-21, miR-221/222, and miR-155. These ncRNAs are pivotal in regulating cell proliferation, metastasis, drug resistance, and apoptosis. Additionally, we discuss experimental approaches that are useful for studying them and highlight the advantages and challenges of each method. We then explain the results of these clinical trials and offer insights for future studies by discussing major existing gaps. On the basis of an extensive number of studies, this review provides valuable insights into the potential of ncRNAs in cancer therapy. Key findings show that even though the functions of ncRNAs are vast and undeniable in cancer, there are still complications associated with their therapeutic use. Moreover, there is an absence of sufficient experiments regarding their application in mouse models, which is an area to work on. By emphasizing the crucial role of ncRNAs, this review underscores the need for innovative approaches and further studies to explore their potential in cancer therapy.
Collapse
Affiliation(s)
- Maryam Solaimani
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Sahar Hosseinzadeh
- Faculty of Pharmacy and Medical Biotechnology, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mozhgan Abasi
- Immunogenetics Research Center, Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, PO Box: 48175/861, Sari, Iran.
| |
Collapse
|
13
|
Zhang Y, Zang C, Mao M, Zhang M, Tang Z, Chen W, Zhu W. Advances in RNA therapy for the treatment of autoimmune diseases. Autoimmun Rev 2025; 24:103753. [PMID: 39842534 DOI: 10.1016/j.autrev.2025.103753] [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: 01/15/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
Autoimmune diseases (ADs) are a group of complex, chronic conditions characterized by disturbance of immune tolerance, with examples including systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, and psoriasis. These diseases have unclear pathogenesis, and traditional therapeutic approaches remain limited. However, advances in high-throughput histology technology and scientific discoveries have led to the identification of various pathogenic factors contributing to ADs. Coupled with improvements in RNA nucleic acid-based drug synthesis, design, and delivery, RNA-based therapies have been extensively investigated for their potential in treating ADs. This paper reviews the progress in the use of miRNAs, lncRNAs, circRNAs, siRNAs, antisense oligonucleotides (ASOs), aptamers, mRNAs, and other RNA-based therapies in ADs, focusing on their therapeutic potential and application prospects, providing insights for future research and clinical treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Ying Zhang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Chenyang Zang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Manyun Mao
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Mi Zhang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Zhenwei Tang
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wangqing Chen
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China.
| | - Wu Zhu
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China.
| |
Collapse
|
14
|
Zhang S, Guo J, He Y, Su Z, Feng Y, Zhang L, Jun Z, Weng X, Yuan Y. Roles of lncRNA in the crosstalk between osteogenesis and angiogenesis in the bone microenvironment. J Zhejiang Univ Sci B 2025; 26:107-123. [PMID: 40015932 PMCID: PMC11867785 DOI: 10.1631/jzus.b2300607] [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: 08/29/2023] [Accepted: 01/16/2024] [Indexed: 03/01/2025]
Abstract
Bone is a highly calcified and vascularized tissue. The vascular system plays a vital role in supporting bone growth and repair, such as the provision of nutrients, growth factors, and metabolic waste transfer. Moreover, the additional functions of the bone vasculature, such as the secretion of various factors and the regulation of bone-related signaling pathways, are essential for maintaining bone health. In the bone microenvironment, bone tissue cells play a critical role in regulating angiogenesis, including osteoblasts, bone marrow mesenchymal stem cells (BMSCs), and osteoclasts. Osteogenesis and bone angiogenesis are closely linked. The decrease in osteogenesis and bone angiogenesis caused by aging leads to osteoporosis. Long noncoding RNAs (lncRNAs) are involved in various physiological processes, including osteogenesis and angiogenesis. Recent studies have shown that lncRNAs could mediate the crosstalk between angiogenesis and osteogenesis. However, the mechanism by which lncRNAs regulate angiogenesis‒osteogenesis crosstalk remains unclear. In this review, we describe in detail the ways in which lncRNAs regulate the crosstalk between osteogenesis and angiogenesis to promote bone health, aiming to provide new directions for the study of the mechanism by which lncRNAs regulate bone metabolism.
Collapse
Affiliation(s)
- Shihua Zhang
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China
- College of Sports and Health, Shandong Sport University, Jinan 250102, China
| | - Jianmin Guo
- School of Life Sciences, South University of Science and Technology, Shenzhen 518055, China
| | - Yuting He
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China
| | - Zhi'ang Su
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China
| | - Yao Feng
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China
| | - Lan Zhang
- College of Sports and Health, Shandong Sport University, Jinan 250102, China
| | - Zou Jun
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Xiquan Weng
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China. ,
| | - Yu Yuan
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China.
| |
Collapse
|
15
|
Gao F, Wang F, Chen Y, Deng B, Yang F, Cao H, Chen J, Chen H, Qi F, Kapranov P. The human genome encodes a multitude of novel miRNAs. Nucleic Acids Res 2025; 53:gkaf070. [PMID: 39964476 PMCID: PMC11833695 DOI: 10.1093/nar/gkaf070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/21/2025] Open
Abstract
Human cells generate a vast complexity of noncoding RNAs, the "RNA dark matter," which includes a vast small RNA (sRNA) transcriptome. The biogenesis, biological relevance, and mechanisms of action of most of these transcripts remain unknown, and they are widely assumed to represent degradation products. Here, we aimed to functionally characterize human sRNA transcriptome by attempting to answer the following question-can a significant number of novel sRNAs correspond to novel members of known classes, specifically, microRNAs (miRNAs)? By developing and validating a miRNA discovery pipeline, we show that at least 2726 novel canonical miRNAs, majority of which represent novel miRNA families, exist in just one human cell line compared to just 1914 known miRNA loci. Moreover, potentially tens of thousands of miRNAs remain to be discovered. Strikingly, many novel miRNAs map to exons of protein-coding genes emphasizing a complex and interleaved architecture of the genome. The existence of so many novel members of a functional class of sRNAs suggest that the human sRNA transcriptome harbors a multitude of novel regulatory molecules. Overall, these results suggest that we are at the very beginning of understanding the true functional complexity of the sRNA component of the "RNA dark matter."
Collapse
Affiliation(s)
- Fan Gao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
- Xiamen Institute for Food and Drug Quality Control, 33 Haishan Road, Xiamen 361012, China
| | - Fang Wang
- Institute of Genomics, School of Medicine, Huaqiao University, 668 Jimei Road, Xiamen 361021, China
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Yue Chen
- Institute of Genomics, School of Medicine, Huaqiao University, 668 Jimei Road, Xiamen 361021, China
| | - Bolin Deng
- Institute of Genomics, School of Medicine, Huaqiao University, 668 Jimei Road, Xiamen 361021, China
| | - Fujian Yang
- Institute of Genomics, School of Medicine, Huaqiao University, 668 Jimei Road, Xiamen 361021, China
| | - Huifen Cao
- Institute of Genomics, School of Medicine, Huaqiao University, 668 Jimei Road, Xiamen 361021, China
| | - Junjie Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Huiling Chen
- Xiamen Institute for Food and Drug Quality Control, 33 Haishan Road, Xiamen 361012, China
| | - Fei Qi
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Philipp Kapranov
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| |
Collapse
|
16
|
Balaraman AK, Afzal M, Moglad E, Babu MA, Priya GP, Bansal P, Rajotiya S, Kondapavuluri BK, Kazmi I, Alzarea SI, Goyal K, Ali H. The interplay of p16INK4a and non-coding RNAs: bridging cellular senescence, aging, and cancer. Biogerontology 2025; 26:50. [PMID: 39907830 DOI: 10.1007/s10522-025-10194-2] [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: 12/24/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
p16INK4a is a crucial tumor suppressor and regulator of cellular senescence, forming a molecular bridge between aging and cancer. Dysregulated p16INK4a expression is linked to both premature aging and cancer progression, where non-coding RNAs (ncRNAs) such as long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and small interfering RNAs (siRNAs) play key roles in modulating its function. These ncRNAs interact with p16INK4a through complex post-transcriptional and epigenetic mechanisms, influencing pathways critical to senescence and tumor suppression. In this review, we explore ncRNAs, including ANRIL, MIR31HG, UCA1, MALAT1, miR-24, miR-30, and miR-141, which collectively regulate p16INK4a expression, promoting or inhibiting pathways associated with cancer and aging. ANRIL and MIR31HG modulate p16INK4a silencing via interactions with polycomb repressive complexes (PRC), while miRNAs such as miR-24 and miR-30 target p16INK4a to influence cellular proliferation and senescence. This regulatory interplay underscores the therapeutic potential of ncRNA-targeted strategies to restore p16INK4a function. We summarize recent studies supporting that ncRNAs that control p16INK4a may be diagnostic biomarkers and therapeutic targets for age-related diseases and cancer.
Collapse
Affiliation(s)
- Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, 63000, Cyberjaya, Selangor, Malaysia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al Kharj, Saudi Arabia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - G Padma Priya
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Pooja Bansal
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Sumit Rajotiya
- NIMS Institute of Pharmacy, NIMS University, Jaipur, Rajasthan, India
| | - Benod Kumar Kondapavuluri
- Department of General Surgery, Consultant Head and Neck Surgical Oncology, Dr.D.Y.Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Al-Jouf, Saudi Arabia
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India.
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| |
Collapse
|
17
|
John A, Almulla N, Elboughdiri N, Gacem A, Yadav KK, Abass AM, Alam MW, Wani AW, Bashir SM, Rab SO, Kumar A, Wani AK. Non-coding RNAs in Cancer: Mechanistic insights and therapeutic implications. Pathol Res Pract 2025; 266:155745. [PMID: 39637712 DOI: 10.1016/j.prp.2024.155745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/23/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Non-coding RNAs have gathered significant attention for their unique roles in biological regulation. Across a broad spectrum of developmental processes and diseases, particularly in human malignancies, ncRNAs play pivotal roles in regulatory mechanisms. MicroRNAs, long noncoding RNAs, and small nucleolar RNAs stand out among the diverse forms of ncRNAs that have been implicated in cancer. MiRNAs, classified as short non-coding RNAs, modulate gene expression by binding to messenger RNA molecules, thereby inhibiting their translation. Altered miRNA expression has been associated with the onset and progression of various malignancies, including lung, breast, and prostate cancer. In contrast, lncRNAs, characterized as longer ncRNAs, exert control over gene expression through various mechanisms, such as chromatin remodelling and gene silencing. This review offers a comprehensive examination of the numerous ncRNAs that have emerged as crucial regulators of gene expression, playing implicated roles in the initiation and progression of diverse cancers.
Collapse
Affiliation(s)
- Arjumand John
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar, Punjab 144411, India
| | - Nuha Almulla
- Department of Biology, Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Noureddine Elboughdiri
- Chemical Engineering Department, College of Engineering, University of Ha'il, P.O. Box 2440, Ha'il 81441, Saudi Arabia
| | - Amel Gacem
- Department of Physics, Faculty of Sciences, University 20 Aout, Skikda 1955, Algeria
| | - Krishna Kumar Yadav
- Department of VLSI Microelectronics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai - 602105, Tamil Nadu, India; Environmental and Atmospheric Sciences Research Group, Scientific Research Center, Al-Ayen University, Thi-Qar, Nasiriyah 64001, Iraq
| | - Anass M Abass
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| | - Mir Waqas Alam
- Department of Physics, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia.
| | - Ab Waheed Wani
- Department of Horticulture, School of Agriculture, Lovely Professional University, Jalandhar, Punjab 144411, India
| | - Showkeen Muzamil Bashir
- Biochemistry & Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, Jammu and Kashmir 190006, India
| | - Safia Obaidur Rab
- Central Labs, King Khalid University, AlQura'a, P.O. Box 960, Abha, Saudi Arabia; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named after the First President of Russia Boris Yeltsin, Ekaterinburg 620002, Russia; Department of Technical Sciences, Western Caspian University, Baku, Azerbaijan; Department of Mechanical Engineering, Karpagam Academy of Higher Education, Coimbatore 641021, India
| | - Atif Khurshid Wani
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar, Punjab 144411, India.
| |
Collapse
|
18
|
Fan Y, Tian D, Lv Z, Peng S, Zhu S. LncRNA-THBS4 affects granulosa cell proliferation and apoptosis in diminished ovarian reserve by regulating PI3K/AKT/mTOR signaling pathway. J Reprod Immunol 2025; 167:104419. [PMID: 39732055 DOI: 10.1016/j.jri.2024.104419] [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: 11/08/2024] [Revised: 11/27/2024] [Accepted: 12/13/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUNDS Recent studies have found Several lncRNAs were proved differential expression in diminished ovarian reserve (DOR) patients, however, the mechanism of DOR caused by lncRNAs is still largely unclear. METHODS High throughput sequencing was performed in ovarian GCs extracted from women with normal ovarian function and women with DOR. Bioinformation analysis was used to analyze the sequencing data and identify the differential expression of lncRNAs. Quantitative RT-PCR (qRT-PCR) was used to verify the sequencing results. Situ fluorescence hybridization (FISH) followed by confocal microscopy and qRT-PCR were used to explore the location and expression of LncRNA-THBS4 in GCs. The significantly enriched signaling pathways of LncRNA-THBS4 were identified by KEGG. The study used RNA interference technology to decipher LncRNA-THBS4 function by silencing LncRNA-THBS4 in GCs. Western blot and qRT-PCR were used to explore the mRNA and protein expressions of key factors of PI3Ks pathway. The pro-apoptotic protein and anti-apoptotic protein were detected by western blot. The proliferation and apoptosis of GCs were detected by MTT assay and Flow cytometry. RESULTS 197 lncRNAs with significant differences in expression levels were detected between control and DOR group by high throughput sequencing. The study found the expression of LncRNA-THBS4 in GCs was positively correlated with Anti-Mullerian hormone (AMH) (p = 0.0020, r = 0.4742)、antral follicle count (AFC) (p = 0.0007, r = 0.5130)、good embryo rate (p = 0.0006, r = 0.5210), negatively correlated with basal FSH level (p = 0.0007, r = -0.5152). LncRNA-THBS4 was mainly localized in the cytoplasm of GCs. LncRNA-THBS4 silencing could inhibit the PI3Ks pathway; decrease the levels of anti-apoptotic protein, inhibit the proliferation of GCs; increase the levels of apoptosis protein, enhance the apoptosis of GCs. CONCLUSIONS The expression level of lncRNA-THBS4 is correlated with ovarian function indicators and pregnancy outcomes in women. LncRNA-THBS4 may participate in the pathogenesis of DOR by affecting the proliferation and apoptosis of GCs via regulating PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yiyue Fan
- School of Medical and Life Sciences/Reproductive & Women-Childrer Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Beiiing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, Nanchong, China.
| | - Dongmei Tian
- School of Medical and Life Sciences/Reproductive & Women-Childrer Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Zili Lv
- School of Medical and Life Sciences/Reproductive & Women-Childrer Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Shiyang Peng
- School of Medical and Life Sciences/Reproductive & Women-Childrer Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Shaomi Zhu
- Chengdu Fifth People's Hospital, (School of Medical and Life Sciences/Affiliated Fifth People's Hospital, Chengdu University of Traditional Chinese Medicine), Chengdu, China.
| |
Collapse
|
19
|
Shen Z, Tao L, Wang Y, Zhu Y, Pan H, Li Y, Jiang S, Zheng J, Cai J, Liu Y, Lin K, Li S, Tong Y, Shangguan L, Xu J, Liang X. Synergistic Anticancer Strategy Targeting ECM Stiffness: Integration of Matrix Softening and Mechanical Signal Transduction Blockade in Primary Liver Cancers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2403040. [PMID: 39703167 PMCID: PMC11809367 DOI: 10.1002/advs.202403040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 10/30/2024] [Indexed: 12/21/2024]
Abstract
The development of primary liver cancer (hepatocellular carcinoma [HCC] and intrahepatic cholangiocarcinoma [ICC]) is linked to its physical microenvironment, particularly extracellular matrix (ECM) stiffness. Potential anticancer strategies targeting ECM stiffness include prevention/reversal of the stiffening process and disruption of the response of cancer cells to mechanical signals from ECM. However, each strategy has limitations. Therefore, the authors propose integrating them to maximize their strengths. Compared with HCC, ICC has a stiffer ECM and a worse prognosis. Therefore, ICC is selected to investigate mechanisms underlying the influence of ECM stiffness on cancer progression and application of the integrated anticancer strategy targeting ECM stiffness. In summary, immunofluorescence results for 181 primary liver cancer tissue chips (ICC, n = 91; HCC, n = 90) and analysis of TCGA mRNA-sequencing demonstrate that ECM stiffness can affect phenotypes of primary liver cancers. The YAP1/ABHD11-AS1/STAU2/ZYX/p-YAP1 pathway is a useful entry point for exploration of specific mechanisms of mechanical signal conduction from the ECM in ICC cells and their impact on cancer progression. Moreover, a synergistic anticancer strategy targeting ECM stiffness (ICCM@NPs + siABHD11-AS1@BAPN) is constructed by integrating ECM softening and blocking intracellular mechanical signal transduction in ICC and can provide insights for the treatment of cancers characterized by stiff ECM.
Collapse
|
20
|
Du T, Meng D, Cao H, Lian Y, Wu R, Liu T, Wang T, Qin C, Song Z, Dong B, Fu Y, Yang Q. Sorbitol induces flavonoid accumulation as a secondary signal via the nanoencapsulated SPc/lncRNA809-MmNAC17 module against Alternaria alternata in Malus micromalus. MOLECULAR HORTICULTURE 2025; 5:5. [PMID: 39885599 PMCID: PMC11783756 DOI: 10.1186/s43897-024-00125-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/29/2024] [Indexed: 02/01/2025]
Abstract
Sorbitol is an important primary metabolite that serves as both a carbon source and signal to pathogens. The leaf diseases caused by Alternata alternata are particularly serious in crabapple (Malus micromalus). Here, we found that sorbitol can enhance the resistance of crabapple to A. alternata R1 by increasing the content of flavonoid catechin. Nanomaterials as an emerging technology tool can efficiently deliver lncRNA to target cells. Here, we found nanoencapsulated lncRNA809 (SPc/lncRNA809) exhibits significant resistance to R1strain. To elucidate the effect of SPc/lncRNA809 on flavonoids catechin synthesis, we observed the expression of lncRNA809 was consistent with that of MmNAC17 which regulates the synthesis of catechin and both could jointly respond to sorbitol. MmNAC17 induced the accumulation of catechin in vivo by directly activating the expression of catechin synthase genes MmF3H and MmLAR. Correspondingly, overexpression of lncRNA809 significantly upregulated the expression of MmNAC17 and enhanced the disease resistance. This study reveals for the first time that sorbitol positively regulates the expression of MmNAC17 through lncRNA809, promoting the accumulation of catechin via the expression of MmF3H and MmLAR, ultimately improving the defense response of M. micromalus. This research provides a crucial foundation for the establishment and application of sorbitol-based signaling regulatory networks.
Collapse
Affiliation(s)
- Tingting Du
- Beijing Forestry University, Beijing, 100000, China
| | - Dong Meng
- Beijing Forestry University, Beijing, 100000, China
| | - Hongyan Cao
- Beijing Forestry University, Beijing, 100000, China
| | - Yi Lian
- Beijing Forestry University, Beijing, 100000, China
| | - Rui Wu
- Beijing Forestry University, Beijing, 100000, China
| | - Tengyue Liu
- Beijing Forestry University, Beijing, 100000, China
| | - Tianyi Wang
- Beijing Forestry University, Beijing, 100000, China
| | - Cai Qin
- Beijing Forestry University, Beijing, 100000, China
| | - Zhihua Song
- Beijing Forestry University, Beijing, 100000, China
| | - Biying Dong
- Beijing Forestry University, Beijing, 100000, China
| | - Yujie Fu
- Beijing Forestry University, Beijing, 100000, China
| | - Qing Yang
- Beijing Forestry University, Beijing, 100000, China.
| |
Collapse
|
21
|
Zhang Y, Yang Y, Li K, Chen L, Yang Y, Yang C, Xie Z, Wang H, Zhao Q. Enhanced Discovery of Alternative Proteins (AltProts) in Mouse Cardiac Development Using Data-Independent Acquisition (DIA) Proteomics. Anal Chem 2025; 97:1517-1527. [PMID: 39813267 PMCID: PMC11781309 DOI: 10.1021/acs.analchem.4c02924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 01/18/2025]
Abstract
Alternative proteins (AltProts) are a class of proteins encoded by DNA sequences previously classified as noncoding. Despite their historically being overlooked, recent studies have highlighted their widespread presence and distinctive biological roles. So far, direct detection of AltProt has been relying on data-dependent acquisition (DDA) mass spectrometry (MS). However, data-independent acquisition (DIA) MS, a method that is rapidly gaining popularity for the analysis of canonical proteins, has seen limited application in AltProt research, largely due to the complexities involved in constructing DIA libraries. In this study, we present a novel DIA workflow that leverages a fragmentation spectra predictor for the efficient construction of DIA libraries, significantly enhancing the detection of AltProts. Our method achieved a 2-fold increase in the identification of AltProts and a 50% reduction in missing values compared to DDA. We conducted a comprehensive comparison of four AltProt databases, four DIA-library construction strategies, and three analytical software tools to establish an optimal workflow for AltProt analysis. Utilizing this workflow, we investigated the mouse heart development process and identified over 50 AltProts with differential expression between embryonic and adult heart tissues. Over 30 unannotated mouse AltProts were validated, including ASDURF, which played a crucial role in cardiac development. Our findings not only provide a practical workflow for MS-based AltProt analysis but also reveal novel AltProts with potential significance in biological functions.
Collapse
Affiliation(s)
- Yuanliang Zhang
- Department
of Applied Biology and Chemical Technology, State Key Laboratory of
Chemical Biology and Drug Discovery, Hong
Kong Polytechnic University, Hong Kong 999077, China
| | - Ying Yang
- Department
of Applied Biology and Chemical Technology, State Key Laboratory of
Chemical Biology and Drug Discovery, Hong
Kong Polytechnic University, Hong Kong 999077, China
| | - Kecheng Li
- Department
of Applied Biology and Chemical Technology, State Key Laboratory of
Chemical Biology and Drug Discovery, Hong
Kong Polytechnic University, Hong Kong 999077, China
| | - Lei Chen
- Department
of Applied Biology and Chemical Technology, State Key Laboratory of
Chemical Biology and Drug Discovery, Hong
Kong Polytechnic University, Hong Kong 999077, China
| | - Yang Yang
- Department
of Applied Biology and Chemical Technology, State Key Laboratory of
Chemical Biology and Drug Discovery, Hong
Kong Polytechnic University, Hong Kong 999077, China
| | - Chenxi Yang
- Department
of Applied Biology and Chemical Technology, State Key Laboratory of
Chemical Biology and Drug Discovery, Hong
Kong Polytechnic University, Hong Kong 999077, China
| | - Zhi Xie
- State
Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Hongwei Wang
- State
Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Qian Zhao
- Department
of Applied Biology and Chemical Technology, State Key Laboratory of
Chemical Biology and Drug Discovery, Hong
Kong Polytechnic University, Hong Kong 999077, China
| |
Collapse
|
22
|
Ng B, Avey DR, Lopes KDP, Fujita M, Vialle RA, Vyas H, Kearns NA, Tasaki S, Iatrou A, Tissera SD, Chang TH, Xu J, Yu C, Sultan F, Menon V, Gaiteri C, De Jager PL, Bennett DA, Wang Y. Spatial Expression of Long Non-Coding RNAs in Human Brains of Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.27.620550. [PMID: 39554066 PMCID: PMC11565709 DOI: 10.1101/2024.10.27.620550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Long non-coding RNAs (lncRNAs) are critical regulators of physiological and pathological processes, with their dysregulation increasingly implicated in aging and Alzheimer's disease (AD). Using spatial transcriptomics, we analyzed 78 postmortem brain sections from 21 ROSMAP participants to map the spatial expression of lncRNAs in the dorsolateral prefrontal cortex of aged human brains. Compared to mRNAs, lncRNAs exhibited greater subregion-specific expression, with enrichment in antisense and lincRNA biotypes. Network analysis identified 193 gene modules across eight subregions, including lncRNA-enriched modules involved in critical biological processes. We also identified AD differentially expressed (DE) lncRNAs, which showed greater subregion specificity than AD DE mRNAs. Gene set enrichment analysis highlighted the involvement of these AD DE lncRNAs in epigenetic regulation and chromatin remodeling, including enrichment for HDAC target genes such as OIP5-AS1. Statistical modeling suggested that interactions between OIP5-AS1 and HDAC proteins, particularly HDAC11, were associated with tau tangles in excitatory neurons and plaque burden in microglia. This study provides a comprehensive resource of lncRNA spatial expression in the aged human brain and uncovers potential functional roles of lncRNAs in AD pathogenesis.
Collapse
|
23
|
Sinha T, Sadhukhan S, Panda AC. Computational Prediction of Gene Regulation by lncRNAs. Methods Mol Biol 2025; 2883:343-362. [PMID: 39702716 DOI: 10.1007/978-1-0716-4290-0_15] [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] [Indexed: 12/21/2024]
Abstract
High-throughput sequencing technologies and innovative bioinformatics tools discovered that most of the genome is transcribed into RNA. However, only a fraction of the RNAs in cell translates into proteins, while the majority of them are categorized as noncoding RNAs (ncRNAs). The ncRNAs with more than 200 nt without protein-coding ability are termed long noncoding RNAs (lncRNAs). Hundreds of studies established that lncRNAs are a crucial RNA family regulating gene expression. Regulatory RNAs, including lncRNAs, modulate gene expression by interacting with RNA, DNA, and proteins. Several databases and computational tools have been developed to explore the functions of lncRNAs in cellular physiology. This chapter discusses the tools available for lncRNA functional analysis and provides a detailed workflow for the computational analysis of lncRNAs.
Collapse
Affiliation(s)
- Tanvi Sinha
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, India
| | - Susovan Sadhukhan
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, India
| | - Amaresh C Panda
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, India.
| |
Collapse
|
24
|
Janga H, Schmerer N, Aznaourova M, Schulte LN. Non-coding RNA Networks in Infection. Methods Mol Biol 2025; 2883:53-77. [PMID: 39702704 DOI: 10.1007/978-1-0716-4290-0_3] [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] [Indexed: 12/21/2024]
Abstract
In the face of global health challenges posed by infectious diseases and the emergence of drug-resistant pathogens, the exploration of cellular non-coding RNA (ncRNA) networks has unveiled new dimensions in infection research. Particularly microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) have emerged as instrumental players in ensuring a balance between protection against hyper-inflammatory conditions and the effective elimination of pathogens. Specifically, ncRNAs, such as the miRNA miR-155 or the lncRNAs MaIL1 (macrophage interferon-regulatory lncRNA 1), and LUCAT1 (lung cancer-associated transcript 1) have been recurrently linked to infectious and inflammatory diseases. Together with other ncRNAs, discussed in this chapter, they form a complex regulatory network shaping the host response to pathogens. Additionally, some pathogens exploit these ncRNAs to establish and sustain infections, underscoring their dual roles in host protection and colonization. Despite the substantial progress made, the vast majority of ncRNA loci remains unexplored, with ongoing research likely to reveal novel ncRNA categories and expand our understanding of their roles in infections. This chapter consolidates current insights into ncRNA-mediated regulatory networks, highlighting their contributions to severe diseases and their potential as targets and biomarkers for innovative therapeutic strategies.
Collapse
Affiliation(s)
| | - Nils Schmerer
- Institute for Lung Research, Philipps University, Marburg, Germany
| | | | - Leon N Schulte
- Institute for Lung Research, Philipps University, Marburg, Germany.
- German Center for Lung Research, Giessen, Germany.
| |
Collapse
|
25
|
Shaikh M, Doshi G. Unraveling non-coding RNAs in breast cancer: mechanistic insights and therapeutic potential. Med Oncol 2024; 42:37. [PMID: 39730979 DOI: 10.1007/s12032-024-02589-x] [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: 09/29/2024] [Accepted: 12/16/2024] [Indexed: 12/29/2024]
Abstract
Breast cancer remains a leading global health challenge requiring innovative, therapeutic strategies to improve patient outcomes. This review explores the pivotal roles of non-coding RNAs (ncRNAs), including long non-coding RNA, micro RNA, and circular RNA, in breast cancer biology. We highlight how these molecules regulate critical signaling pathways, influence tumor microenvironments, and contribute to treatment resistance. Our findings underscore the potential of ncRNAs as biomarkers for early diagnosis and as treatment targets for personalized treatment strategies. To pave the way for innovative cancer management approaches, we investigate the complex interactions of ncRNAs and their impact on tumor progression. This comprehensive review enhances our understanding of breast cancer biology while emphasizing the translational significance of ncRNA research in developing effective treatment strategies. Additional research and clinical studies are required to confirm the diagnostic and medicinal value of ncRNAs in breast cancer. Investigating the complex networks of ncRNA interactions and their links to other biological pathways can lead to the discovery of new treatment targets. Furthermore, leveraging advanced technologies, such as machine learning and multi-omics methods, will be critical in improving our understanding of ncRNAs biomarkers and translating these insights into impactful clinical applications.
Collapse
Affiliation(s)
- Muqtada Shaikh
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, 400 056, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, 400 056, India.
| |
Collapse
|
26
|
Singh G, Darwin R, Panda KC, Afzal SA, Katiyar S, Dhakar RC, Mani S. Gene expression and hormonal signaling in osteoporosis: from molecular mechanisms to clinical breakthroughs. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024:1-36. [PMID: 39729311 DOI: 10.1080/09205063.2024.2445376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Osteoporosis is well noted to be a universal ailment that realization impaired bone mass and micro architectural deterioration thus enhancing the probability of fracture. Despite its high incidence, its management remains highly demanding because of the multifactorial pathophysiology of the disease. This review highlights recent findings in the management of osteoporosis particularly, gene expression and hormonal control. Some of the newest approaches regarding the subject are described, including single-cell RNA sequencing and long non-coding RNAs. Also, the review reflects new findings on hormonal signaling and estrogen and parathyroid hormone; patient-specific approaches due to genetic and hormonal variation. Potential new biomarkers and AI comprised as factors for improving the ability to anticipate and manage fractures. These hold great potential of new drugs, combination therapies and gene based therapies for osteoporosis in the future. Further studies and cooperation of scientists and clinicians will help to apply such novelties into practical uses in the sphere of medicine in order to enhance the treatment of patients with osteoporosis.
Collapse
Affiliation(s)
- Gurinderdeep Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala, India
| | - Ronald Darwin
- School of Pharmaceutical Sciences, Vels Institute of Science Technology & Advanced Studies, Chennai, India
| | - Krishna Chandra Panda
- Department of Pharmaceutical Chemistry, Roland Institute of Pharmaceutical Sciences, Berhampur, India
| | - Shaikh Amir Afzal
- Department of Pharmaceutics, SCES's Indira College of Pharmacy, Pune, India
| | - Shashwat Katiyar
- Department of Biochemistry, School of Life Sciences and Biotechnology, Chhatrapati Shahu Ji Maharaj University, Kanpur, India
| | - Ram C Dhakar
- SRG Hospital and Medical College, Jhalawar, India
| | - Sangeetha Mani
- Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| |
Collapse
|
27
|
Xiang Z, Luo Y, Yu J, Ma H, Zhao Y. Comprehensive Transcriptome-Wide Profiling of 5-Methylcytosine Modifications in Long Non-Coding RNAs in a Rat Model of Traumatic Brain Injury. Curr Issues Mol Biol 2024; 46:14497-14513. [PMID: 39727999 DOI: 10.3390/cimb46120871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/09/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024] Open
Abstract
Traumatic brain injury (TBI) poses a major global health challenge, leading to serious repercussions for those affected and imposing considerable financial strains on families and healthcare systems. RNA methylation, especially 5-methylcytosine (m5C), plays a crucial role as an epigenetic modification in regulating RNA at the level of post-transcriptional regulation. However, the impact of TBI on the m5C methylation profile of long non-coding RNAs (lncRNAs) remains unexplored. In the present study, we conducted a thorough transcriptome-wide examination of m5C methylation in lncRNAs in a rat TBI model utilizing MeRIP-Seq. Our results revealed significant differences in the amount and distribution of m5C methylation in lncRNAs between TBI and control groups, indicating profound changes in m5C methylation following TBI. Bioinformatic analyses linked these specifically methylated transcripts to pathways involved in immune response, neural repair, and lipid metabolism, providing insight into possible mechanisms underlying TBI pathology. These findings offer novel perspectives on the post-transcriptional modifications in lncRNA m5C methylation following TBI, which may contribute to understanding the disease mechanisms and developing targeted therapeutic strategies.
Collapse
Affiliation(s)
- Zhijun Xiang
- Emergency Center, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
| | - Yixing Luo
- Emergency Center, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
| | - Jiangtao Yu
- Emergency Center, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
| | - Haoli Ma
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yan Zhao
- Emergency Center, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
- Hubei Clinical Research Center for Emergency and Resuscitation, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan 430071, China
| |
Collapse
|
28
|
Kumari S, Lausted C, Scherler K, Ng AHC, Lu Y, Lee I, Hood L, Wang K. Approaches and Challenges in Characterizing the Molecular Content of Extracellular Vesicles for Biomarker Discovery. Biomolecules 2024; 14:1599. [PMID: 39766306 PMCID: PMC11674167 DOI: 10.3390/biom14121599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer nanoparticles released from all known cells and are involved in cell-to-cell communication via their molecular content. EVs have been found in all tissues and body fluids, carrying a variety of biomolecules, including DNA, RNA, proteins, metabolites, and lipids, offering insights into cellular and pathophysiological conditions. Despite the emergence of EVs and their molecular contents as important biological indicators, it remains difficult to explore EV-mediated biological processes due to their small size and heterogeneity and the technical challenges in characterizing their molecular content. EV-associated small RNAs, especially microRNAs, have been extensively studied. However, other less characterized RNAs, including protein-coding mRNAs, long noncoding RNAs, circular RNAs, and tRNAs, have also been found in EVs. Furthermore, the EV-associated proteins can be used to distinguish different types of EVs. The spectrum of EV-associated RNAs, as well as proteins, may be associated with different pathophysiological conditions. Therefore, the ability to comprehensively characterize EVs' molecular content is critical for understanding their biological function and potential applications in disease diagnosis. Here, we set out to provide an overview of EV-associated RNAs and proteins as well as approaches currently being used to characterize them.
Collapse
Affiliation(s)
- Suman Kumari
- Institute for Systems Biology, Seattle, WA 98109, USA; (S.K.); (C.L.); (K.S.); (L.H.)
| | - Christopher Lausted
- Institute for Systems Biology, Seattle, WA 98109, USA; (S.K.); (C.L.); (K.S.); (L.H.)
| | - Kelsey Scherler
- Institute for Systems Biology, Seattle, WA 98109, USA; (S.K.); (C.L.); (K.S.); (L.H.)
| | - Alphonsus H. C. Ng
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; (A.H.C.N.); (Y.L.)
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Yue Lu
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; (A.H.C.N.); (Y.L.)
| | - Inyoul Lee
- Institute for Systems Biology, Seattle, WA 98109, USA; (S.K.); (C.L.); (K.S.); (L.H.)
| | - Leroy Hood
- Institute for Systems Biology, Seattle, WA 98109, USA; (S.K.); (C.L.); (K.S.); (L.H.)
| | - Kai Wang
- Institute for Systems Biology, Seattle, WA 98109, USA; (S.K.); (C.L.); (K.S.); (L.H.)
| |
Collapse
|
29
|
Zhang S, Xu B, Zhang JL, Liu SH, Xiang X, Liu P. Prognostic role of long noncoding RNA CASC11 in cancer patients: A meta-analysis. Medicine (Baltimore) 2024; 103:e40823. [PMID: 39686470 PMCID: PMC11651512 DOI: 10.1097/md.0000000000040823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Long noncoding RNA (lncRNA) is a significant component of the noncoding genome and refers to RNA molecules that exceed 200 nucleotides in length. It plays a crucial role in both promoting and suppressing cancer by regulating the proliferation, invasion, and metastasis of tumor cells. We have found a correlation between cancer susceptibility candidate 11 (CASC11) experssion and tumorigenesis development prognosis in a number of studies on tumors. Hence, this meta-analysis was conducted to further investigate the effects of CASC11 expression on clinicopathological features and outcome. Furthermore, CASC11 can act both as a therapeutic target and a cancer biomarker. METHOD We conducted a thorough search of PubMed, Embase, Web of Science, and Cochrane Library to identify all eligible studies until September 20, 2023. These studies examined the potential relationship between the expression levels of CASC11 and survival or the range of pathological feature in cancer patients. The impact of CASC11 expression on overall survival (OS) was evaluated using pooled hazard ratios and 95% confidence intervals (CI). The relationship between CASC11 expression and clinicopathological features was assessed through pooled odds ratios and 95% CI. RESULTS A total of 11 studies, involving 660 patients, were examined in this analysis. The results revealed that the overexpression of CASC11 was significantly associated with poor OS (hazard ratios = 2.07, 95%CI = 1.64-2.60) in cancer. Further subgroup analysis demonstrated that the overexpression of CASC11 was consistently linked to poorer OS in diverse types of cancer, including digestive system neoplasm, respiratory neoplasms, and gynecologic tumor. CONCLUSION Overall, this analysis established a strong correlation between CASC11 expression and tumor prognosis, suggesting its potential as a predictive marker for tumor progression in diverse cancer types.
Collapse
Affiliation(s)
- Song Zhang
- Department of Hepatic-Biliary-Pancreatic Surgery, The Fourth People’s Hospital of Neijiang, Sichuan, China
| | - Bo Xu
- Department of Hepatic-Biliary-Pancreatic Surgery, The Fourth People’s Hospital of Neijiang, Sichuan, China
| | - Ji-Ling Zhang
- Department of Hepatic-Biliary-Pancreatic Surgery, The Fourth People’s Hospital of Neijiang, Sichuan, China
| | - Shun-Hai Liu
- Department of Hepatic-Biliary-Pancreatic Surgery, The First People’s Hospital o Neijiang, Sichuan, China
| | - Xin Xiang
- Department of Hepatic-Biliary-Pancreatic Surgery, The First People’s Hospital o Neijiang, Sichuan, China
| | - Pan Liu
- Department of Hepatic-Biliary-Pancreatic Surgery, The First People’s Hospital o Neijiang, Sichuan, China
| |
Collapse
|
30
|
Elimam H, Alhamshry NAA, Hatawsh A, Elfar N, Moussa R, Radwan AF, Abd-Elmawla MA, Elkashlan AM, Zaki MB, Abdel-Reheim MA, Mohammed OA, Doghish AS. Natural products and long noncoding RNA signatures in gallbladder cancer: a review focuses on pathogenesis, diagnosis, and drug resistance. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9549-9571. [PMID: 39028332 DOI: 10.1007/s00210-024-03279-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/02/2024] [Indexed: 07/20/2024]
Abstract
Gallbladder cancer (GBC) is an aggressive and lethal malignancy with a poor prognosis. Long noncoding RNAs (lncRNAs) and natural products have emerged as key orchestrators of cancer pathogenesis through widespread dysregulation across GBC transcriptomes. Functional studies have revealed that lncRNAs interact with oncoproteins and tumor suppressors to control proliferation, invasion, metastasis, angiogenesis, stemness, and drug resistance. Curcumin, baicalein, oleanolic acid, shikonin, oxymatrine, arctigenin, liensinine, fangchinoline, and dioscin are a few examples of natural compounds that have demonstrated promising anticancer activities against GBC through the regulation of important signaling pathways. The lncRNAs, i.e., SNHG6, Linc00261, GALM, OIP5-AS1, FOXD2-AS1, MINCR, DGCR5, MEG3, GATA6-AS, TUG1, and DILC, are key players in regulating the aforementioned processes. For example, the lncRNAs FOXD2-AS1, DILC, and HOTAIR activate oncogenes such as DNMT1, Wnt/β-catenin, BMI1, and c-Myc, whereas MEG3 and GATA6-AS suppress the tumor proteins NF-κB, EZH2, and miR-421. Clinically, specific lncRNAs can serve as diagnostic or prognostic biomarkers based on overexpression correlating with advanced TNM stage, metastasis, chemoresistance, and poor survival. Therapeutically, targeting aberrant lncRNAs with siRNA or antisense oligos disrupts their oncogenic signaling and inhibits GBC progression. Overall, dysfunctional lncRNA regulatory circuits offer multiple avenues for precision medicine approaches to improve early GBC detection and overcome this deadly cancer. They have the potential to serve as novel biomarkers as they are detectable in bodily fluids and tissues. These findings enhance gallbladder treatments, mitigating resistance to chemo- and radiotherapy.
Collapse
Affiliation(s)
- Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt.
| | - Nora A A Alhamshry
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Abdulrahman Hatawsh
- Biotechnology School, 26th of July Corridor, Sheikh Zayed City, Nile University, Giza, 12588, Egypt
| | - Nourhan Elfar
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, New Administrative Capital, Cairo, 11578, Egypt
- Egyptian Drug Authority (EDA), Ministry of Health and Population, Cairo, 11567, Egypt
| | - Rewan Moussa
- Faculty of Medicine, Helwan University, Cairo, 11795, Egypt
| | - Abdullah F Radwan
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Cairo, 11829, Egypt
| | - Mai A Abd-Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Akram M Elkashlan
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Egypt
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, 11961, Shaqra, Saudi Arabia.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62521, Egypt.
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
- Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt
| |
Collapse
|
31
|
Wang S, Qi X, Liu D, Xie D, Jiang B, Wang J, Wang X, Wu G. The implications for urological malignancies of non-coding RNAs in the the tumor microenvironment. Comput Struct Biotechnol J 2024; 23:491-505. [PMID: 38249783 PMCID: PMC10796827 DOI: 10.1016/j.csbj.2023.12.016] [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: 10/03/2023] [Revised: 12/08/2023] [Accepted: 12/16/2023] [Indexed: 01/23/2024] Open
Abstract
Urological malignancies are a major global health issue because of their complexity and the wide range of ways they affect patients. There's a growing need for in-depth research into these cancers, especially at the molecular level. Recent studies have highlighted the importance of non-coding RNAs (ncRNAs) – these don't code for proteins but are crucial in controlling genes – and the tumor microenvironment (TME), which is no longer seen as just a background factor but as an active player in cancer progression. Understanding how ncRNAs and the TME interact is key for finding new ways to diagnose and predict outcomes in urological cancers, and for developing new treatments. This article reviews the basic features of ncRNAs and goes into detail about their various roles in the TME, focusing specifically on how different ncRNAs function and act in urological malignancies.
Collapse
Affiliation(s)
- Shijin Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Xiaochen Qi
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Deqian Xie
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Bowen Jiang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Jin Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Xiaoxi Wang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning, China
| |
Collapse
|
32
|
Zhao T, Ma F. Roles of Long Noncoding RNA in Prostate Cancer Pathogenesis. Clin Genitourin Cancer 2024; 22:102213. [PMID: 39357460 DOI: 10.1016/j.clgc.2024.102213] [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: 03/22/2024] [Revised: 08/20/2024] [Accepted: 08/24/2024] [Indexed: 10/04/2024]
Abstract
Prostate cancer stands as the most common cancer in men, and research into its genesis and spread is still vital. The idea that the human genome's transcriptional activity is more widespread than previously thought has received empirical validation through the application of deep sequencing-based transcriptome profiling techniques. An assortment of noncoding transcripts longer than 200 nucleotides is referred to as long noncoding RNAs (lncRNAs). Transposable elements comprise a substantial portion of the human genome, with projections indicating that their prospective proportion may reach 90%. Considering they can interact directly with proteins, alter the transcriptional activity of coding genes, and perhaps encode proteins, lncRNAs possess the capability to regulate a variety of biological processes. LncRNAs have been recognized to be key factors in the development of several types of human cancers, including lung, colorectal, and breast cancers, alongside other pathological processes that have a significant impact on the diagnosis and survival of cancer individuals. Furthermore, lncRNAs' discernible expression patterns throughout various cancer scenarios significantly raise their potential as biomarkers and therapeutic targets. We conducted an extensive analysis of the prevailing academic literature on the interaction between lncRNAs and prostate cancer in order to present a solid foundation for potential future studies on the prevention and intervention of prostate cancer. The discourse additionally expands on lncRNAs' prospective applications as targets and biomarkers for medical therapies.
Collapse
Affiliation(s)
- Tongyue Zhao
- Department of Clinical Medicine, Chengdu Medical College, Chengdu City, Sichuan, China
| | - Feng Ma
- Department of Medical Oncology, Jiashan Hospital of Traditional Chinese Medicine, Jiaxing City, China.
| |
Collapse
|
33
|
Kadian LK, Verma D, Lohani N, Yadav R, Ranga S, Gulshan G, Pal S, Kumari K, Chauhan SS. Long non-coding RNAs in cancer: multifaceted roles and potential targets for immunotherapy. Mol Cell Biochem 2024; 479:3229-3254. [PMID: 38413478 DOI: 10.1007/s11010-024-04933-1] [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: 06/06/2023] [Accepted: 01/05/2024] [Indexed: 02/29/2024]
Abstract
Cancer remains a major global health concern with high mortality rates mainly due to late diagnosis and poor prognosis. Long non-coding RNAs (lncRNAs) are emerging as key regulators of gene expression in human cancer, functioning through various mechanisms including as competing endogenous RNAs (ceRNAs) and indirectly regulating miRNA expression. LncRNAs have been found to have both oncogenic and tumor-suppressive roles in cancer, with the former promoting cancer cell proliferation, migration, invasion, and poor prognosis. Recent research has shown that lncRNAs are expressed in various immune cells and are involved in cancer cell immune escape and the modulation of the tumor microenvironment, thus highlighting their potential as targets for cancer immunotherapy. Targeting lncRNAs in cancer or immune cells could enhance the anti-tumor immune response and improve cancer immunotherapy outcomes. However, further research is required to fully understand the functional roles of lncRNAs in cancer and the immune system and their potential as targets for cancer immunotherapy. This review offers a comprehensive examination of the multifaceted roles of lncRNAs in human cancers, with a focus on their potential as targets for cancer immunotherapy. By exploring the intricate mechanisms underlying lncRNA-mediated regulation of cancer cell proliferation, invasion, and immune evasion, we provide insights into the diverse therapeutic applications of these molecules.
Collapse
Affiliation(s)
- Lokesh K Kadian
- Dept of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
- Dept of Dermatology, Indiana University School of Medicine, Indianapolis, 46202, USA
| | - Deepika Verma
- Dept of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Neelam Lohani
- Dept of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Ritu Yadav
- Dept of Genetics, MD University, Rohtak, 124001, India
| | - Shalu Ranga
- Dept of Genetics, MD University, Rohtak, 124001, India
| | - Gulshan Gulshan
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai, Maharashtra, India
| | - Sanghapriya Pal
- Dept of Biochemistry, Maulana Azad Medical College and Associated Hospital, New Delhi, 110002, India
| | - Kiran Kumari
- Dept of Forensic Science, Lovely Professional University, Jalandhar, Punjab, 144411, India
| | - Shyam S Chauhan
- Dept of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
34
|
Alissa M, Aldurayhim M, Abdulaziz O, Alsalmi O, Awad A, Algopishi UB, Alharbi S, Safhi AY, Khan KH, Uffar C. From molecules to heart regeneration: Understanding the complex and profound role of non-coding RNAs in stimulating cardiomyocyte proliferation for cardiac repair. Curr Probl Cardiol 2024; 49:102857. [PMID: 39306148 DOI: 10.1016/j.cpcardiol.2024.102857] [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: 09/07/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024]
Abstract
Recent studies of noncoding genomes have shown important implications for regulating gene expression and genetic programs during development and their association with health, including cardiovascular disease. There are nearly 2,500 microRNAs (miRNAs), 12,000 long-chain non-coding RNAs (lncRNA), and nearly 4,000 circular RNAs (circles). Even though they do not code for proteins, they make up nearly 99% of the human genome. Non-coding RNA families (ncRNAs) have recently been discovered and established as novel and necessary controllers of cardiovascular risk factors and cellular processes and, therefore, have the potential to improve the diagnosis and prediction of cardiovascular disease. The increase in the prevalence of cardiovascular disease can be explained by the shortcomings of existing therapies, which focus only on the non-coding RNAs that protein codes for. On the other hand, recent studies point to the possibility of using ncRNAs in the early detection and intervention of CVD. These findings suggest that developing diagnostic tools and therapies based on miRNAs, lncRNAs, and circRNAs will potentially enhance the clinical management of patients with cardiovascular disease. Cardiovascular diseases include CH, HF, RHD, ACS, MI, AS, MF, ARR, and PAH, of which CH is the most common cardiovascular disease, followed by HF and RHD. This paper aims to elucidate the biological and clinical significance of miRNAs, increase, and circles, as well as their expression profiles and the possibility of regulating non-coding transcripts in cardiovascular diseases to improve the application of ncRNAs in diagnosis and treatment.
Collapse
Affiliation(s)
- Mohammed Alissa
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Mohammed Aldurayhim
- Department of Medical Laboratory, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Osama Abdulaziz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21974, Saudi Arabia
| | - Ohud Alsalmi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21974, Saudi Arabia
| | - Alsamghan Awad
- King Khalid University, College of Medicine, Family Medicine department, Saudi Arabia
| | | | - Sarah Alharbi
- Department of Medical Laboratory, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Awaji Y Safhi
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Khadijah Hassan Khan
- Department of Laboratory, King Faisal Medical Complex, Ministry of Health, Taif 26514, Saudi Arabia
| | - Christin Uffar
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| |
Collapse
|
35
|
Jafari SH, Lajevardi ZS, Zamani Fard MM, Jafari A, Naghavi S, Ravaei F, Taghavi SP, Mosadeghi K, Zarepour F, Mahjoubin-Tehran M, Rahimian N, Mirzaei H. Imaging Techniques and Biochemical Biomarkers: New Insights into Diagnosis of Pancreatic Cancer. Cell Biochem Biophys 2024; 82:3123-3144. [PMID: 39026059 DOI: 10.1007/s12013-024-01437-z] [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] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
Pancreatic cancer (PaC) incidence is increasing, but our current screening and diagnostic strategies are not very effective. However, screening could be helpful in the case of PaC, as recent evidence shows that the disease progresses gradually. Unfortunately, there is no ideal screening method or program for detecting PaC in its early stages. Conventional imaging techniques, such as abdominal ultrasound, CT, MRI, and EUS, have not been successful in detecting early-stage PaC. On the other hand, biomarkers may be a more effective screening tool for PaC and have greater potential for further evaluation compared to imaging. Recent studies on biomarkers and artificial intelligence (AI)-enhanced imaging have shown promising results in the early diagnosis of PaC. In addition to proteins, non-coding RNAs are also being studied as potential biomarkers for PaC. This review consolidates the current literature on PaC screening modalities to provide an organized framework for future studies. While conventional imaging techniques have not been effective in detecting early-stage PaC, biomarkers and AI-enhanced imaging are promising avenues of research. Further studies on the use of biomarkers, particularly non-coding RNAs, in combination with imaging modalities may improve the accuracy of PaC screening and lead to earlier detection of this deadly disease.
Collapse
Affiliation(s)
- Seyed Hamed Jafari
- Medical Imaging Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Radiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Sadat Lajevardi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Masoud Zamani Fard
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ameneh Jafari
- Chronic Respiratory Diseases Research Center, NRITLD, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soroush Naghavi
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ravaei
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Pouya Taghavi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Kimia Mosadeghi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Zarepour
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran; Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
36
|
Kaur P, Sharma P, Bhatia P, Singh M. Recent advances on biogenesis, functions and therapeutic potential of long noncoding RNAs in T cell acute lymphoblastic leukemia. Discov Oncol 2024; 15:729. [PMID: 39612075 DOI: 10.1007/s12672-024-01618-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
T-cell Acute Lymphoblastic Leukemia (T-ALL) is a highly aggressive form of ALL with at least 25% relapse rates. The high relapse rates are often linked to poor prognoses. More detailed studies for novel therapeutic targets for the treatment of T-ALL are required as the genetic and transcriptomic data currently available on T-ALL pathophysiology is insufficient. Long non-coding RNAs are emerging as important players in the regulation of tumour proliferation and metastasis. Studies on various cancers have revealed their potential as biomarkers and therapeutic targets in treatment. This review describes the characterization, biosynthesis, and role of long non-coding RNA in T-ALL and highlights their potential as next generation molecule in development of promising diagnostic, prognostic and/or therapeutic markers.
Collapse
Affiliation(s)
- Parminder Kaur
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pankaj Sharma
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Prateek Bhatia
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Minu Singh
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
37
|
Ngowi EE, Lu T, Liu Q, Xie X, Wang N, Luo L, Deng L, Zhou Y, Zhang Z, Qiao A. Biofluid-Derived Exosomal LncRNAs: Their Potential in Obesity and Related Comorbidities. BIOLOGY 2024; 13:976. [PMID: 39765643 PMCID: PMC11673191 DOI: 10.3390/biology13120976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 01/03/2025]
Abstract
Obesity has escalated into a critical global health crisis, tripling in prevalence since the mid-1970s. This increase mirrors the rise in metabolic-associated diseases such as type 2 diabetes (T2D) and its complications, certain cancers, and cardiovascular conditions. While substantial research efforts have enriched our understanding and led to the development of innovative management strategies for these diseases, the suboptimal response rates of existing therapies remain a major obstacle to effectively managing obesity and its associated conditions. Over the years, inter-organ communication (IOC) has emerged as a crucial factor in the development and progression of metabolic disorders. Exosomes, which are nano-sized vesicular couriers released by cells, play a significant role in this communication by transporting proteins, lipids, and nucleic acids across cellular landscapes. The available evidence indicates that exosomal RNAs present in biofluids such as blood, urine, milk, vitreous humor (VH), and cerebrospinal fluid (CSF) are altered in numerous diseases, suggesting their diagnostic and therapeutic potential. Long non-coding RNAs contained in exosomes (exo-lncRNAs) have attracted considerable interest, owing to their ability to interact with critical components involved in a multitude of metabolic pathways. Recent studies have found that alterations in exo-lncRNAs in biofluids correlate with several metabolic parameters in patients with metabolic-associated conditions; however, their exact roles remain largely unclear. This review highlights the diagnostic and therapeutic potential of exosomal lncRNAs in obesity and its associated conditions, emphasizing their role in IOC and disease progression, aiming to pave the way for further research in this promising domain.
Collapse
Affiliation(s)
- Ebenezeri Erasto Ngowi
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Tuyan Lu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Qing Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Xianghong Xie
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Ning Wang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Liping Luo
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Lijuan Deng
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Yinghua Zhou
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Zhihong Zhang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
| | - Aijun Qiao
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; (E.E.N.); (T.L.); (Q.L.); (X.X.); (N.W.); (L.L.); (L.D.) (Y.Z.); (Z.Z.)
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
38
|
Zhao Z, Yang Y, Iqbal A, Wu Q, Zhou L. Biological Insights and Recent Advances in Plant Long Non-Coding RNA. Int J Mol Sci 2024; 25:11964. [PMID: 39596034 PMCID: PMC11593582 DOI: 10.3390/ijms252211964] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Long non-coding RNA (lncRNA) refers to an RNA molecule longer than 200 nucleotides (nt) that plays a significant role in regulating essential molecular and biological processes. It is commonly found in animals, plants, and viruses, and is characterized by features such as epigenetic markers, developmental stage-specific expression, and tissue-specific expression. Research has shown that lncRNA participates in anatomical processes like plant progression, while also playing a crucial role in plant disease resistance and adaptation mechanisms. In this review, we provide a concise overview of the formation mechanism, structural characteristics, and databases related to lncRNA in recent years. We primarily discuss the biological roles of lncRNA in plant progression as well as its involvement in response to biotic and abiotic stresses. Additionally, we examine the current challenges associated with lncRNA and explore its potential application in crop production and breeding. Studying plant lncRNAs is highly significant for multiple reasons: It reveals the regulatory mechanisms of plant growth and development, promotes agricultural production and food security, and drives research in plant genomics and epigenetics. Additionally, it facilitates ecological protection and biodiversity conservation.
Collapse
Affiliation(s)
- Zhihao Zhao
- National Key Laboratory for Tropical Crop Breeding, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China; (Z.Z.); (Y.Y.); (Q.W.)
- Hainan Key Laboratory of Tropical Oil Crops Biology/Coconut Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wenchang 571339, China;
- Industrial Development Department, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Yaodong Yang
- National Key Laboratory for Tropical Crop Breeding, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China; (Z.Z.); (Y.Y.); (Q.W.)
- Hainan Key Laboratory of Tropical Oil Crops Biology/Coconut Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wenchang 571339, China;
| | - Amjad Iqbal
- Hainan Key Laboratory of Tropical Oil Crops Biology/Coconut Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wenchang 571339, China;
- Department of Food Science & Technology, Abdul Wali Khan University Mardan, Khyber Pakhtunkhwa 23200, Pakistan
| | - Qiufei Wu
- National Key Laboratory for Tropical Crop Breeding, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China; (Z.Z.); (Y.Y.); (Q.W.)
- Hainan Key Laboratory of Tropical Oil Crops Biology/Coconut Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wenchang 571339, China;
| | - Lixia Zhou
- National Key Laboratory for Tropical Crop Breeding, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China; (Z.Z.); (Y.Y.); (Q.W.)
- Hainan Key Laboratory of Tropical Oil Crops Biology/Coconut Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wenchang 571339, China;
| |
Collapse
|
39
|
Sishuai S, Lingui G, Pengtao L, Xinjie B, Junji W. Advances in regulating endothelial-mesenchymal transformation through exosomes. Stem Cell Res Ther 2024; 15:391. [PMID: 39482726 PMCID: PMC11529026 DOI: 10.1186/s13287-024-04010-w] [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: 07/30/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
Endothelial-mesenchymal transformation (EndoMT) is the process through which endothelial cells transform into mesenchymal cells, affecting their morphology, gene expression, and function. EndoMT is a potential risk factor for cardiovascular and cerebrovascular diseases, tumor metastasis, and fibrosis. Recent research has highlighted the role of exosomes, a mode of cellular communication, in the regulation of EndoMT. Exosomes from diseased tissues and microenvironments can promote EndoMT, increase endothelial permeability, and compromise the vascular barrier. Conversely, exosomes derived from stem cells or progenitor cells can inhibit the EndoMT process and preserve endothelial function. By modifying exosome membranes or contents, we can harness the advantages of exosomes as carriers, enhancing their targeting and ability to inhibit EndoMT. This review aims to systematically summarize the regulation of EndoMT by exosomes in different disease contexts and provide effective strategies for exosome-based EndoMT intervention.
Collapse
Affiliation(s)
- Sun Sishuai
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Gu Lingui
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Pengtao
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Bao Xinjie
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| | - Wei Junji
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
40
|
Wang Y, Zhao Y, Liu Q, Yang J, Xu Z, Du W, Tang G, Zhang C, Si X, Wang J. Identifying functional cuproptosis-related long non-coding RNAs in patients with bladder cancer. Transl Cancer Res 2024; 13:5178-5189. [PMID: 39525026 PMCID: PMC11543048 DOI: 10.21037/tcr-23-2367] [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: 12/25/2023] [Accepted: 08/29/2024] [Indexed: 11/16/2024]
Abstract
Background Bladder cancer is the most common malignancy of the urinary tract and one of the most common cancers in the world. Cuproptosis is a novel type of cell death associated with tumorigenesis. In this study, we assessed the correlation between cuproptosis-related genes and tumorigenesis. Moreover, we constructed a prognostic signature. Methods Pearson correlation analysis and univariate Cox regression were utilized to extract cuproptosis-related long non-coding RNAs (lncRNAs) predicting prognosis in The Cancer Genome Atlas (TCGA) database. The least absolute shrinkage and selection operator (LASSO) Cox regression was utilized to establish a cuproptosizs-related prognostic signature. A nomogram signature was generated to predict individual survival. Results We obtained 19 cuproptosis-related genes and 14 prognostic cuproptosis-related lncRNAs. We constructed a seven-prognostic risk signature. Time-dependent receiver operating characteristic (ROC) curves demonstrated good predictive power (1-, 3-, and 5-year survival rates of 0.711, 0.673, and 0.684, respectively). The high-risk group reported a worse prognosis than the low-risk group, and the risk signature was identified as an independent factor. The biological process of risk-related genes primarily involved tumorigenesis and migration. The high-risk group expressed high chemokines and T cell inhibition and low antigen-presenting cells. Conclusions Cuproptosis-related lncRNAs are central to tumorigenesis, providing a novel therapeutic target for patients with bladder cancer. We constructed an individualized predictive signature based on cuproptosis-related lncRNAs.
Collapse
Affiliation(s)
- Yunchao Wang
- Department of Urology, the First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Yihan Zhao
- Department of Urology, the First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Qing Liu
- Department of Medical Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jiwei Yang
- Department of Urology, the First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Zhipeng Xu
- Department of Urology, the First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Wenzhi Du
- Department of Urology, the First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Guanbao Tang
- Department of Urology, the First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Chuanpai Zhang
- Department of Urology, the First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Xiaoqing Si
- Department of Dermatology, the First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Jianning Wang
- Department of Urology, the First Affiliated Hospital of Shandong First Medical University, Jinan, China
| |
Collapse
|
41
|
Liang P, Zhu M, Sun X, Wang L, Li B, Ming S, Younis M, Yang J, Wu Y, Huang X. LncRNA-mRNA co-expression analysis reveals aquaporin-9-promoted neutrophil extracellular trap formation and inflammatory activation in sepsis. Int Immunopharmacol 2024; 140:112916. [PMID: 39133961 DOI: 10.1016/j.intimp.2024.112916] [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: 06/03/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024]
Abstract
Sepsis is a life-threatening condition caused by an excessive inflammatory response to an infection. However, the precise regulatory mechanism of sepsis remains unclear. Using a strand-specific RNA-sequencing, we identified 115 hub differentially expressed long noncoding RNAs (lncRNAs) and 443 mRNAs in septic patients, primarily participated in crucial pathways including neutrophil extracellular trap (NET) formation and toll-like receptor signaling. Notably, NETs related gene aquaporin-9 (AQP9) and its associated lncRNAs exhibited significant upregulation in septic neutrophils. Functional experiments revealed AQP9 interacts with its lncRNAs to augment the formation of neutrophil NETs. In murine sepsis models, AQP9 inhibition with phloretin reduced proinflammatory cytokine production and lung damage. These findings provide crucial insights into the regulatory role of AQP9 in sepsis, unraveling its interaction with associated lncRNAs in transmitting downstream signals, holding promise in informing the development of novel therapeutic strategies aimed at ameliorating the debilitating effects of sepsis.
Collapse
Affiliation(s)
- Pingping Liang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Manman Zhu
- Department of Clinical Laboratory, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Xingzi Sun
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Li Wang
- Department of Obstetrics and Gynecology, Perinatal Medical Center, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Bin Li
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Siqi Ming
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Muhammad Younis
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Jianhua Yang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China.
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China; Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China.
| |
Collapse
|
42
|
Pinkney HR, Ross CR, Hodgson TO, Pattison ST, Diermeier SD. Discovery of prognostic lncRNAs in colorectal cancer using spatial transcriptomics. NPJ Precis Oncol 2024; 8:230. [PMID: 39390212 PMCID: PMC11467462 DOI: 10.1038/s41698-024-00728-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024] Open
Abstract
Colorectal cancer (CRC) exhibits significant genetic and epigenetic diversity, evolving into sub-clonal populations with varied metastatic potentials and treatment responses. Predicting metastatic disease in CRC patients remains challenging, underscoring the need for reliable biomarkers. While most research on therapeutic targets and biomarkers has focused on proteins, non-coding RNAs such as long non-coding RNAs (lncRNAs) comprise most of the transcriptome and demonstrate superior tissue- and cancer-specific expression. We utilised spatial transcriptomics to investigate lncRNAs in CRC tumours, offering more precise cell-type-specific expression data compared to bulk RNA sequencing. Our analysis identified 301 lncRNAs linked to malignant CRC regions, which we validated with public data. Further validation using RNA-FISH revealed three lncRNAs (LINC01978, PLAC4, and LINC01303) that are detectable in stage II tumours but not in normal epithelium and are upregulated in metastatic tissues. These lncRNAs hold potential as biomarkers for early risk assessment of metastatic disease.
Collapse
Affiliation(s)
- Holly R Pinkney
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | | | | | | | - Sarah D Diermeier
- Department of Biochemistry, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
43
|
Kubaski Benevides AP, Marin AM, Wosniaki DK, Oliveira RN, Koerich GM, Kusma BN, Munhoz EC, Zanette DL, Aoki MN. Expression of HOTAIR and PTGS2 as potential biomarkers in chronic myeloid leukemia patients in Brazil. Front Oncol 2024; 14:1443346. [PMID: 39450252 PMCID: PMC11499243 DOI: 10.3389/fonc.2024.1443346] [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: 06/03/2024] [Accepted: 09/16/2024] [Indexed: 10/26/2024] Open
Abstract
Chronic myeloid leukemia (CML) is a clonal myeloproliferative neoplasm in which all the patients has the translocation (9;22) that generates de BCR::ABL1 tyrosine kinase. Despite this disease possessing a good biomarker (BCR::ABL1 transcripts level) for diagnosis and prognosis, many studies has been performed to investigate other molecules, such as the long noncoding RNAs (lncRNAs) and mRNAs, as potential biomarkers with the aim of predicting a change in BCR::ABL1 levels and as an associated biomarker. A RNAseq was performed comparing 6 CML patients with high BCR::ABL1 expression with 6 healthy control individuals, comprising the investigation cohort to investigate these molecules. To validate the results obtained by RNAseq, samples of 87 CML patients and 42 healthy controls were used in the validation cohort by RT-qPCR assays. The results showed lower expression of HOTAIR and PTGS2 in CML patients. The HOTAIR expression is inversely associated with BCR::ABL1 expression in imatinib-treated CML patients, and to PTGS2 showing that CML patients with high BCR::ABL1 expression showed reduced PTGS2 expression.
Collapse
Affiliation(s)
- Ana Paula Kubaski Benevides
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Anelis Maria Marin
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Denise K. Wosniaki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Rafaela Noga Oliveira
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Gabriela Marino Koerich
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Bianca Nichele Kusma
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | | | - Dalila Luciola Zanette
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Mateus Nóbrega Aoki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| |
Collapse
|
44
|
Li J, Wang H, Zhang S, Quan L, Zhou X. Identification and validation of an m7G-related lncRNAs signature for predicting prognosis, immune response and therapy landscapes in ovarian cancer. Front Genet 2024; 15:1466422. [PMID: 39440245 PMCID: PMC11493627 DOI: 10.3389/fgene.2024.1466422] [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: 07/18/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024] Open
Abstract
Background Ovarian cancer is the most mortality malignancy in gynecology. N7-methylguanosine (m7G) is one of the most prevalent RNA modifications in the development and progression of cancer. The aim of this study is to investigate the effect of m7G-related lncRNA on ovarian cancer in terms of instruction prognosis and immunotherapy. Methods After integrating and processing the RNA expression profiles with the clinical sample information in the TCGA database, we initially screened to the m7G-related lncRNAs by Spearman correlation analysis, and subsequently obtained a prognostic model constructed by five m7G-related lncRNAs with Univariate Cox analysis, LASSO regression analysis, and Multivariate Cox regression analysis, after which we further evaluated and validated the prognostic value of the model using Kaplan-Meier survival analysis, Principal component analysis, Nomogram, and ROC curve. In addition, based on this risk model, we explored the differentially enriched pathways and functions of the high and low risk groups, and characterized the immune cells, immune functions, gene mutations, and drug sensitivity between the two groups. Results After a series of rigorous filtering, we finally attained a prognostic risk model consisting of KRT7-AS, USP30-AS1, ZFHX4-AS1, ACAP2-IT1, and TWSG1-DT which is excellent in predicting the prognostic survival of ovarian cancer patients as well as existing as an independent prognostic factor. Moreover, the model has certain relevance in the immune cells and functions between high and low risk groups, and simultaneously, the signature has the role of guiding the option of immunotherapy and chemotherapeutic drugs. Conclusion Altogether, our study established a tight connection between m7G-associated lncRNAs and ovarian cancer, with potential that the prognostic patterns contribute to steering the prognosis of ovarian cancer patients, measuring the efficacy of immunotherapeutic approaches, and detecting effective chemotherapeutic agents.
Collapse
Affiliation(s)
| | | | | | | | - Xin Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
45
|
Maqbool M, Hussain MS, Bisht AS, Kumari A, Kamran A, Sultana A, Kumar R, Khan Y, Gupta G. Connecting the dots: LncRNAs in the KRAS pathway and cancer. Pathol Res Pract 2024; 262:155570. [PMID: 39226802 DOI: 10.1016/j.prp.2024.155570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/17/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024]
Abstract
Long non-coding RNAs (lncRNAs) have been identified as important participants in several biological functions, particularly their complex interactions with the KRAS pathway, which provide insights into the significant roles lncRNAs play in cancer development. The KRAS pathway, a central signaling cascade crucial for cell proliferation, survival, and differentiation, stands out as a key therapeutic target due to its aberrant activation in many human cancers. Recent investigations have unveiled a myriad of lncRNAs, such as H19, ANRIL, and MEG3, intricately modulating the KRAS pathway, influencing both its activation and repression through various mechanisms, including epigenetic modifications, transcriptional regulation, and post-transcriptional control. These lncRNAs function as fine-tuners, delicately orchestrating the balance required for normal cellular function. Their dysregulation has been linked to the development and progression of multiple malignancies, including lung, pancreatic, and colorectal carcinomas, which frequently harbor KRAS mutations. This scrutiny delves into the functional diversity of specific lncRNAs within the KRAS pathway, elucidating their molecular mechanisms and downstream effects on cancer phenotypes. Additionally, it underscores the diagnostic and prognostic potential of these lncRNAs as indicators for cancer detection and assessment. The complex regulatory network that lncRNAs construct within the context of the KRAS pathway offers important insights for the creation of focused therapeutic approaches, opening new possibilities for precision medicine in oncology. However, challenges such as the dual roles of lncRNAs in different cancer types and the difficulty in therapeutically targeting these molecules highlight the ongoing debates and need for further research. As ongoing studies unveil the complexities of lncRNA-mediated KRAS pathway modulation, the potential for innovative cancer interventions becomes increasingly promising.
Collapse
Affiliation(s)
- Mudasir Maqbool
- Department of Pharmaceutical Sciences, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Md Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand 248007, India.
| | - Ajay Singh Bisht
- School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Patel Nagar, Dehradun, Uttarakhand 248001, India
| | - Alka Kumari
- University institute of pharmacy, Chandigarh University, Gharaun, Punjab 140413, India
| | - Almaz Kamran
- HIMT College of Pharmacy, Plot No. 08, Knowledge Park - 1, Greater Noida, Uttar Pradesh 201310, India
| | - Ayesha Sultana
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya University, Deralakatte, Mangalore, Karnataka, India
| | - Rajesh Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Yumna Khan
- Institute of Biotechnology and Genetic Engineering (Health Division), The University of Agriculture, Peshawar, Khyber Pakhtunkhwa 25000, Pakistan
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Punjab, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| |
Collapse
|
46
|
Pathoor NN, Ganesh PS. Unveiling the nexus: Long non-coding RNAs and the PI3K/Akt pathway in oral squamous cell carcinoma. Pathol Res Pract 2024; 262:155540. [PMID: 39142241 DOI: 10.1016/j.prp.2024.155540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 08/16/2024]
Abstract
The PI3K/Akt pathway plays a critical role in the progression and treatment of oral squamous cell carcinoma (OSCC). Recent research has uncovered the involvement of long non-coding RNAs (lncRNAs) in regulating this pathway, influencing OSCC cell proliferation, survival, and metastasis. This review explores the latest findings on how certain lncRNAs act as either cancer promoters or cancer inhibitors within the PI3K/Akt signaling pathway. Certain lncRNAs act as oncogenic or tumor-suppressive agents, making them potential diagnostic and prognostic markers. Targeting these lncRNAs may lead to novel therapeutic strategies. The evolving fields of precision medicine and artificial intelligence promise advancements in OSCC diagnosis and treatment, enabling more personalized and effective patient care.
Collapse
Affiliation(s)
- Naji Naseef Pathoor
- Department of Microbiology, Centre for infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu 600077, India
| | - Pitchaipillai Sankar Ganesh
- Department of Microbiology, Centre for infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu 600077, India.
| |
Collapse
|
47
|
Li Z, Li X, Lin J, Wang Y, Cao H, Zhou J. Reevaluation by the CRISPR/Cas9 knockout approach revealed that multiple pluripotency-associated lncRNAs are dispensable for pluripotency maintenance while Snora73a/b is essential for pluripotency exit. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2198-2212. [PMID: 38995489 DOI: 10.1007/s11427-023-2594-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 03/15/2024] [Indexed: 07/13/2024]
Abstract
Many long noncoding RNAs (lncRNAs) have been identified through siRNA-based screening as essential regulators of embryonic stem cell (ESC) pluripotency. However, the biological and molecular functions of most lncRNAs remain unclear. Here, we employed CRISPR/Cas9-mediated knockout technology to explore the functions of 8 lncRNAs previously reported to promote pluripotency in mouse ESCs. Unexpectedly, all of these lncRNAs were dispensable for pluripotency maintenance and proliferation in mouse ESCs when disrupted individually or in combination. Single-cell transcriptomic analysis also showed that the knockout of these lncRNAs has a minimal impact on pluripotency gene expression and cell identity. We further showed that several small hairpin RNAs (shRNAs) previously used to knock down lncRNAs caused the downregulation of pluripotency genes in the corresponding lncRNA-knockout ESCs, indicating that off-target effects likely responsible for the pluripotency defects caused by these shRNAs. Interestingly, linc1343-knockout and linc1343-knockdown ESCs failed to form cystic structures and exhibited high expression of pluripotency genes during embryoid body (EB) differentiation. By reintroducing RNA products generated from the linc1343 locus, we found that two snoRNAs, Snora73a and Snora73b, but not lncRNAs, could rescue pluripotency silencing defects during EB differentiation of linc1343 knockout ESCs. Our results suggest that the 8 previously annotated pluripotency-regulating lncRNAs have no overt functions in conventional ESC culture; however, we identified snoRNA products derived from an annotated lncRNA locus as essential regulators for silencing pluripotency genes.
Collapse
Affiliation(s)
- Zhen Li
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
| | - Xuefei Li
- Dermatology Hospital, Southern Medical University, Guangzhou, 510091, China
| | - Jingxia Lin
- Dermatology Hospital, Southern Medical University, Guangzhou, 510091, China
| | - Yangming Wang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, 100871, China
| | - Huiqing Cao
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China.
| | - Jiajian Zhou
- Dermatology Hospital, Southern Medical University, Guangzhou, 510091, China.
| |
Collapse
|
48
|
Yin Z, Wang J, Zhu C, Xu C, Fang J, Li Q. Identification and Verification of a Novel Disulfidptosis-Related lncRNAs Prognostic Signature to Predict the Prognosis and Immune Activity of Head and Neck Squamous Carcinoma. IRANIAN JOURNAL OF PUBLIC HEALTH 2024; 53:2328-2340. [PMID: 39544861 PMCID: PMC11557754 DOI: 10.18502/ijph.v53i10.16720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/14/2024] [Indexed: 11/17/2024]
Abstract
Background We aimed to explore the prediction value of disulfidptosis-related long noncoding RNAs (lncRNAs) on the prognosis and immunotherapy efficiency of patients with head and neck squamous carcinoma (HNSCC). Methods Clinical and RNA-seq information were collected from The Cancer Genome Atlas (TCGA) and Genome Data Sharing (GDC) portal. The Pearson correlation analysis, univariate COX regression analysis, the least absolute shrinkage and selection operator (LASSO) COX regression were employed to construct the disulfidptosis-related lncRNAs (DRLs) prognostic model. The Kaplan-Meier survival curve, principal component analysis (PCA), receiver operating characteristic (ROC) curves and areas under the curves (AUCs) were used to examine the accuracy of the prognostic model. ssGSEA, mutation and functional and gene set enrichment analysis was performed to quantify the immune cell infiltration, immune function and functional enrichments. Finally, the mRNA expression of the DRLs was verified by real-time PCR (RT-PCR) in HNSCC cells. Results A new DRLs prognostic model (AC083967.1, AC106820.5, AC245041.2, AL590617.2, AP002478.1, and VPS9D1-AS1) with an independent prognostic value of HNSCC patients was successfully identified. In addition, the DRLs prognostic model was related with immune signature and drug therapy response. Meanwhile, the mRNA expression level of the 6 DRLs detected by RT-PCR was consistent with the results of bioinformatic analysis. Conclusion We developed a new DRLs prognostic model of HNSCC, which could effectively predicate the prognosis and therapy response of HNSCC patients and provide insights into personalized therapeutics.
Collapse
Affiliation(s)
- Zi Yin
- Department of Pathology, School of Basic Medicine, Hubei University of Medicine, Shiyan, China
- Clinical Pathology Test and Consultation Center, Hubei University of Medicine, Shiyan, China
| | - Jue Wang
- Clinical Pathology Test and Consultation Center, Hubei University of Medicine, Shiyan, China
| | - Changqing Zhu
- Department of Pathology, School of Basic Medicine, Hubei University of Medicine, Shiyan, China
- Department of Pathology, Shiyan Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Chenli Xu
- Department of Pathology, School of Basic Medicine, Hubei University of Medicine, Shiyan, China
- Forensic Judicial Appraisal Institute, Hubei University of Medicine, Shiyan, China
| | - Juan Fang
- Department of Pathology, School of Basic Medicine, Hubei University of Medicine, Shiyan, China
- Clinical Pathology Test and Consultation Center, Hubei University of Medicine, Shiyan, China
| | - Qiaoqin Li
- Department of Pathology, School of Basic Medicine, Hubei University of Medicine, Shiyan, China
- Clinical Pathology Test and Consultation Center, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
49
|
Fischer KD, Tiwari S, Thier B, Qiu LC, Lin TC, Paschen A, Imig J. Long non-coding RNA GRASLND links melanoma differentiation and interferon-gamma response. Front Mol Biosci 2024; 11:1471100. [PMID: 39398277 PMCID: PMC11466874 DOI: 10.3389/fmolb.2024.1471100] [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: 07/26/2024] [Accepted: 09/13/2024] [Indexed: 10/15/2024] Open
Abstract
Melanoma is a highly malignant tumor, that stands as the most lethal form of skin cancer and is characterized by notable phenotypic plasticity and intratumoral heterogeneity. Melanoma plasticity is involved in tumor growth, metastasis and therapy resistance. Long non-coding RNAs (lncRNAs) could influence plasticity due to their regulatory function. However, their role and mode of action are poorly studied. Here, we show a relevance of lncRNA GRASLND in melanoma differentiation and IFNγ signaling. GRASLND knockdown revealed switching of differentiated, melanocytic melanoma cells towards a dedifferentiated, slow-proliferating and highly-invasive cell state. Interestingly, GRASLND is overexpressed in differentiated melanomas and associated with poor prognosis. Accordingly, we found GRASLND expressed in immunological "cold" tumors and it negatively correlates with gene signatures of immune response activation. In line, silencing of GRASLND under IFNγ enhanced the expression of IFNγ-stimulated genes, including HLA-I antigen presentation, demonstrating suppressive activity of GRASLND on IFNγ signaling. Our findings demonstrate that in differentiated melanomas elevated expression of GRASLND interferes with anti-tumor effects of IFNγ, suggesting a role of GRASLND in tumor immune evasion.
Collapse
Affiliation(s)
- Kim Denise Fischer
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Faculty of Chemistry and Chemical Biology, Technical University of Dortmund, Dortmund, Germany
| | - Shashank Tiwari
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Beatrice Thier
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Lin Christina Qiu
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Faculty of Chemistry and Chemical Biology, Technical University of Dortmund, Dortmund, Germany
| | - Tzu-Chen Lin
- Faculty of Chemistry and Chemical Biology, Technical University of Dortmund, Dortmund, Germany
| | - Annette Paschen
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Jochen Imig
- Chemical Genomics Centre, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| |
Collapse
|
50
|
Bhat AA, Afzal M, Moglad E, Thapa R, Ali H, Almalki WH, Kazmi I, Alzarea SI, Gupta G, Subramaniyan V. lncRNAs as prognostic markers and therapeutic targets in cuproptosis-mediated cancer. Clin Exp Med 2024; 24:226. [PMID: 39325172 PMCID: PMC11427524 DOI: 10.1007/s10238-024-01491-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
Long non-coding RNAs (lncRNAs) have emerged as crucial regulators in various cellular processes, including cancer progression and stress response. Recent studies have demonstrated that copper accumulation induces a unique form of cell death known as cuproptosis, with lncRNAs playing a key role in regulating cuproptosis-associated pathways. These lncRNAs may trigger cell-specific responses to copper stress, presenting new opportunities as prognostic markers and therapeutic targets. This paper delves into the role of lncRNAs in cuproptosis-mediated cancer, underscoring their potential as biomarkers and targets for innovative therapeutic strategies. A thorough review of scientific literature was conducted, utilizing databases such as PubMed, Google Scholar, and ScienceDirect, with search terms like 'lncRNAs,' 'cuproptosis,' and 'cancer.' Studies were selected based on their relevance to lncRNA regulation of cuproptosis pathways and their implications for cancer prognosis and treatment. The review highlights the significant contribution of lncRNAs in regulating cuproptosis-related genes and pathways, impacting copper metabolism, mitochondrial stress responses, and apoptotic signaling. Specific lncRNAs are potential prognostic markers in breast, lung, liver, ovarian, pancreatic, and gastric cancers. The objective of this article is to explore the role of lncRNAs as potential prognostic markers and therapeutic targets in cancers mediated by cuproptosis.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al Kharj, Saudi Arabia
| | - Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|