1
|
Zarandi PK, Ghiasi M, Heiat M. The role and function of lncRNA in ageing-associated liver diseases. RNA Biol 2025; 22:1-8. [PMID: 39697114 PMCID: PMC11660375 DOI: 10.1080/15476286.2024.2440678] [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] [Revised: 10/09/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Liver diseases are a significant global health issue, characterized by elevated levels of disorder and death. The substantial impact of ageing on liver diseases and their prognosis is evident. Multiple processes are involved in the ageing process, which ultimately leads to functional deterioration of this organ. The process of liver ageing not only renders the liver more susceptible to diseases but also compromises the integrity of other organs due to the liver's critical function in metabolism regulation. A growing body of research suggests that long non-coding RNAs (lncRNAs) play a significant role in the majority of pathophysiological pathways. They regulate gene expression through a variety of interactions with microRNAs (miRNAs), messenger RNAs (mRNAs), DNA, or proteins. LncRNAs exert a major influence on the progression of age-related liver diseases through the regulation of cell proliferation, necrosis, apoptosis, senescence, and metabolic reprogramming. A concise overview of the current understanding of lncRNAs and their potential impact on the development of age-related liver diseases will be provided in this mini-review.
Collapse
Affiliation(s)
- Peyman Kheirandish Zarandi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Cancer Biology Signaling Pathway Interest Group (CBSPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohsen Ghiasi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Zhang T, Ren Z, Mao R, Yi W, Wang B, Yang H, Wang H, Liu Y. LINC00278 and BRG1: A key regulatory axis in male obesity and preadipocyte adipogenesis. Metabolism 2025; 168:156194. [PMID: 40107651 DOI: 10.1016/j.metabol.2025.156194] [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: 11/10/2024] [Revised: 03/09/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Obesity is a significant public health concern directly associated with adipogenesis. Long non-coding RNAs (lncRNAs) have emerged as critical regulators of adipogenesis. However, the roles of sex-specific lncRNAs in adipose tissue are not well comprehended. In this study, we used lncRNA microarrays to profile lncRNAs expression in visceral adipose tissues from obese and lean individuals, identifying LINC00278 as significantly and exclusively expressed in males. Elevated levels of LINC00278 were associated with higher body mass index (BMI) and non-remission after bariatric surgery in individuals with obesity. Mechanistic studies further revealed that METTL14 regulates the m6A methylation of LINC00278, which in turn binds with BRG1, activating the PPAR-γ2 pathway and promoting adipogenesis. Additionally, adipose-specific LINC00278 knock-in in C57BL/6 J mice resulted in adipocyte enlargement, increased body weight, higher body fat percentage, and impaired glucose metabolism. Treatment with the BRG1 inhibitor, BRM/BRG1 ATP Inhibitor-1, significantly alleviated the obesity phenotype in these mice. Our findings highlight the critical role of LINC00278 in male adipogenesis, suggesting that targeting the LINC00278-BRG1 axis could be a potential therapeutic strategy for managing obesity and related metabolic disorders in males.
Collapse
Affiliation(s)
- Tongtong Zhang
- Obesity and Metabolism Medicine-Engineering Integration Laboratory, Department of General Surgery, The Third People's Hospital of Chengdu, Chengdu, China; Medical Research Center, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China.
| | - Zhengyun Ren
- College of Medicine, Southwest Jiaotong University, Chengdu, China; Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Rui Mao
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Yi
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Bin Wang
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Huawu Yang
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Haibo Wang
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Yanjun Liu
- Obesity and Metabolism Medicine-Engineering Integration Laboratory, Department of General Surgery, The Third People's Hospital of Chengdu, Chengdu, China; Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China.
| |
Collapse
|
3
|
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
|
4
|
Uusi-Mäkelä J, Kauppinen M, Seppälä J, Jaatinen S, Ryback B, Rantapero T, Rodriguez-Martinez A, Nykter M, Rautajoki KJ. Tumor-associated long non-coding RNAs show variable expression across diffuse gliomas and effect on cell growth upon silencing in glioblastoma. Sci Rep 2025; 15:16220. [PMID: 40346283 PMCID: PMC12064817 DOI: 10.1038/s41598-025-99984-9] [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: 08/06/2024] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
Long noncoding RNAs (lncRNAs) have been recently recognized as critical components of cancer biology linked to oncogenic processes. Certain lncRNAs are known to act as oncogenes, and the disease-specific expression of many lncRNAs makes them informative biomarkers. We identified 22 uncharacterized lncRNAs from RNA-seq data of 169 glioblastoma (GBM) tumor samples sequenced by The Cancer Genome Atlas (TCGA) consortium and studied their expression in TCGA diffuse glioma cohort including also IDH-mutant astrocytomas and oligodendrogliomas as well as in normal brain samples from the Genotype-Tissue Expression cohort. All of the 22 lncRNAs were clearly upregulated in diffuse gliomas samples compared to the normal brain. Interestingly, 20 (91%) of these lncRNAs had significant expression differences between tumor grades and/or entities, and 14 (64%) were associated with overall patient survival. All 22 lncRNAs were expressed in at least one of the studied GBM cell lines and 10 (45%) were expressed in all four. When six of the lncRNAs were silenced in the SNB19 GBM cell line, the knock-down was associated with reduced growth and colony formation for three lncRNAs: TCONS_l2_00001282, lnc-GBMT-6, and lnc-NBN-1. In conclusion, the studied lncRNAs are associated with survival in patients with diffuse glioma and have functional relevance in GBM.
Collapse
Affiliation(s)
- Joonas Uusi-Mäkelä
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Maria Kauppinen
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- IT Management, Helsinki University Hospital, Helsinki, Finland
| | - Janne Seppälä
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Serafiina Jaatinen
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Birgitta Ryback
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Tommi Rantapero
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Alejandra Rodriguez-Martinez
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Kirsi J Rautajoki
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
5
|
Fujiwara N, Ueno T, Yamazaki T, Hirose T. Architectural RNAs: A class of long noncoding RNAs functioning as scaffolds for membraneless organelles. Biochim Biophys Acta Gen Subj 2025:130815. [PMID: 40348038 DOI: 10.1016/j.bbagen.2025.130815] [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: 01/05/2025] [Revised: 04/25/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
Architectural RNAs (arcRNAs) are long noncoding RNAs that serve as structural scaffolds for membraneless organelles (MLOs), facilitating cellular organization and dynamic responses to stimuli. Acting as blueprints for MLO assembly, arcRNAs recruit specific proteins and nucleic acids to establish and maintain the internal structure of MLOs while coordinating their spatial relationships with other organelles. This organized framework enables precise spatiotemporal regulation, allowing for targeted control of transcription, RNA processing, and cellular responses to stress. Notably, arcRNAs exhibit the "semi-extractable" feature, a property derived from their stable binding to cellular structures, making them partially resistant to conventional RNA extraction methods. This unique feature serves as a useful criterion for identifying novel arcRNAs, providing an opportunity to accelerate research in long noncoding RNAs and deepen our understanding of their functional roles in cellular processes.
Collapse
Affiliation(s)
- Naoko Fujiwara
- Graduate School of Frontier Biosciences, The University of Osaka, Suita 565-0871, Japan
| | - Tsuyoshi Ueno
- Graduate School of Frontier Biosciences, The University of Osaka, Suita 565-0871, Japan
| | - Tomohiro Yamazaki
- Graduate School of Frontier Biosciences, The University of Osaka, Suita 565-0871, Japan
| | - Tetsuro Hirose
- Graduate School of Frontier Biosciences, The University of Osaka, Suita 565-0871, Japan.
| |
Collapse
|
6
|
Zhang L, Li X, Gao H, Chang W, Li P. Gut microbiota-lncRNA/circRNA crosstalk: implications for different diseases. Crit Rev Microbiol 2025; 51:499-513. [PMID: 38967384 DOI: 10.1080/1040841x.2024.2375516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 05/23/2024] [Accepted: 06/26/2024] [Indexed: 07/06/2024]
Abstract
The gut microbiota features an abundance of diverse microorganisms and represents an important component of human physiology and metabolic homeostasis, indicating their roles in a wide array of physiological and pathological processes in the host. Maintaining balance in the gut microbiota is critical for normal functionality as microbial dysbiosis can lead to the occurrence and development of diseases through various mechanisms. Long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) are non-coding RNAs that perform important regulatory functions for many processes. Furthermore, the gut microbiota and lncRNAs/circRNAs are known to interact in a range of both physiological and pathological activities. In this article, we review existing research relevant to the interaction between the gut microbiota and lncRNAs/circRNAs and investigate the role of their crosstalk in the pathogenesis of different diseases. Studies have shown that, the gut microbiota can target lncRNAs ENO1-IT1, BFAL1, and LINC00152 to regulate colorectal cancer development via various signaling pathways. In addition, the gut microbiota can influence mental diseases and lung tumor metastasis by modulating circRNAs such as circNF1-419, circ_0001239, circHIPK2 and mmu_circ_0000730. These findings provide a theoretical basis for disease prevention and treatment and suggest that gut microbiota-lncRNA/circRNA crosstalk has high clinical value.
Collapse
Affiliation(s)
- Lei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Xin Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Huijuan Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Wenguang Chang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
7
|
Mainkar G, Ghiringhelli M, Zangi L. The Potential of RNA Therapeutics in Treating Cardiovascular Disease. Drugs 2025; 85:659-676. [PMID: 40175855 DOI: 10.1007/s40265-025-02173-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2025] [Indexed: 04/04/2025]
Abstract
Despite significant advances in cardiology over the past few decades, cardiovascular diseases (CVDs) remain the leading cause of global mortality and morbidity. This underscores the need for novel therapeutic interventions that go beyond symptom management to address the underlying causal mechanisms of CVDs. RNA-based therapeutics represent a new class of drugs capable of regulating specific genetic and molecular pathways, positioning them as strong candidates for targeting the root causes of a wide range of diseases. Moreover, owing to the vast diversity in RNA form and function, these molecules can be utilized to induce changes at different levels of gene expression regulation, making them suitable for a broad array of medical applications, even within a single disease context. Several RNA-based therapies are currently being investigated for their potential to address various CVD pathologies. These include treatments aimed at promoting cardiac revascularization and regeneration, preventing cardiomyocyte apoptosis, reducing harmful circulating cholesterols and fats, lowering blood pressure, reversing cardiac fibrosis and remodeling, and correcting the genetic basis of inherited CVDs. In this review, we discuss the current landscape of RNA therapeutics for CVDs, with an emphasis on their classifications, modes of action, advancements in delivery strategies and considerations for their implementation, as well as CVD targets with proven therapeutic potential.
Collapse
Affiliation(s)
- Gayatri Mainkar
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Matteo Ghiringhelli
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lior Zangi
- Icahn School of Medicine at Mount Sinai, Cardiovascular Research Institute, New York, NY, 10029, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
8
|
Wang J, Liu ZX, Huang ZH, Wen J, Rao ZZ. Long non-coding RNA in the regulation of cell death in hepatocellular carcinoma. World J Clin Oncol 2025; 16:104061. [PMID: 40290684 PMCID: PMC12019274 DOI: 10.5306/wjco.v16.i4.104061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/02/2025] [Accepted: 02/25/2025] [Indexed: 03/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the predominant form of primary liver cancer, accounting for 90% of all cases. Currently, early diagnosis of HCC can be achieved through serum alpha-fetoprotein detection, B-ultrasound, and computed tomography scanning; however, their specificity and sensitivity are suboptimal. Despite significant advancements in HCC biomarker detection, the prognosis for patients with HCC remains unfavorable due to tumor heterogeneity and limited understanding of its pathogenesis. Therefore, it is crucial to explore more sensitive HCC biomarkers for improved diagnosis, monitoring, and management of the disease. Long non-coding RNA (lncRNA) serves as an auxiliary carrier of genetic information and also plays diverse intricate regulatory roles that greatly contribute to genome complexity. Moreover, investigating gene expression regulation networks from the perspective of lncRNA may provide insights into the diagnosis and prognosis of HCC. We searched the PubMed database for literature, comprehensively classified regulated cell death mechanisms and systematically reviewed research progress on lncRNA-mediated cell death pathways in HCC cells. Furthermore, we prospectively summarize its potential implications in diagnosing and treating HCC.
Collapse
Affiliation(s)
- Jiang Wang
- Children Medical Center, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Zi-Xuan Liu
- Children Medical Center, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Zhi-Hong Huang
- Children Medical Center, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Jie Wen
- Department of Pediatric Orthopedics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Zhou-Zhou Rao
- Department of Physiology, Hunan Normal University School of Medicine, Changsha 410003, Hunan Province, China
| |
Collapse
|
9
|
Coltman BL, Motheramgari K, Tatto N, Gasser B. Identification and functional analysis of growth rate associated long non-coding RNAs in Komagataella phaffii. Comput Struct Biotechnol J 2025; 27:1693-1705. [PMID: 40352477 PMCID: PMC12063151 DOI: 10.1016/j.csbj.2025.04.028] [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: 02/11/2025] [Revised: 04/19/2025] [Accepted: 04/21/2025] [Indexed: 05/14/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) are a regulatory feature that have been reported to operate on both transcriptional and translational levels. With DNA-based prediction still limited, lncRNAs are most reliably identified through genome-guided mapping of RNA-Seq data. Reports of lncRNAs in yeast have been increasing in recent years and changes in their expression levels have often been associated with stressful conditions. As the transition to near zero-growth conditions likely imposes stress, we used RNA-Seq data from the non-conventional, biotechnologically established yeast Komagataella phaffii, cultivated in glucose-limited retentostats, to identify the expression of lncRNAs. Using an adapted bioinformatics pipeline, we identified 168 mostly novel lncRNAs from the K. phaffii retentostat RNA-Seq data, 36 of which demonstrate likely growth-associated expression changes. lncRNA expression levels were associated to that of possible interaction partners, in both cis and trans, suggesting potential roles in regulatory adaptations. Our analysis indicates that lncRNAs likely contribute to how K. phaffii responds to changing environmental conditions, as exemplified here by the adaptation to extremely slow growth.
Collapse
Affiliation(s)
- Benjamin Luke Coltman
- CD-Laboratory for Growth-decoupled Protein Production in Yeast at Department of Biotechnology, BOKU University, Vienna, Austria
- Department of Biotechnology and Food Science, Institute of Microbiology and Microbial Biotechnology, BOKU University, Vienna, Austria
- Division of Microbial Ecology, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | | | - Nadine Tatto
- Department of Biotechnology and Food Science, Institute of Microbiology and Microbial Biotechnology, BOKU University, Vienna, Austria
- Austrian Centre of Industrial Biotechnology (acib), Vienna, Austria
- Vienna Biocenter Core Facilities GmbH (VBCF), Next Generation Sequencing, Vienna, Austria
| | - Brigitte Gasser
- CD-Laboratory for Growth-decoupled Protein Production in Yeast at Department of Biotechnology, BOKU University, Vienna, Austria
- Department of Biotechnology and Food Science, Institute of Microbiology and Microbial Biotechnology, BOKU University, Vienna, Austria
- Austrian Centre of Industrial Biotechnology (acib), Vienna, Austria
| |
Collapse
|
10
|
Jiang Y, Saeed TN, Alfarttoosi KH, Bishoyi AK, Rekha MM, Kundlas M, Jain B, Rizaev J, Taher WM, Alwan M, Jawad MJ, Ali Al-Nuaimi AM. The intersection of ferroptosis and non-coding RNAs: a novel approach to ovarian cancer. Eur J Med Res 2025; 30:300. [PMID: 40247379 PMCID: PMC12007203 DOI: 10.1186/s40001-025-02559-7] [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: 02/08/2025] [Accepted: 04/06/2025] [Indexed: 04/19/2025] Open
Abstract
Understanding the core principles of ovarian cancer has been significantly improved through the exploration of Ferroptosis, a type of cell death triggered by iron that leads to an increase in lipid peroxides. Current research has shed light on the critical functions of non-coding RNAs, such as circRNAs, lncRNAs, and miRNAs, in regulating ferroptosis in ovarian cancer. The aim of this paper is to comprehensively analyze how ncRNAs influence the development of ferroptosis in ovarian cancer cells. In-depth exploration is undertaken to understand the intricate ways in which ncRNAs regulate essential elements of ferroptosis, including iron management and lipid peroxidation levels. We also investigate their significant involvement in the progression of this type of cellular demise. It should be emphasized that ncRNAs can impact the synthesis of crucial proteins, such as GPX4, a key contributor to the cellular defense against oxidation, and ACSL4, involved in lipid formation. In addition, we examine the correlation between ncRNAs and well-known pathways associated with oxidative stress and cell death. The consequences of these discoveries are noteworthy, since focusing on particular ncRNAs could potentially render ovarian cancer cells more vulnerable to ferroptosis, effectively combating drug resistance problems. This discussion highlights the growing significance of ncRNAs in governing ferroptosis and their potential as useful biomarkers and treatment targets for ovarian cancer. We intend to promote additional research into the involvement of ncRNAs in controlling ferroptosis, based on current findings, with the ultimate goal of informing targeted therapeutic strategies and improving long-term treatment outcomes for individuals suffering from OC.
Collapse
Affiliation(s)
- Youyi Jiang
- School of Civil Engineering, Chongqing Jiaotong University, Chongqing, China
| | - Tamara Nazar Saeed
- Department of Medical Laboratory Technics, College of Health and Medical Technology, Alnoor University, Mosul, Iraq.
| | | | - Ashok Kumar Bishoyi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, 360003, Gujarat, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Mayank Kundlas
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Bhavik Jain
- Chitkara Centre for Research and Development, Chitkara University, Baddi, Himachal Pradesh, 174103, India
| | - Jasur Rizaev
- Department of Public Health and Healthcare Management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | |
Collapse
|
11
|
Chen Y, Wang J, Gu L, Chen H, Gai Z, Hu R, Qing B, Yuan Y, Xia Z. lncRNA NR_146969 promotes the progression of lung adenocarcinoma. Exp Cell Res 2025; 447:114535. [PMID: 40147711 DOI: 10.1016/j.yexcr.2025.114535] [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: 02/15/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Emerging research suggests that dysregulation of long non-coding RNAs (lncRNAs) is closely linked to the onset and progression of cancer. In this study, we used lncRNA array technology to identify differentially expressed lncRNAs in lung adenocarcinoma patients and normal lung tissues. The study further explored the clinical significance and function of candidate lncRNAs in lung adenocarcinoma (LUAD). The results showed that lncRNA NR_146969 was upregulated in LAUD specimens and was associated with lymph node metastasis and clinical staging in LUAD patients. METHODS The biological functions of lncRNA NR_146969 were observed using CCK-8, colony formation, transwell assay and xenograft tumor model. Explore the potential mechanism of action of lncRNA NR_146969 by FISH, dual luciferase reporter assay and recovery assay. RESULTS Overall, lncRNA NR_146969 plays an oncogenic role in LUAD. Mechanically, lncRNA NR_146969 targets SLC6A14 via miR-26a-1-3p, leading to phosphorylation of the AKT/mTOR pathway, which promotes LUAD growth and metastasis. CONCLUSION Therefore, targeting lncRNA NR_146969 may provide a new therapeutic strategy for LUAD.
Collapse
Affiliation(s)
- Ying Chen
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Hunan Provincial Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Juan Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Hunan Provincial Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Linguo Gu
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Hunan Provincial Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Hongzuo Chen
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Hunan Provincial Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Zhengling Gai
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Hunan Provincial Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Rui Hu
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Hunan Provincial Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Bei Qing
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Hunan Provincial Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Yunchang Yuan
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Hunan Provincial Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Zhenkun Xia
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Hunan Provincial Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
12
|
Dakal TC, Xu C, Kumar A. Advanced computational tools, artificial intelligence and machine-learning approaches in gut microbiota and biomarker identification. FRONTIERS IN MEDICAL TECHNOLOGY 2025; 6:1434799. [PMID: 40303946 PMCID: PMC12037385 DOI: 10.3389/fmedt.2024.1434799] [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: 07/23/2024] [Accepted: 10/16/2024] [Indexed: 05/02/2025] Open
Abstract
The microbiome of the gut is a complex ecosystem that contains a wide variety of microbial species and functional capabilities. The microbiome has a significant impact on health and disease by affecting endocrinology, physiology, and neurology. It can change the progression of certain diseases and enhance treatment responses and tolerance. The gut microbiota plays a pivotal role in human health, influencing a wide range of physiological processes. Recent advances in computational tools and artificial intelligence (AI) have revolutionized the study of gut microbiota, enabling the identification of biomarkers that are critical for diagnosing and treating various diseases. This review hunts through the cutting-edge computational methodologies that integrate multi-omics data-such as metagenomics, metaproteomics, and metabolomics-providing a comprehensive understanding of the gut microbiome's composition and function. Additionally, machine learning (ML) approaches, including deep learning and network-based methods, are explored for their ability to uncover complex patterns within microbiome data, offering unprecedented insights into microbial interactions and their link to host health. By highlighting the synergy between traditional bioinformatics tools and advanced AI techniques, this review underscores the potential of these approaches in enhancing biomarker discovery and developing personalized therapeutic strategies. The convergence of computational advancements and microbiome research marks a significant step forward in precision medicine, paving the way for novel diagnostics and treatments tailored to individual microbiome profiles. Investigators have the ability to discover connections between the composition of microorganisms, the expression of genes, and the profiles of metabolites. Individual reactions to medicines that target gut microbes can be predicted by models driven by artificial intelligence. It is possible to obtain personalized and precision medicine by first gaining an understanding of the impact that the gut microbiota has on the development of disease. The application of machine learning allows for the customization of treatments to the specific microbial environment of an individual.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, India
| | - Caiming Xu
- Beckman Research Institute of City of Hope, Monrovia, CA, United States
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Abhishek Kumar
- Manipal Academy of Higher Education (MAHE), Manipal, India
- Institute of Bioinformatics, International Technology Park, Bangalore, India
| |
Collapse
|
13
|
Mouhou E, Genty F, El M'selmi W, Chouali H, Zagury JF, Le Clerc S, Proudhon C, Noirel J. High tissue specificity of lncRNAs maximises the prediction of tissue of origin of circulating DNA. Sci Rep 2025; 15:12941. [PMID: 40234550 PMCID: PMC12000428 DOI: 10.1038/s41598-024-82393-9] [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: 03/21/2024] [Accepted: 12/05/2024] [Indexed: 04/17/2025] Open
Abstract
Several studies have made it possible to envision a translational application of plasma DNA sequencing in cancer diagnosis and monitoring. However, the extremely low concentration of circulating tumour DNA (ctDNA) fragments among the total cell-free DNA (cfDNA) remains a formidable challenge to overcome and statistical models have yet to be improved enough to become of practical use. In this study, we set about appraising the predictive value of a variety of binary classification models based on cfDNA sequencing using fragmentation features extracted around transcription start sites (TSSs). We investigated (1) features summarising mapped fragment density around each TSS, (2) long non-coding RNA (lncRNA) genes versus coding genes and (3) selection criteria to generate gene classes to be assigned by the model. Given that, in healthy samples, most of the cfDNA comes from lymphomyeloid lineages, we could identify the model parametrisation with the best accuracy in those lineages using publicly available datasets of healthy patients' cfDNA. Our results show that (1) the way tissue-specific gene classes are defined matters more than what fragmentation features are included, and (2) in particular, lncRNAs are more tissue specific than coding genes and stand out in terms of both sensitivity and specificity in our results.
Collapse
Affiliation(s)
- Elyas Mouhou
- Laboratoire GBCM (EA7528), Conservatoire national des arts et métiers (CNAM), Paris, France
| | - Fabien Genty
- Infotel Conseil, 13, rue Madeleine-Michelis, Neuilly-sur-Seine, France
| | | | - Hanae Chouali
- BioinfOmics, GenoToul Bioinformatics facility, Université Fédérale de Toulouse, INRAE, Castanet-Tolosan, France
- MIAT, Université Fédérale de Toulouse, INRAE, Castanet-Tolosan, France
| | - Jean-François Zagury
- Laboratoire GBCM (EA7528), Conservatoire national des arts et métiers (CNAM), Paris, France
| | - Sigrid Le Clerc
- Laboratoire GBCM (EA7528), Conservatoire national des arts et métiers (CNAM), Paris, France
| | | | - Josselin Noirel
- Laboratoire GBCM (EA7528), Conservatoire national des arts et métiers (CNAM), Paris, France.
| |
Collapse
|
14
|
Ribeiro AL, Dallagiovanna B. The Role of Long Non-Coding RNAs in Human Endoderm Differentiation. Noncoding RNA 2025; 11:29. [PMID: 40278506 PMCID: PMC12029278 DOI: 10.3390/ncrna11020029] [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: 02/26/2025] [Revised: 04/03/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
The human genome sequencing revealed a vast complexity of transcripts, with over 80% of the genome being transcribed into non-coding RNAs. In particular, long non-coding RNAs (lncRNAs) have emerged as critical regulators of various cellular processes, including embryonic development and stem cell differentiation. Despite extensive efforts to identify and characterize lncRNAs, defining their mechanisms of action in state-specific cellular contexts remains a significant challenge. Only recently has the involvement of lncRNAs in human endoderm differentiation of pluripotent stem cells begun to be addressed, creating an opportunity to explore the mechanisms by which lncRNAs exert their functions in germ layer formation, lineage specification, and commitment. This review summarizes current findings on the roles of lncRNAs in endoderm differentiation, highlighting the functional mechanisms and regulatory aspects underlying their involvement in cell fate decisions leading to endoderm development. The key lncRNAs implicated in endoderm differentiation are discussed, along with their interaction with transcription factors and RNA-binding proteins and modulation of signaling pathways essential for endoderm development. Gaining insight into the regulatory roles of lncRNAs in endoderm differentiation enhances the understanding of developmental biology and provides a foundation for discovering novel lncRNAs involved in cell fate determination.
Collapse
Affiliation(s)
| | - Bruno Dallagiovanna
- Stem Cells Basic Biology Laboratory, Carlos Chagas Institute—FIOCRUZ/PR, Curitiba 81350-010, Brazil;
| |
Collapse
|
15
|
Tataranu LG. Liquid Biopsy as a Diagnostic and Monitoring Tool in Glioblastoma. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:716. [PMID: 40283007 PMCID: PMC12028463 DOI: 10.3390/medicina61040716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 03/26/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025]
Abstract
Glioblastoma (GBM) is the most prevalent and aggressive primary central nervous system (CNS) tumor in adults. GBMs exhibit genetic and epigenetic heterogeneity, posing difficulties in surveillance and being associated with high rates of recurrence and mortality. Nevertheless, due to the high infiltrating ability of glioblastoma cells, and regardless of the considerable progress made in radiotherapeutic, chemotherapeutic, and surgical protocols, the treatment of GBM is still inefficient. Conventional diagnostic approaches, such as neuroimaging techniques and tissue biopsies, which are invasive maneuvers, present certain challenges and limitations in providing real-time information, and are incapable of differentiating pseudo-progression related to treatment from real tumor progression. Liquid biopsy, the analysis of biomarkers such as nucleic acids (DNA/RNA), circulating tumor cells (CTCs), extracellular vesicles (EVs), or tumor-educated platelets (TEPs) that are present in body fluids, provides a minimally invasive and dynamic method of diagnosis and continuous monitoring for GBM. It represents a new preferred approach that enables a superior manner to obtain data on possible tumor risk, prognosis, and recurrence assessment. This article is a literature review that aims to provide updated information about GBM biomarkers in body fluids and to analyze their clinical efficiency.
Collapse
Affiliation(s)
- Ligia Gabriela Tataranu
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Neurosurgery, Bagdasar-Arseni Emergency Clinical Hospital, 041915 Bucharest, Romania
| |
Collapse
|
16
|
Wingfield JL, Puthanveettil SV. Decoding the complex journeys of RNAs along neurons. Nucleic Acids Res 2025; 53:gkaf293. [PMID: 40243060 PMCID: PMC12004114 DOI: 10.1093/nar/gkaf293] [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: 12/29/2024] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Neurons are highly polarized, specialized cells that must overcome immense challenges to ensure the health and survival of the organism in which they reside. They can spread over meters and persist for decades yet communicate at sub-millisecond and millimeter scales. Thus, neurons require extreme levels of spatial-temporal control. Neurons employ molecular motors to transport coding and noncoding RNAs to distal synapses. Intracellular trafficking of RNAs enables neurons to locally regulate protein synthesis and synaptic activity. The way in which RNAs get loaded onto molecular motors and transported to their target locations, particularly following synaptic plasticity, is explored below.
Collapse
Affiliation(s)
- Jenna L Wingfield
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, United States
| | - Sathyanarayanan V Puthanveettil
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, United States
| |
Collapse
|
17
|
Chen P, Gong Q, Wang H, Wang C, Wang W, Wu J, Wu Z, Wang L. Analgesic Mechanism of Emodin in Neuropathic Pain Through Inhibiting P2X4 Purinoceptor Signaling. Mol Neurobiol 2025:10.1007/s12035-025-04906-5. [PMID: 40195215 DOI: 10.1007/s12035-025-04906-5] [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: 11/30/2024] [Accepted: 03/31/2025] [Indexed: 04/09/2025]
Abstract
Neuropathic pain (NeP) is a most intractable health problem due to its unsatisfactory treatment effect. Emodin, a natural anthraquinone derivative extracted from Rheum palmatum and Polygonam cuspidatum, exhibits the analgesic effects in various NeP models. However, the underlying mechanisms remain elusive. This study employed whole transcriptome sequencing and metabolomics to elucidate emodin's analgesic mechanism in the spinal cord of chronic constriction injury (CCI) rats. Fifteen-day emodin treatment reversed hyperalgesia and deficit of sciatic nerve function induced by CCI and significantly decreased the concentrations of TNF-α, IL- 1β, IL- 6, IL- 18, and BDNF in the spinal cord of the CCI rats. Transcriptome sequencing revealed altered expression of 85 mRNAs in the spinal cord of emodin-treated and CCI rats, with 53 mRNAs upregulated and 32 mRNAs downregulated. Notably, seven genes (P2RX4, CXCL10, ALOX5, SCN4 A, AURKB, AQP9) overlapped with established NeP targets. Untargeted metabolomic analyses identified 67 significantly altered metabolites (46 upregulated, 32 downregulated) in the spinal cord upon emodin treatment. Integrative analysis highlighted shared pathways between differentially expressed genes and metabolites, including arachidonic acid metabolism, cAMP signaling pathway, and Fc epsilon RI signaling pathway. Western blot and immunofluorescent staining further proved the decreased expression of IBA1, P2X4R, p38 MAPK, p-p38 MAPK, NF-κB, p-NF-κB, and TNF-α, IL- 1β. In conclusion, this study demonstrated that emodin played the analgesic effect in the CCI rats, possibly through suppression of P2X4 purinoceptor signaling in spinal microglia, suggesting a potential therapeutic target for NeP.
Collapse
Affiliation(s)
- Peng Chen
- Basic Medical School, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China.
| | - Qian Gong
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hao Wang
- Basic Medical School, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Chen Wang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou, China
| | - Wenjing Wang
- Basic Medical School, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Jing Wu
- Basic Medical School, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Zhibing Wu
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Long Wang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
18
|
Le LTT. Long non coding RNA function in epigenetic memory with a particular emphasis on genomic imprinting and X chromosome inactivation. Gene 2025; 943:149290. [PMID: 39880342 DOI: 10.1016/j.gene.2025.149290] [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: 01/14/2024] [Revised: 12/13/2024] [Accepted: 01/25/2025] [Indexed: 01/31/2025]
Abstract
Cells preserve and convey certain gene expression patterns to their progeny through the mechanism called epigenetic memory. Epigenetic memory, encoded by epigenetic markers and components, determines germline inheritance, genomic imprinting, and X chromosome inactivation. First discovered long non coding RNAs were implicated in genomic imprinting and X-inactivation and these two phenomena clearly demonstrate the role of lncRNAs in epigenetic memory regulation. Undoubtedly, lncRNAs are well-suited for regulating genes in close proximity at imprinted loci. Due to prolonged association with the transcription site, lncRNAs are able to guide chromatin modifiers to certain locations, thereby enabling accurate temporal and spatial regulation. Nevertheless, the current state of knowledge regarding lncRNA biology and imprinting processes is still in its nascent phase. Herein, we provide a synopsis of recent scientific advancements to enhance our comprehension of lncRNAs and their functions in epigenetic memory, with a particular emphasis on genomic imprinting and X chromosome inactivation, thus gaining a deeper understanding of the role of lncRNAs in epigenetic regulatory networks.
Collapse
Affiliation(s)
- Linh T T Le
- Faculty of Biotechnology, Ho Chi Minh City Open University, Ho Chi Minh City 700000 Viet Nam
| |
Collapse
|
19
|
Riquier S, Carthy S, Hughes GM, Touzalin F, Haerty W, Huang Z, Teeling EC. RNA-Seq analysis reveals the long noncoding RNAs associated with immunity in wild Myotis myotis bats. BMC Genomics 2025; 26:345. [PMID: 40188093 PMCID: PMC11972528 DOI: 10.1186/s12864-025-11485-1] [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: 01/30/2025] [Accepted: 03/13/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Bats possess a uniquely adapted immune system that enables them to live with viral infections without the expected maladies. The molecular basis and regulation of bats' immune response is still not fully understood. Long non-coding RNAs (lncRNAs) represent an emerging class of molecules with critical regulatory roles in multiple biological processes, including immunity. We hypothesise that lncRNA-based regulation in bats may enable them to limit disease and live with viral pathogens. RESULTS We developed a lncRNA prediction pipeline to annotate the long non-coding transcriptome across multiple bat tissues and at the population level. Characterisation of our lncRNA dataset based on 100 blood transcriptomes from wild Myotis myotis bats revealed lower and more tissue-specific expression compared with coding genes, reduced GC content and shorter length distributions, consistent with lncRNA profiles observed in other species. Using WGCNA network analyses and gene ontology, we identified two mRNA-lncRNA co-expression modules in Myotis myotis associated with distinct immune response: one linked to T-cell activation and vial processes, and the other to inflammation. From these immune-related lncRNAs, we selected four candidates with high translational potential for regulating viral infections and inflammation. These include a newly identified lncRNA, BatLnc1, with potential antiviral functions; the M. myotis ortholog of TUG1, implicated in viral-host interactions; and well-known lncRNAs MALAT1 and NEAT1, recognised for their roles in inflammatory regulation. CONCLUSIONS We conducted the first ab initio prediction of lncRNAs in a non-model bat species, the wild-caught M. myotis. Our network analysis revealed significant variation in immune status among a subset of individuals, potentially due to pathogenic conditions. From these variations, we identified lncRNAs most likely associated with immune response in bats. This initial exploration lays the groundwork for future experimental validations of lncRNA functions, offering promising insights into their role in bat immunity.
Collapse
Affiliation(s)
- Sebastien Riquier
- School of Biology and Environmental Science, University College Dublin, Belfield, Dublin, Ireland
| | - Samuel Carthy
- School of Biology and Environmental Science, University College Dublin, Belfield, Dublin, Ireland
| | - Graham M Hughes
- School of Biology and Environmental Science, University College Dublin, Belfield, Dublin, Ireland
| | - Frederic Touzalin
- School of Biology and Environmental Science, University College Dublin, Belfield, Dublin, Ireland
- School of Biodiversity, One Health & Veterinary Medicine, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, UK
| | - Wilfried Haerty
- Earlham Institute, Norwich, UK
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Zixia Huang
- School of Biology and Environmental Science, University College Dublin, Belfield, Dublin, Ireland
| | - Emma C Teeling
- School of Biology and Environmental Science, University College Dublin, Belfield, Dublin, Ireland.
| |
Collapse
|
20
|
Sharma NK, Mishra DC, Kumar B, Srivastava S, Chaturvedi KK, Singh AK, Madival SD, Budhlakoti N, Jha GK. Beyond the genome: unveiling tissue-specific non-coding RNAs in clove ( Syzygium aromaticum L.). 3 Biotech 2025; 15:81. [PMID: 40071125 PMCID: PMC11891123 DOI: 10.1007/s13205-025-04251-3] [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: 08/30/2024] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Clove (Syzygium aromaticum), valued for its role in food preservation and medicine, has recently drawn research interest for its noncoding RNAs (ncRNAs). This study discovers 3274 long noncoding RNAs (lncRNAs) and 2404 circular RNAs (circRNAs) from publicly available RNAseq data. We identified the regulation of 834 genes through miRNA-lncRNA-mRNA network interactions. Additionally, 35 lncRNAs were predicted as precursors for 17 microRNAs (miRNAs), highlighting their role in post-transcriptional regulation. Tissue-specific analysis of circRNAs revealed their interaction with 1047 miRNAs and competing for binding sites on 2382 messenger RNAs (mRNAs). These results underscore their involvement in complex regulatory networks. To support further research and development, we developed SaroNcRDb (http://backlin.cabgrid.res.in/saroncrdb/), a web resource providing detailed insights into the types, chromosomal locations, tissue distributions, and interactions of identified ncRNAs. The findings pave the way for future studies to harness the regulatory roles of ncRNAs in improving Clove's agronomic traits and secondary metabolite production.
Collapse
Affiliation(s)
- Nitesh Kumar Sharma
- Division of Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, New Delhi, 110012 India
- The Graduate School, ICAR-Indian Agricultural Research Institute, New Delhi, 110012 India
| | - Dwijesh Chandra Mishra
- Division of Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, New Delhi, 110012 India
- Department of Environmental and Public Health, College of Medicine, University of Cincinnati, Cincinnati, OH 45221 USA
| | - Baibhav Kumar
- Division of Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, New Delhi, 110012 India
- The Graduate School, ICAR-Indian Agricultural Research Institute, New Delhi, 110012 India
| | - Sudhir Srivastava
- Division of Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, New Delhi, 110012 India
| | - Krishna Kumar Chaturvedi
- Division of Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, New Delhi, 110012 India
| | - Awani Kumar Singh
- Centre for Protected Cultivation Technology, ICAR-Indian Agricultural Research Institute, New Delhi, 110012 India
| | - Sharanbasappa D. Madival
- Division of Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, New Delhi, 110012 India
- The Graduate School, ICAR-Indian Agricultural Research Institute, New Delhi, 110012 India
| | - Neeraj Budhlakoti
- Division of Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, New Delhi, 110012 India
| | - Girish Kumar Jha
- Division of Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, New Delhi, 110012 India
| |
Collapse
|
21
|
Yang J. Unveiling the multifaceted roles of long non-coding RNA CTBP1-DT in human diseases: Special attention to its microprotein-encoding potential. Pathol Res Pract 2025; 268:155870. [PMID: 40020329 DOI: 10.1016/j.prp.2025.155870] [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: 11/18/2024] [Revised: 01/30/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
C-terminal binding protein 1 divergent transcript (CTBP1-DT) is a novel long non-coding RNA (lncRNA) located on human chromosome 4p16.3. Numerous studies have shown that CTBP1-DT plays a critical regulatory role in various human malignancies and non-malignant diseases. In several cancers, the expression of CTBP1-DT is upregulated, closely associated with the risk of 12 types of cancer, and strongly correlated with the clinical pathological features and poor prognosis of 10 of these cancers. Mechanistically, CTBP1-DT is stimulated by the transcription factors ETV5 and Sp1, or methylated by YTHDC1. By competitively inhibiting 12 microRNAs, it activates 3 signaling pathways that influence malignant behaviors of tumor cells, including proliferation, apoptosis, cell cycle arrest, migration, invasion, immune evasion, and chemoresistance. Importantly, it also encodes the microprotein DNA damage up-regulated protein (DDUP), which mediates cisplatin resistance through sustained response to DNA damage signals. Furthermore, CTBP1-DT has been implicated in the progression of non-malignant diseases such as diabetes and related conditions, cardiovascular diseases, and osteoarthritis. This review summarizes the latest research on the RNA and protein functions of CTBP1-DT in human diseases, outlines various molecular regulatory networks centered around CTBP1-DT, and discusses the opportunities and challenges of its clinical applications.
Collapse
Affiliation(s)
- Jingjie Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Key Laboratory of Cancer Immunology and Biotherapy, Tianjin 300060, China.
| |
Collapse
|
22
|
Bao Y, Luo Y, Zhai H, Lu J, Zhang M, Wang N. Long noncoding RNA MIAT regulates VSMC migration by sponging miR-326. Sci Prog 2025; 108:368504251335854. [PMID: 40233150 PMCID: PMC12035257 DOI: 10.1177/00368504251335854] [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] [Indexed: 04/17/2025]
Abstract
ObjectiveThe current study aimed to investigate the role of the myocardial infarction-associated transcript (MIAT)/microRNA-326 (miR-326) axis in regulating the migration of vascular smooth muscle cells (VSMCs) during the progression of atherosclerosis (AS).MethodsBioinformatic analysis of MIAT and miR-326 in two AS-related GEO datasets was performed via the online web tool GEO2R. MIAT and miR-326 expression in 46 paired plasma samples and in oxidized low-density lipoprotein (ox-LDL)-treated VSMCs was analysed via RT-qPCR. Western blot analysis was used to determine the expression of monocyte chemotactic protein 1 (MCP-1) after diverse ox-LDL treatments. The correlation between MIAT and miR-326 was analysed by Spearman correlation analysis. Transwell assays were performed to determine the changes in migration after different MIAT or miR-326 interventions. RNA-fluorescence in situ hybridization (FISH) assays were performed to determine the subcellular localization of MIAT and miR-326. The targeted binding effect between MIAT and miR-326 was confirmed via a luciferase assay.ResultsMIAT was upregulated and miR-326 was downregulated in 46 plasma samples from patients with AS compared with those from patients without AS (non-AS). A negative correlation was found between MIAT and miR-326 (r = - 0.6591, P < 0.0001). The expression of MIAT in plaque samples from advanced AS patients was markedly greater than that in plaque samples from early AS patients according to the GEO dataset GSE28829 (P < 0.0001). The expression of miR-326 in platelet samples from patients with first acute myocardial infarction (FAMI) was significantly lower than that in healthy controls (P = 0.0034). MCP-1 was upregulated in ox-LDL-treated VSMCs. MIAT knockdown by specific MIAT small interfering RNAs (siRNAs) suppressed VSMC migration. Upregulation of miR-326 by transfection of miR-326 mimics also inhibited VSMC migration. Dual luciferase assays indicated that miR-326 targets MIAT. The upregulation of MIAT increased the migration of VSMCs. However, this effect was attenuated by a miR-326 mimic.ConclusionsMIAT was upregulated and miR-326 was downregulated in AS plasma and in ox-LDL-treated VSMCs. MIAT binds to miR-326 via theoretical miRNA response elements. MIAT promoted migration by sponging miR-326 in ox-LDL-induced VMSCs. The MIAT/miR-326 axis may represent a novel therapeutic target for the treatment of AS, offering potential insights into AS progression and its clinical management.
Collapse
MESH Headings
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Humans
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Cell Movement/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Lipoproteins, LDL/metabolism
- Lipoproteins, LDL/pharmacology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/cytology
- Cells, Cultured
- Male
- Chemokine CCL2/metabolism
- Chemokine CCL2/genetics
Collapse
Affiliation(s)
- Yuxin Bao
- Fourth Department of Orthopaedics, Central Hospital Affiliated to Shenyang Medical College, Shenyang, P. R. China
| | - Yinzhou Luo
- Fourth Department of Orthopaedics, Central Hospital Affiliated to Shenyang Medical College, Shenyang, P. R. China
- Third Department of Orthopaedics, Bazhong Central Hospital, Bazhong, Sichuan, P. R. China
| | - Hanjie Zhai
- Fourth Department of Orthopaedics, Central Hospital Affiliated to Shenyang Medical College, Shenyang, P. R. China
| | - Jie Lu
- Department of Cardiology, Shenyang Fourth People's Hospital, China Medical University, Shenyang,
P. R. China
| | - Man Zhang
- Second Department of Cardiology, Central Hospital Affiliated to Shenyang Medical College, Shenyang, P. R. China
| | - Ningning Wang
- Second Department of Cardiology, Central Hospital Affiliated to Shenyang Medical College, Shenyang, P. R. China
- Health Center, Central Hospital Affiliated to Shenyang Medical College, Shenyang, Liaoning, P. R. China
| |
Collapse
|
23
|
Yu X, Su N, Luo J, Zhang D, Zhang H, Duan M, Shi N. Long noncoding RNA USP30-AS1 promotes influenza A virus replication by enhancing PHB1 function. Vet Microbiol 2025; 303:110444. [PMID: 40020267 DOI: 10.1016/j.vetmic.2025.110444] [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: 08/27/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/16/2025]
Abstract
Long noncoding RNAs (lncRNAs) are important regulators of gene expression. Although evidence accumulated over the past decade shows that lncRNAs have key roles in the interaction between viruses and hosts, the functions of the majority of differentially expressed lncRNAs in response to viral infections remain uncharacterized so far. In this study, we have identified that USP30 antisense RNA 1 (USP30-AS1), a host antisense lncRNA, is hijacked by influenza A virus (IAV) to assist its replication. We show that USP30-AS1 is IAV-induced via the Janus protein tyrosine kinase-signal transducer and the activator of transcription (JAK-STAT) signaling pathway. Functionally, ectopic expression of USP30-AS1 significantly promotes IAV replication. Conversely, silencing USP30-AS1 suppresses IAV replication. Mechanistically, USP30-AS1 directly binds prohibitin 1 (PHB1) and modulates its protein stability and function. On the one hand, the binding of USP30-AS1 sequesters PHB1 away from the E3 ubiquitin ligase, tripartite motif containing 21 (TRIM21), thereby protecting the protein stability of PHB1. On the other hand, USP30-AS1 serves as a molecular scaffold for enhancing the interaction between PHB1 and interferon regulatory factor 3 (IRF3), which in turn impedes the nuclear import of IRF3. Therefore, our data unveil an important role of USP30-AS1 in promoting viral replication by modulating PHB1 stability and functions, providing a new insight into the role of lncRNAs in the interplay between IAV and host.
Collapse
Affiliation(s)
- Xiuhua Yu
- Department of Pediatric Respiration, Children's Medical Center, The First Hospital of Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Ning Su
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Jinna Luo
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Daining Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China
| | - Hansi Zhang
- College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Ming Duan
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China.
| | - Ning Shi
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
24
|
Xu M, Gan D, Zhang X, He X, Wu RX, Yin Y, Jin R, Li L, Tan Y, Chen F, Li X, Tian B. SLC30A4-AS1 Mediates the Senescence of Periodontal Ligament Stem Cells in Inflammatory Environments via the Alternative Splicing of TP53BP1. Cell Prolif 2025; 58:e13778. [PMID: 39572253 PMCID: PMC11969240 DOI: 10.1111/cpr.13778] [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: 09/03/2024] [Revised: 10/25/2024] [Accepted: 11/09/2024] [Indexed: 04/05/2025] Open
Abstract
Periodontal ligament stem cells (PDLSCs) are key cells that suppress periodontal damage during both the progression and recovery stages of periodontitis. Although substantial evidence has demonstrated that incubation under an inflammatory condition may accelerate senescence of PDLSCs, whether cellular senescence in response to inflammatory incubation contributes to cell dysfunction remain unexplored. In this study, we first observed inflammation-caused PDLSC senescence in periodontitis based on comparisons of matched patients, and this cellular senescence was demonstrated in healthy cells that were subjected to inflammatory conditions. We subsequently designed further experiments to investigate the possible mechanism underlying inflammation-induced PDLSC senescence with a particular focus on the role of long noncoding RNAs (lncRNAs). LncRNA microarray analysis and functional gain/loss studies revealed SLC30A4-AS1 as a regulator of inflammation-mediated PDLSC senescence. By full-length transcriptome sequencing, we found that SLC30A4-AS1 interacted with SRSF3 to affect the alternative splicing (AS) of TP53BP1 and alter the expression of TP53BP1-204. Further functional studies showed that decreased expression of TP53BP1-204 reversed PDLSC senescence, and SLC30A4-AS1 overexpression-induced PDLSC senescence was abolished by TP53BP1-204 knockdown. Our data suggest for the first time that SLC30A4-AS1 plays a key role in regulating PDLSC senescence in inflammatory environments by modulating the AS of TP53BP1.
Collapse
Affiliation(s)
- Mei Xu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Dian Gan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Xi‐Yu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Xiao‐Tao He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Rui Xin Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Yuan Yin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Rui Jin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Lin Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Yu‐Jie Tan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Fa‐Ming Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Xuan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Bei‐Min Tian
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
25
|
Lisi M, Santini T, D'Andrea T, Salvatori B, Setti A, Paiardini A, Nutarelli S, Nicoletti C, Pellegrini F, Fucile S, Bozzoni I, Martone J. SERTM2: a neuroactive player in the world of micropeptides. EMBO Rep 2025; 26:2044-2076. [PMID: 40108405 PMCID: PMC12019361 DOI: 10.1038/s44319-025-00404-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: 08/11/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/22/2025] Open
Abstract
In this study, we analyze the long noncoding RNA, lncMN3, that is predominantly expressed in motor neurons and shows potential coding capabilities. Utilizing custom antibodies, we demonstrate the production of a lncMN3-derived type I transmembrane micropeptide, SERTM2. Patch-clamp experiments performed on both wild-type and SERTM2 knockout motor neurons, differentiated in vitro from mouse embryonic stem cells, show a difference in the resting membrane potential and overall decreased excitability upon SERTM2 depletion. In vivo studies indicate that the absence of the peptide impairs treadmill test performance. At the mechanistic level, we identify a two-pore domain potassium channel, TASK1, known to be a major determinant of the resting membrane potential in motor neurons, as a SERTM2 interactor. Our study characterizes one of the first lncRNA-derived micropeptides involved in neuronal physiology.
Collapse
Affiliation(s)
- Michela Lisi
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
- Center for Life Nano-& Neuro-Science, Fondazione Istituto Italiano di Tecnologia, Rome, Italy
| | - Tiziana Santini
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
- Center for Life Nano-& Neuro-Science, Fondazione Istituto Italiano di Tecnologia, Rome, Italy
| | | | - Beatrice Salvatori
- Center for Life Nano-& Neuro-Science, Fondazione Istituto Italiano di Tecnologia, Rome, Italy
| | - Adriano Setti
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | | | - Sofia Nutarelli
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Carmine Nicoletti
- DAHFMO-Unit of Histology and Medical Embryology, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Flaminia Pellegrini
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Sergio Fucile
- IRCCS Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Irene Bozzoni
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy.
- Center for Life Nano-& Neuro-Science, Fondazione Istituto Italiano di Tecnologia, Rome, Italy.
- Center for Human Technologies, Istituto Italiano di Tecnologia, Genoa, Italy.
| | - Julie Martone
- Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy.
| |
Collapse
|
26
|
Zhang Y, Xu Y, Zhang Y, Wang S, Zhao M. The multiple functions and mechanisms of long non-coding RNAs in regulating breast cancer progression. Front Pharmacol 2025; 16:1559408. [PMID: 40223929 PMCID: PMC11985786 DOI: 10.3389/fphar.2025.1559408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 03/14/2025] [Indexed: 04/15/2025] Open
Abstract
Breast cancer (BC) is a malignant tumor that has the highest morbidity and mortality rates in the female population, and its high tendency to metastasize is the main cause of poor clinical prognosis. Long non-coding RNAs (lncRNAs) have been extensively documented to exhibit aberrant expression in various cancers and influence tumor progression via multiple molecular pathways. These lncRNAs not only modulate numerous aspects of gene expression in cancer cells, such as transcription, translation, and post-translational modifications, but also play a crucial role in the reprogramming of energy metabolism by regulating metabolic regulators, which is particularly significant in advanced BC. This review examines the characteristics and mechanisms of lncRNAs in regulating BC cells, both intracellularly (e.g., cell cycle, autophagy) and extracellularly (e.g., tumor microenvironment). Furthermore, we explore the potential of specific lncRNAs and their regulatory factors as molecular markers and therapeutic targets. Lastly, we summarize the application of lncRNAs in the treatment of advanced BC, aiming to offer novel personalized therapeutic options for patients.
Collapse
Affiliation(s)
- Yongsheng Zhang
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
- Department of Anesthesia and Perioperative Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Yanjiao Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yanping Zhang
- Department of Anesthesia and Perioperative Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Shoushi Wang
- Department of Anesthesia and Perioperative Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Mingqiang Zhao
- Department of Anesthesia and Perioperative Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| |
Collapse
|
27
|
Yang J, Zhang D, Jiang W. Long noncoding RNA as an emerging regulator of endoderm differentiation: progress and perspectives. CELL REGENERATION (LONDON, ENGLAND) 2025; 14:11. [PMID: 40133743 PMCID: PMC11937447 DOI: 10.1186/s13619-025-00230-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/09/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025]
Abstract
Accumulated studies have demonstrated that long noncoding RNAs (lncRNAs) play crucial regulatory roles in diverse biological processes, such as embryonic development and cell differentiation. Comprehensive transcriptome analysis identifies extensive lncRNAs, gradually elucidating their functions across various contexts. Recent studies have highlighted the essential role of lncRNAs in definitive endoderm differentiation, underscoring their importance in early development. In this review, we have analyzed the features of overlapping, proximal, and desert lncRNAs, classified by genomic location, in pluripotent stem cells (PSCs) and the differentiation derivatives. Furthermore, we focus on the endoderm lineage and review the latest advancements in lncRNA identification and their distinct regulatory mechanisms. By consolidating current knowledge, we aim to provide a clearer perspective on how lncRNAs contribute to endoderm differentiation in different manners.
Collapse
Affiliation(s)
- Jie Yang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, 430062, China.
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Wei Jiang
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
28
|
Costa S, La Rocca G, Cavalieri V. Epigenetic Regulation of Chromatin Functions by MicroRNAs and Long Noncoding RNAs and Implications in Human Diseases. Biomedicines 2025; 13:725. [PMID: 40149701 PMCID: PMC11939841 DOI: 10.3390/biomedicines13030725] [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: 02/07/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
The bulk of RNA produced from the genome of complex organisms consists of a very large number of transcripts lacking protein translational potential and collectively known as noncoding RNAs (ncRNAs). Initially thought to be mere products of spurious transcriptional noise, ncRNAs are now universally recognized as pivotal players in cell regulatory networks across a broad spectrum of biological processes. Owing to their critical regulatory roles, ncRNA dysfunction is closely associated with the etiopathogenesis of various human malignancies, including cancer. As such, ncRNAs represent valuable diagnostic biomarkers as well as potential targets for innovative therapeutic intervention. In this review, we focus on microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), the two most extensively studied classes in the field of ncRNA biology. After outlining key concepts of miRNA and lncRNA biogenesis pathways, we examine their multiple roles in mediating epigenetic regulation of gene expression and chromatin organization. Finally, by providing numerous examples of specific miRNAs and lncRNAs, we discuss how dysregulation of these mechanisms contributes to the onset and/or progression of various human diseases.
Collapse
Affiliation(s)
| | | | - Vincenzo Cavalieri
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STeBiCeF), University of Palermo, Viale delle Scienze Bld. 16, 90128 Palermo, Italy
| |
Collapse
|
29
|
Schwarzmueller LJ, Adam RS, Moreno LF, Nijman LE, Logiantara A, Eleonora S, Bril O, Vromans S, de Groot NE, Giugliano FP, Stepanova E, Muncan V, Elbers CC, Lenos KJ, Zwijnenburg DA, van Eijndhoven MAJ, Pegtel DM, van Neerven SM, Loayza-Puch F, Dadali T, Broom WJ, Maier MA, Koster J, Vermeulen L, Léveillé N. Identifying colorectal cancer-specific vulnerabilities in the Wnt-driven long non-coding transcriptome. Gut 2025; 74:571-585. [PMID: 39562049 PMCID: PMC12013597 DOI: 10.1136/gutjnl-2024-332752] [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] [Received: 04/29/2024] [Accepted: 10/31/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Aberrant Wnt pathway activation is a key driver of colorectal cancer (CRC) and is essential to sustain tumour growth and progression. Although the downstream protein-coding target genes of the Wnt cascade are well known, the long non-coding transcriptome has not yet been fully resolved. OBJECTIVE In this study, we aim to comprehensively reveal the Wnt-regulated long non-coding transcriptome and exploit essential molecules as novel therapeutic targets. DESIGN We used global run-on sequencing to define β-catenin-regulated long non-coding RNAs (lncRNAs) in CRC. CRISPRi dropout screens were subsequently used to establish the functional relevance of a subset of these lncRNAs for long-term expansion of CRC. RESULTS We uncovered that LINC02418 is essential for cancer cell clonogenic outgrowth. Mechanistically, LINC02418 regulates MYC expression levels to promote CRC stem cell functionality and prevent terminal differentiation. Furthermore, we developed effective small interfering RNA (siRNA)-based therapeutics to target LINC02418 RNA in vivo. CONCLUSION We propose that cancer-specific Wnt-regulated lncRNAs provide novel therapeutic opportunities to interfere with the Wnt pathway, which has so far defied effective pharmacological inhibition.
Collapse
Affiliation(s)
- Laura J Schwarzmueller
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Ronja S Adam
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Leandro F Moreno
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Lisanne E Nijman
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Adrian Logiantara
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Steven Eleonora
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Oscar Bril
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Sophie Vromans
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Nina E de Groot
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Francesca Paola Giugliano
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ekaterina Stepanova
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Vanesa Muncan
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Clara C Elbers
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Kristiaan J Lenos
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Danny A Zwijnenburg
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | | | - Dirk Michiel Pegtel
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Sanne M van Neerven
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Fabricio Loayza-Puch
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tulin Dadali
- Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts, USA
| | - Wendy J Broom
- Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts, USA
| | - Martin A Maier
- Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts, USA
| | - Jan Koster
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Nicolas Léveillé
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Deng L, Gòdia M, Derks MFL, Harlizius B, Farhangi S, Tang Z, Groenen MAM, Madsen O. Comprehensive expression genome-wide association study of long non-coding RNAs in four porcine tissues. Genomics 2025; 117:111026. [PMID: 40049421 DOI: 10.1016/j.ygeno.2025.111026] [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: 08/29/2024] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs), a type of non-coding RNA molecules, are known to play critical regulatory roles in various biological processes. However, the functions of the majority of lncRNAs remain largely unknown, and little is understood about the regulation of lncRNA expression. In this study, high-throughput DNA genotyping and RNA sequencing were applied to investigate genomic regions associated with lncRNA expression, commonly referred to as lncRNA expression quantitative trait loci (eQTLs). We analyzed the liver, lung, spleen, and muscle transcriptomes of 100 three-way crossbred sows to identify lncRNA transcripts, explore genomic regions that might influence lncRNA expression, and identify potential regulators interacting with these regions. RESULT We identified 6380 lncRNA transcripts and 3733 lncRNA genes. Correlation tests between the expression of lncRNAs and protein-coding genes were performed. Subsequently, functional enrichment analyses were carried out on protein-coding genes highly correlated with lncRNAs. Our correlation results of these protein-coding genes uncovered terms that are related to tissue specific functions. Additionally, heatmaps of lncRNAs and protein-coding genes at different correlation levels revealed several distinct clusters. An expression genome-wide association study (eGWAS) was conducted using 535,896 genotypes and 1829, 1944, 2089, and 2074 expressed lncRNA genes for liver, spleen, lung, and muscle, respectively. This analysis identified 520,562 significant associations and 6654, 4525, 4842, and 7125 eQTLs for the respective tissues. Only a small portion of these eQTLs were classified as cis-eQTLs. Fifteen regions with the highest eQTL density were selected as eGWAS hotspots and potential mechanisms of lncRNA regulation in these hotspots were explored. However, we did not identify any interactions between the transcription factors or miRNAs in the hotspots and the lncRNAs, nor did we observe a significant enrichment of regulatory elements in these hotspots. While we could not pinpoint the key factors regulating lncRNA expression, our results suggest that the regulation of lncRNAs involves more complex mechanisms. CONCLUSION Our findings provide insights into several features and potential functions of lncRNAs in various tissues. However, the mechanisms by which lncRNA eQTLs regulate lncRNA expression remain unclear. Further research is needed to explore the regulation of lncRNA expression and the mechanisms underlying lncRNA interactions with small molecules and regulatory proteins.
Collapse
Affiliation(s)
- Liyan Deng
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen, the Netherlands; Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China; Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| | - Marta Gòdia
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen, the Netherlands
| | - Martijn F L Derks
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen, the Netherlands; Topigs Norsvin Research Center, 's-Hertogenbosch, the Netherlands
| | | | - Samin Farhangi
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen, the Netherlands
| | - Zhonglin Tang
- Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute, Chinese Academy of Agricultural Sciences, Foshan 528226, China; Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Martien A M Groenen
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen, the Netherlands
| | - Ole Madsen
- Animal Breeding and Genomics, Wageningen University & Research, Wageningen, the Netherlands.
| |
Collapse
|
31
|
Biayna J, Dumbović G. Decoding subcellular RNA localization one molecule at a time. Genome Biol 2025; 26:45. [PMID: 40033325 PMCID: PMC11874642 DOI: 10.1186/s13059-025-03507-8] [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: 07/25/2024] [Accepted: 02/13/2025] [Indexed: 03/05/2025] Open
Abstract
Eukaryotic cells are highly structured and composed of multiple membrane-bound and membraneless organelles. Subcellular RNA localization is a critical regulator of RNA function, influencing various biological processes. At any given moment, RNAs must accurately navigate the three-dimensional subcellular environment to ensure proper localization and function, governed by numerous factors, including splicing, RNA stability, modifications, and localizing sequences. Aberrant RNA localization can contribute to the development of numerous diseases. Here, we explore diverse RNA localization mechanisms and summarize advancements in methods for determining subcellular RNA localization, highlighting imaging techniques transforming our ability to study RNA dynamics at the single-molecule level.
Collapse
Affiliation(s)
- Josep Biayna
- Goethe University Frankfurt, Center for Molecular Medicine, Institute for Cardiovascular Regeneration, Frankfurt, Germany
| | - Gabrijela Dumbović
- Goethe University Frankfurt, Center for Molecular Medicine, Institute for Cardiovascular Regeneration, Frankfurt, Germany.
- Cardio-Pulmonary Institute (CPI), Goethe University, Frankfurt, Frankfurt, Germany.
- German Center of Cardiovascular Research (DZHK), Partner Site Rhein/Main, Frankfurt, Germany.
| |
Collapse
|
32
|
Sun Q, Zhai W, Wang H, Gao Z, Liu H. A novel lncRNA MSTRG.59348.1 regulates muscle cells proliferation and innate immunity of Megalobrama amblycephala. Int J Biol Macromol 2025; 294:139445. [PMID: 39756731 DOI: 10.1016/j.ijbiomac.2024.139445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/07/2025]
Abstract
In mammals, long non-coding RNAs (lncRNAs) play a regulatory role in gene expression, contribute to immune responses, and aid in pathogen elimination, primarily through interactions with RNA-binding proteins (RBPs). However, the role of lncRNAs in fish innate immunity and their interaction with RBPs remains uncertain. To investigate the immunomodulatory role of lncRNAs in Megalobrama amblycephala, we identified the novel lncRNA MSTRG.59348.1 and examined its function in the innate immune response to Aeromonas hydrophila infection. Localization studies in hepatocytes revealed that MSTRG.59348.1 is primarily located in the nucleus, suggesting its potential involvement in gene regulation, possibly through chromatin modification or other nuclear processes. The expression of MSTRG.59348.1 was significantly up-regulated after lipopolysaccharide (LPS) stimulation in liver cells. RNA-seq analysis of muscle cells revealed that genes differentially expressed following MSTRG.59348.1 overexpression were enriched in immune pathways. MSTRG.59348.1 overexpression significantly inhibited the expression of sting and ifn, and significantly up-regulated muscle cell viability and promoted cell proliferation by targeting sting, ifn, nf-κb1, and bcl2. Screening by RNA pull-down and mass spectrometry identified 57 RBPs interacting with MSTRG.59348.1, with functions enriched in immune pathways. Our results suggest that MSTRG.59348.1 plays a crucial regulatory role in fish antibacterial response, marking it as a significant subject for future research in innate immunity.
Collapse
Affiliation(s)
- Qianhui Sun
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affair, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China; Xiaogan Academy of Agricultural Sciences, Xiaogan 432100, China
| | - Wenya Zhai
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affair, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Huanling Wang
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affair, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Zexia Gao
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affair, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Hong Liu
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affair, Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China.
| |
Collapse
|
33
|
Sun C, Zhou C, Daneshvar K, Ben Saad A, Kratkiewicz AJ, Toles BJ, Arghiani N, Hess A, Chen JY, Pondick JV, York SR, Li W, Moran SP, Gentile SD, Rahman RU, Li Z, Zhou P, Sparks RP, Habboub T, Kim BM, Choi MY, Affo S, Schwabe RF, Popov YV, Mullen AC. Conserved long noncoding RNA TILAM promotes liver fibrosis through interaction with PML in HSCs. Hepatology 2025; 81:853-869. [PMID: 38563629 PMCID: PMC11825499 DOI: 10.1097/hep.0000000000000822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 02/01/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND AND AIMS Fibrosis is the common end point for all forms of chronic liver injury, and the progression of fibrosis leads to the development of end-stage liver disease. Activation of HSCs and their transdifferentiation into myofibroblasts results in the accumulation of extracellular matrix proteins that form the fibrotic scar. Long noncoding RNAs regulate the activity of HSCs and provide targets for fibrotic therapies. APPROACH AND RESULTS We identified long noncoding RNA TILAM located near COL1A1 , expressed in HSCs, and induced with liver fibrosis in humans and mice. Loss-of-function studies in human HSCs and human liver organoids revealed that TILAM regulates the expression of COL1A1 and other extracellular matrix genes. To determine the role of TILAM in vivo, we annotated the mouse ortholog ( Tilam ), generated Tilam- deficient green fluorescent protein-reporter mice, and challenged these mice in 2 different models of liver fibrosis. Single-cell data and analysis of single-data and analysis of Tilam-deficient reporter mice revealed that Tilam is induced in murine HSCs with the development of fibrosis in vivo. Tilam -deficient reporter mice revealed that Tilam is induced in murine HSCs with the development of fibrosis in vivo. Furthermore, loss of Tilam expression attenuated the development of fibrosis in the setting of in vivo liver injury. Finally, we found that TILAM interacts with promyelocytic leukemia nuclear body scaffold protein to regulate a feedback loop by which TGF-β2 reinforces TILAM expression and nuclear localization of promyelocytic leukemia nuclear body scaffold protein to promote the fibrotic activity of HSCs. CONCLUSIONS TILAM is activated in HSCs with liver injury and interacts with promyelocytic leukemia nuclear body scaffold protein to drive the development of fibrosis. Depletion of TILAM may serve as a therapeutic approach to combat the development of end-stage liver disease.
Collapse
Affiliation(s)
- Cheng Sun
- Department of Medicine, Division of Gastroenterology, Chan Medical School, University of Massachusetts, Worcester, Massachusetts, USA
| | - Chan Zhou
- Department of Population and Quantitative Health Sciences, Chan Medical School, University of Massachusetts, Worcester, Massachusetts USA
| | - Kaveh Daneshvar
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Amel Ben Saad
- Department of Medicine, Division of Gastroenterology, Chan Medical School, University of Massachusetts, Worcester, Massachusetts, USA
| | - Arcadia J. Kratkiewicz
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin J. Toles
- Department of Medicine, Division of Gastroenterology, Chan Medical School, University of Massachusetts, Worcester, Massachusetts, USA
| | - Nahid Arghiani
- Department of Medicine, Division of Gastroenterology, Chan Medical School, University of Massachusetts, Worcester, Massachusetts, USA
| | - Anja Hess
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer Y. Chen
- Department of Medicine, Liver Center, University of California, San Francisco, California, USA
| | - Joshua V. Pondick
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Samuel R. York
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Wenyang Li
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sean P. Moran
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefan D. Gentile
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Raza Ur Rahman
- Department of Medicine, Division of Gastroenterology, Chan Medical School, University of Massachusetts, Worcester, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Zixiu Li
- Department of Population and Quantitative Health Sciences, Chan Medical School, University of Massachusetts, Worcester, Massachusetts USA
| | - Peng Zhou
- Department of Population and Quantitative Health Sciences, Chan Medical School, University of Massachusetts, Worcester, Massachusetts USA
| | - Robert P. Sparks
- Department of Medicine, Division of Gastroenterology, Chan Medical School, University of Massachusetts, Worcester, Massachusetts, USA
| | - Tim Habboub
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Byeong-Moo Kim
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Y. Choi
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Silvia Affo
- Department of Liver, Digestive System, and Metabolism, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Robert F. Schwabe
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Yury V. Popov
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Alan C. Mullen
- Department of Medicine, Division of Gastroenterology, Chan Medical School, University of Massachusetts, Worcester, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
34
|
Bonacci RE, McGill M, Le NTA, Barkarar M, Finnegan C, Wilson M, Ajagbe O, Udekwu CC, Gorski K, Manohar J, Sboner A, Ogunwobi OO. Upregulation of the interferon-inducible antiviral gene RSAD2 in neuroendocrine prostate cancer via PVT1 exon 9 dependent and independent pathways. J Biol Chem 2025; 301:108370. [PMID: 40024473 PMCID: PMC11994405 DOI: 10.1016/j.jbc.2025.108370] [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: 07/30/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
PVT1 exon 9 overexpression is a newly uncovered aberration in prostate cancer (PCa). We have previously demonstrated the exon 9 region of PVT1 is overexpressed in some patient PCa tissues and caused development of neuroendocrine prostate cancer (NEPC) in vitro and in vivo. In this study, we focused on elucidating downstream mechanisms induced by PVT1 exon 9 overexpression with the goal of further understanding its role in NEPC development. RNA-seq analysis of a PVT1 exon 9 overexpressing PCa model revealed significant enrichment of genes responsible for inducing inflammatory processes including RSAD2. We observed RSAD2 overexpression in all NEPC models examined whereas PVT1 exon 9 was overexpressed only in a subset of the NEPC models. We identified two distinct pathways in which RSAD2 is overexpressed: one dependent and one independent on PVT1 exon 9 overexpression. Knockdown of RSAD2 suppressed cell proliferation and migration suggestive of its role as a therapeutic target in NEPC. We identified RSAD2 induces increased cell proliferation, colony formation, and may be involved in the transition between CRPC and NEPC. Distinct differences between PVT1 exon 9-dependent and PVT1 exon 9-independent NEPC models include differences in type II interferon signaling and AR modulation. PVT1 exon 9 binds to RSAD2 protein and disruption of binding significantly impedes downstream interferon gamma secretion by PVT1 exon 9-dependent NEPC cells. These novel findings indicate the importance of these two independent pathways in NEPC, the need to identify relevant NEPC patient populations and study strategies for targeting PVT1 exon 9 and/or RSAD2.
Collapse
Affiliation(s)
- Rachel E Bonacci
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Meghan McGill
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Nu Thuy Anh Le
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Murtaza Barkarar
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Colin Finnegan
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Maya Wilson
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Oluwabusola Ajagbe
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Chinedum C Udekwu
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Kathryn Gorski
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Jyothi Manohar
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Andrea Sboner
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Olorunseun O Ogunwobi
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA.
| |
Collapse
|
35
|
An X, Sun L, Zheng H, Xiao Y, Sun W, Yu D. Mitochondria-associated non-coding RNAs and their impact on drug resistance. Front Pharmacol 2025; 16:1472804. [PMID: 40078288 PMCID: PMC11897306 DOI: 10.3389/fphar.2025.1472804] [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: 07/30/2024] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
Drug resistance is a prevalent challenge in clinical disease treatment, often leading to disease relapse and poor prognosis. Therefore, it is crucial to gain a deeper understanding of the molecular mechanisms underlying drug resistance and to develop targeted strategies for its effective prevention and management. Mitochondria, as vital energy-producing organelles within cells, have been recognized as key regulators of drug sensitivity. Processes such as mitochondrial fission, fusion, mitophagy, changes in membrane potential, reactive oxygen species (ROS) accumulation, and oxidative phosphorylation (OXPHOS) are all linked to drug sensitivity. Non-coding RNAs (ncRNAs) enriched in mitochondria (mtncRNA), whether transcribed from mitochondrial DNA (mtDNA) or from the nucleus and transported to mitochondria, can regulate the transcription and translation of mtDNA, thus influencing mitochondrial function, including mitochondrial substance exchange and energy metabolism. This, in turn, directly or indirectly affects cellular sensitivity to drugs. This review summarizes the types of mtncRNAs associated with drug resistance and the molecular mechanisms regulating drug resistance. Our aim is to provide insights and strategies for overcoming drug resistance by modulating mtncRNAs.
Collapse
Affiliation(s)
- Xingna An
- Department of Core Facility, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Lina Sun
- Department of Hematology-Oncology, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Huan Zheng
- Department of Hematology-Oncology, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Yinghui Xiao
- Department of Core Facility, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Weixia Sun
- Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Dehai Yu
- Department of Core Facility, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
36
|
Ruffo P, Traynor BJ, Conforti FL. Advancements in genetic research and RNA therapy strategies for amyotrophic lateral sclerosis (ALS): current progress and future prospects. J Neurol 2025; 272:233. [PMID: 40009238 PMCID: PMC11865122 DOI: 10.1007/s00415-025-12975-8] [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: 11/12/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
This review explores the intricate landscape of neurodegenerative disease research, focusing on Amyotrophic Lateral Sclerosis (ALS) and the intersection of genetics and RNA biology to investigate the causative pathogenetic basis of this fatal disease. ALS is a severe neurodegenerative disease characterized by the progressive loss of motor neurons, leading to muscle weakness and paralysis. Despite significant research advances, the exact cause of ALS remains largely unknown. Thanks to the application of next-generation sequencing (NGS) approaches, it was possible to highlight the fundamental role of rare variants with large effect sizes and involvement of portions of non-coding RNA, providing valuable information on risk prediction, diagnosis, and treatment of age-related diseases, such as ALS. Genetic research has provided valuable insights into the pathophysiology of ALS, leading to the development of targeted therapies such as antisense oligonucleotides (ASOs). Regulatory agencies in several countries are evaluating the commercialization of Qalsody (Tofersen) for SOD1-associated ALS, highlighting the potential of gene-targeted therapies. Furthermore, the emerging significance of microRNAs (miRNAs) and long RNAs are of great interest. MiRNAs have emerged as promising biomarkers for diagnosing ALS and monitoring disease progression. Understanding the role of lncRNAs in the pathogenesis of ALS opens new avenues for therapeutic intervention. However, challenges remain in delivering RNA-based therapeutics to the central nervous system. Advances in genetic screening and personalized medicine hold promise for improving the management of ALS. Ongoing clinical trials use genomic approaches for patient stratification and drug targeting. Further research into the role of non-coding RNAs in the pathogenesis of ALS and their potential as therapeutic targets is crucial to the development of effective treatments for this devastating disease.
Collapse
Affiliation(s)
- Paola Ruffo
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
- Medical Genetics Laboratory, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Francesca Luisa Conforti
- Medical Genetics Laboratory, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| |
Collapse
|
37
|
Zhao S, Wang Y, Zhou L, Li Z, Weng Q. Exploring the Potential of tsRNA as Biomarkers for Diagnosis and Treatment of Neurogenetic Disorders. Mol Neurobiol 2025:10.1007/s12035-025-04760-5. [PMID: 40009263 DOI: 10.1007/s12035-025-04760-5] [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: 06/04/2024] [Accepted: 02/08/2025] [Indexed: 02/27/2025]
Abstract
tRNA-derived small RNA (tsRNA) is a recently discovered small non-coding RNA (ncRNA) molecule that widely exists in prokaryotic and eukaryotic transcriptomes and is produced by specific cleavage of mature tRNA or precursor tRNA. In recent years, with the development of high-throughput sequencing technology, tsRNA has been found to have a variety of biological functions, including gene expression regulation, stress signal activation, etc. In addition, it has been found that these molecules are abnormally expressed in various diseases and participate in various pathological processes, which play an important role. At present, more and more studies have shown that the expression level of tsRNA changes significantly during the development of neurogenetic diseases. This review provides an overview of the classification and biological functions of tsRNAs, with a particular emphasis on their roles in neurogenetic disorders and their potential as diagnostic biomarkers and therapeutic targets. Despite the nascent stage of tsRNA research, their relevance to the diagnosis and treatment of neurogenetic diseases warrants further investigation.
Collapse
Affiliation(s)
- Shiqi Zhao
- Department of Neurology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, China
- Health Science Center, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Yujia Wang
- Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Liqun Zhou
- Health Science Center, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Zhe Li
- Department of Neurology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, China.
| | - Qiuyan Weng
- Department of Neurology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, China.
| |
Collapse
|
38
|
Song GY, Yu QH, Xing XK, Fan XM, Xu SG, Zhang WB, Wu YY, Zhang N, Chao TZ, Wang F, Ding CS, Guo CY, Ma L, Sun CY, Duan SY, Xu P. The YTHDC1 reader protein recognizes and regulates the lncRNA MEG3 following its METTL3-mediated m 6A methylation: a novel mechanism early during radiation-induced liver injury. Cell Death Dis 2025; 16:127. [PMID: 39994235 PMCID: PMC11850776 DOI: 10.1038/s41419-025-07417-2] [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: 07/25/2024] [Revised: 12/20/2024] [Accepted: 01/31/2025] [Indexed: 02/26/2025]
Abstract
While apoptotic cell death is known to be central to the pathogenesis of radiation-induced liver injury (RILI), the mechanistic basis for this apoptotic activity remains poorly understood. N6-methyladenosine (m6A) modifications are the most common form of reversible methylation observed on lncRNAs in eukaryotic cells, with their presence leading to pronounced changes in the activity of a range of biological processes. The degree to which m6A modification plays a role in the induction of apoptotic cell death in response to ionizing radiation (IR) in the context of RILI remains to be established. Here, IR-induced apoptosis was found to significantly decrease the levels of m6A present, with a pronounced decrease in the expression of methyltransferase-like 3 (METTL3) at 2 d post radiation in vitro. From a mechanistic perspective, a methylated RNA immunoprecipitation assay found that lncRNA MEG3 was a major METTL3 target. The expression of MEG3 was upregulated via METTL3-mediated m6A in a process that was dependent on YTHDC1, ultimately reversing the miR-20b-mediated inhibition of BNIP2 expression. Together, these findings demonstrate that the responsivity of METTL3 activity to IR plays a role in IR-induced apoptotic cell death, leading to the reverse of miR-20b-mediated BNIP2 inhibition through the YTHDC1-dependent m6A modification of MEG3, suggesting that this process may play a central role in RILI incidence.
Collapse
Affiliation(s)
- Gui-Yuan Song
- Laboratory of Radiation-induced Diseases and Molecule-targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China
| | - Qing-Hua Yu
- Laboratory of Radiation-induced Diseases and Molecule-targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
- School of Public Health, Weifang Medical University, Weifang, Shandong, China
| | - Xue-Kun Xing
- School of Public Health, Guilin Medical University, Guilin, Guangxi, China
| | - Xin-Ming Fan
- Department of Radiotherapy, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, China
| | - Si-Guang Xu
- Key Laboratory of Medical Tissue Regeneration of Henan Province, Xinxiang Medical University, Xinxiang, Henan, China
| | - Wen-Bo Zhang
- Key Laboratory of Medical Tissue Regeneration of Henan Province, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yao-Yao Wu
- Laboratory of Radiation-induced Diseases and Molecule-targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
| | - Nan Zhang
- Laboratory of Radiation-induced Diseases and Molecule-targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
| | - Tian-Zhu Chao
- Laboratory of Radiation-induced Diseases and Molecule-targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
| | - Fei Wang
- Laboratory of Radiation-induced Diseases and Molecule-targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
| | - Cheng-Shi Ding
- Laboratory of Radiation-induced Diseases and Molecule-targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
| | - Cun-Yang Guo
- Laboratory of Radiation-induced Diseases and Molecule-targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
- School of Public Health, Binzhou Medical University, Yantai, Shandong, China
| | - Li Ma
- Department of Radiotherapy, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, China
| | - Chang-Ye Sun
- Key Laboratory of Medical Tissue Regeneration of Henan Province, Xinxiang Medical University, Xinxiang, Henan, China
| | - Shu-Yan Duan
- Laboratory of Radiation-induced Diseases and Molecule-targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
| | - Ping Xu
- Laboratory of Radiation-induced Diseases and Molecule-targeted Drugs, School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China.
| |
Collapse
|
39
|
Doghish AS, Mahmoud A, Abd-Elmawla MA, Zaki MB, Aborehab NM, Hatawsh A, Radwan AF, Sayed GA, Moussa R, Abdel-Reheim MA, Mohammed OA, Elimam H. Innovative perspectives on glioblastoma: the emerging role of long non-coding RNAs. Funct Integr Genomics 2025; 25:43. [PMID: 39992471 DOI: 10.1007/s10142-025-01557-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025]
Abstract
Glioblastoma (GBM) is a highly aggressive and treatment-resistant brain tumor. Recent advancements have highlighted the crucial role of long noncoding RNAs (lncRNAs) in GBM's molecular biology. Unlike protein-coding RNAs, lncRNAs regulate gene expression through transcription, post-transcriptional modifications, and chromatin remodeling. Some lncRNAs, like HOTAIR, CCAT2, CRNDE, and MALAT1, promote GBM development by affecting tumor suppressors and various signaling pathways like PI3K/Akt, mTOR, EGFR, NF-κB, and Wnt/β-catenin. Conversely, certain lncRNAs such as TUG1, MEG3, and GAS8-AS1 act as tumor suppressors and are associated with better prognosis. The study presented in the manuscript aims to explore the involvement of lncRNAs in GBM, focusing on their roles in tumor progression, proliferation, invasion, and potential implications for early detection and immunotherapy. The research seeks to elucidate the mechanisms by which specific lncRNAs influence GBM characteristics and highlight their potential as therapeutic targets or biomarkers in managing this aggressive form of brain cancer.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11231, Egypt
| | - Abdelhamid Mahmoud
- Biotechnology School, 26 of July Corridor, Nile University, Sheikh Zayed City, Giza, 12588, Egypt
| | - Mai A Abd-Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City 32897, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Menoufia National University, Km Cairo-Alexandria Agricultural Road, Menofia, Egypt
| | - Nora M Aborehab
- Department of Biochemistry, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Abdulrahman Hatawsh
- Biotechnology School, 26 of July Corridor, Nile University, Sheikh Zayed City, Giza, 12588, Egypt
| | - Abdullah F Radwan
- Department of Pharmacy, Kut University College, Al Kut, Wasit, 52001, Iraq
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Cairo, 11829, Egypt
| | - Ghadir A Sayed
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Cairo, 11829, Egypt
| | - Rewan Moussa
- Faculty of Medicine, Helwan University, Cairo, 11795, Egypt
| | | | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City 32897, Egypt.
| |
Collapse
|
40
|
Zhang X, Cai Y, Sit BHM, Jian RX, Malki Y, Zhang Y, Ong CCY, Li Q, Lam RPK, Rainer TH. Cell-Free Nucleic Acids for Early Diagnosis of Acute Ischemic Stroke: A Systematic Review and Meta-Analysis. Int J Mol Sci 2025; 26:1530. [PMID: 40003998 PMCID: PMC11855205 DOI: 10.3390/ijms26041530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Rapid identification of acute ischemic stroke (AIS) is challenging in both pre-hospital and hospital settings. We aimed to identify the most promising cell-free nucleic acids (cfNAs) as diagnostic biomarkers for IS within 72 h from symptom onset. We searched PubMed, Web of Science, EMBASE, and Cochrane Library for published articles that evaluated blood cfNAs in the early diagnosis of AIS until 10 May 2023. The diagnostic performances of individual cfNAs were pooled by random-effects meta-analysis based on the fold change of biomarkers' level between AIS and non-AIS patients. Of 2955 records, 66 articles reporting 143 different cfNAs met the inclusion criteria. The median sample size was 110, and 21.4% of the studies performed validation. Among selected high-quality studies, miR-106b-5p, miR-124, miR-155, lncRNA H19, and cfDNA showed good diagnostic performance. Data from four studies on cfDNA involving 355 AIS patients and 97 controls were pooled in the meta-analysis, which showed a significant fold change between AIS and controls (pooled ratio 1.48, 95% confidence interval 1.23-1.79, p < 0.001). This review highlights that cfDNA, miR-106b-5p, miR-124, miR-155, and lncRNA H19 are the most promising biomarkers for AIS diagnosis, and further research is needed for verification.
Collapse
Affiliation(s)
- Xiaodan Zhang
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| | - Yuee Cai
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| | - Brian Hon Man Sit
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| | - Rain Xiaoyu Jian
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| | - Yasine Malki
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China;
| | - Yilin Zhang
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| | - Christopher Chi Yat Ong
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| | - Qianyun Li
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| | - Rex Pui Kin Lam
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| | - Timothy Hudson Rainer
- Department of Emergency Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.Z.); (Y.C.); (B.H.M.S.); (R.X.J.); (Y.Z.); (C.C.Y.O.); (Q.L.); (R.P.K.L.)
| |
Collapse
|
41
|
Shi K, Huang LD, Li D, Luo WM, Liu HS, Ding DX, Guo Q, Liu YF. Aberrant SNHG expression predicts poor prognosis in esophageal cancer using meta-analysis and bioinformatics analysis. BMC Gastroenterol 2025; 25:63. [PMID: 39920577 PMCID: PMC11804041 DOI: 10.1186/s12876-025-03621-8] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 01/16/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Small nucleolar RNA host gene (SNHG) family were reported involved in various biological processes and may be used as a promising prognostic marker in esophageal cancer (EC). A meta-analysis was performed to investigate the relationship between SNHG expression and prognosis of EC in this study. METHODS Relevant databases were browsed to obtain suitable publications. Hazard ratio (HR) with 95% confidence interval (CI) were extracted to explore the association between SNHG expression and EC prognosis. Odds ratio (OR) with 95%CI were extracted to assess the association between SNHG expression and other clinicopathological parameters. Sensitivity analysis and publication bias were performed to explore the reliability and robustness of the results. Bio-informatics has been explored in order to confirm our conclusions more comprehensively. RESULTS 16 studies comprising 1229 patients were enrolled. The results showed that increasing SNHG expression indicated worse overall survival (HR: 1.392, 95%CI = 0.876-1.908). SNHG2, SNHG5, and SNHG12 were down-regulated, while other SNHGs were up-regulated in EC. In populations with low expression of SNHG2, SNHG5, and SNHG12, increasing SNHG expression predicted a favorable cancer prognosis (HR: 0.511, 95%CI = 0.322-0.700). Conversely, in populations with high expression of other SNHGs, SNHG expression indicated poor prognosis (OR: 2.340, 95%CI = 1.744-2.936). Elevated SNHG expression also implied advanced TNM stage (OR 1.578, 95%CI = 1.273-1.956) and lymph node metastasis (OR: 1.533, 95%CI = 1.205-1.950). CONCLUSION Increased expression of SNHG2, SNHG5, and SNHG12, and decreased expression of other SNHGs tended to have a favorable prognosis in patients with EC. These findings suggest that SNHG may serve as a prognostic marker and therapeutic target for EC.
Collapse
Affiliation(s)
- Ke Shi
- Department of Thoracic Surgery, Beilun District People's Hospital of Ningbo, Ningbo City, China
| | - Li-De Huang
- Department of Pain management, People's Hospital of Shiyan City, Hubei Medical University, Shiyan City, China
| | - Dan Li
- Department of Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan City, China
| | - Wei-Min Luo
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan City, China
| | - Hua-Song Liu
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan City, China
| | - Dong-Xiao Ding
- Department of Thoracic Surgery, Beilun District People's Hospital of Ningbo, Ningbo City, China.
| | - Qiang Guo
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan City, China.
| | - Yue-Feng Liu
- Department of Ophthalmology, Taihe Hospital, Hubei University of Medicine, Shiyan City, China.
| |
Collapse
|
42
|
Afroze N, Sundaram MK, Haque S, Hussain A. Long non-coding RNA involved in the carcinogenesis of human female cancer - a comprehensive review. Discov Oncol 2025; 16:122. [PMID: 39912983 PMCID: PMC11803034 DOI: 10.1007/s12672-025-01848-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/22/2025] [Indexed: 02/07/2025] Open
Abstract
Recent years have seen an increase in our understanding of lncRNA and their role in various disease states. lncRNA molecules have been shown to contribute to carcinogenesis and influence the various cancer hallmarks and signalling pathways. It is pertinent to understand the specific contributions and mechanisms of action of these molecules in various cancers. This review provides an overview of the various lncRNA entities that influence and regulate the gynaecological cancers, namely, cervical, breast, ovarian and uterine cancers. The review curates a list of the key players and their effect on cellular processes. lncRNA molecules show immense potential to be used as diagnostic and prognostic indicators and in therapeutic strategies. Several phytochemicals, small molecules, RNA-based regulators, oligos and gene editing tools show promise as a therapeutic strategy. While this review highlights the promising developments in this field, it also underscores the necessity for further research to delineate the complex role of lncRNAs in cancer.
Collapse
Affiliation(s)
- Nazia Afroze
- School of Life Sciences, Manipal Academy of Higher Education, Dubai Campus, P.O. Box 345050, Dubai, United Arab Emirates
| | - Madhumitha K Sundaram
- School of Life Sciences, Manipal Academy of Higher Education, Dubai Campus, P.O. Box 345050, Dubai, United Arab Emirates
| | - Shafiul Haque
- Department of Nursing, College of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
- School of Medicine, Universidad Espiritu Santo, Samborondon, Ecuador
| | - Arif Hussain
- School of Life Sciences, Manipal Academy of Higher Education, Dubai Campus, P.O. Box 345050, Dubai, United Arab Emirates.
| |
Collapse
|
43
|
He Y, Li J, Chen Y, Ren B, Zhou Z, Liu J, Gao H, Li F, Li B, Liu L, Shen H. Expression and Function of Long Non-coding RNA in Endemic Cretinism. Mol Neurobiol 2025; 62:1770-1787. [PMID: 39031326 DOI: 10.1007/s12035-024-04358-3] [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] [Received: 03/02/2024] [Accepted: 07/09/2024] [Indexed: 07/22/2024]
Abstract
Endemic cretinism (EC) is one of the most severe iodine deficiency disorders, leading to typical symptoms such as neurodevelopmental impairments or mental deficits. In addition to environmental factors, the pathogenesis of its genetic contribution remains unclear. The study revealed the differential expression profiles of long non-coding RNA(lncRNA) and messenger RNA(mRNA) based on high-throughput RNA-seq. GO and KEGG analyses were used to annotate the function and pathway of differentially expressed (DE) mRNA and co-expressed mRNA. The protein-protein interaction(PPI) network was established. The expression levels of three lncRNAs and six mRNAs were validated by quantitative real-time PCR analysis (qRT-PCR) and subjected to correlation analysis. Compared to controls, a total of 864 lncRNAs and 393 mRNAs were differentially expressed. The PPI network had 149 nodes and 238 edges, and three key protein-coding genes were observed. Levels of LINC01220 and target mRNA IDO1 were statistically elevated in EC patients. Differentially expressed lncRNA may be a new potential player in EC. LINC01220 and IDO1 might interact with each other to participate in EC. The biological process of regulation of postsynaptic membrane potential and the Rap1 signaling pathway might exert a regulating role in the pathophysiological process of EC. Our findings could provide more theoretical and experimental evidence for investigating the pathophysiological mechanisms of EC.
Collapse
Affiliation(s)
- Yanhong He
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Jianshuang Li
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Heilongjiang Province 163319, Daqing City, People's Republic of China
| | - Yun Chen
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Bingxuan Ren
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Zheng Zhou
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Jinjin Liu
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Haiyan Gao
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Fan Li
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Baoxiang Li
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Lixiang Liu
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China
| | - Hongmei Shen
- Chinese Centre for Disease Control and Prevention, Centre for Endemic Disease Control, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China.
- Commission & Education Bureau of Heilongjiang Province, Key Laboratory of Etiology and Epidemiology, National Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China.
- Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Heilongjiang Province 150081, Harbin City, People's Republic of China.
| |
Collapse
|
44
|
Farberov S, Ziv O, Lau JY, Ben-Tov Perry R, Lubelsky Y, Miska E, Kudla G, Ulitsky I. Structural features within the NORAD long noncoding RNA underlie efficient repression of Pumilio activity. Nat Struct Mol Biol 2025; 32:287-299. [PMID: 39327473 PMCID: PMC7617650 DOI: 10.1038/s41594-024-01393-5] [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: 10/18/2022] [Accepted: 08/27/2024] [Indexed: 09/28/2024]
Abstract
Long noncoding RNAs (lncRNAs) are increasingly appreciated for their important functions in mammalian cells. However, how their functional capacities are encoded in their sequences and manifested in their structures remains largely unknown. Some lncRNAs bind to and modulate the availability of RNA-binding proteins, but the structural principles that underlie this mode of regulation are unknown. The NORAD lncRNA is a known decoy for Pumilio proteins, which modulate the translation and stability of hundreds of messenger RNAs and, consequently, a regulator of genomic stability and aging. Here we probed the RNA structure and long-range RNA-RNA interactions formed by human NORAD inside cells under different stressful conditions. We discovered a highly modular structure consisting of well-defined domains that contribute independently to NORAD function. Following arsenite stress, most structural domains undergo relaxation and form interactions with other RNAs that are targeted to stress granules. We further revealed a unique structural organization that spatially clusters the multiple Pumilio binding sites along NORAD and consequently contributes to the derepression of Pumilio targets. We then applied these structural principles to design an effective artificial decoy for the let-7 microRNA. Our work demonstrates how the sequence of a lncRNA spatially clusters its function into separated domains and how structural principles can be employed for the rational design of lncRNAs with desired activities.
Collapse
Affiliation(s)
- Svetlana Farberov
- Department of Immunology and Regenerative Biology and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Omer Ziv
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK.
- Eleven Therapeutics, Cambridge, UK.
| | - Jian You Lau
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh, UK
| | - Rotem Ben-Tov Perry
- Department of Immunology and Regenerative Biology and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Yoav Lubelsky
- Department of Immunology and Regenerative Biology and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Eric Miska
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK.
| | - Grzegorz Kudla
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh, UK.
| | - Igor Ulitsky
- Department of Immunology and Regenerative Biology and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
45
|
Han Y, Pu Q, Fan T, Wei T, Xu Y, Zhao L, Liu S. Long non-coding RNAs as promising targets for controlling disease vector mosquitoes. INSECT SCIENCE 2025; 32:24-41. [PMID: 38783627 DOI: 10.1111/1744-7917.13383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024]
Abstract
Hematophagous female mosquitoes are important vectors of numerous devastating human diseases, posing a major public health threat. Effective prevention and control of mosquito-borne diseases rely considerably on progress in understanding the molecular mechanisms of various life activities, and accordingly, the molecules that regulate the various life activities of mosquitoes are potential targets for implementing future vector control strategies. Many long non-coding RNAs (lncRNAs) have been identified in mosquitoes and significant progress has been made in determining their functions. Here, we present a comprehensive overview of the research advances on mosquito lncRNAs, including their molecular identification, function, and interaction with other non-coding RNAs, as well as their synergistic regulatory roles in mosquito life activities. We also highlight the potential roles of competitive endogenous RNAs in mosquito growth and development, as well as in insecticide resistance and virus-host interactions. Insights into the biological functions and mechanisms of lncRNAs in mosquito life activities, viral replication, pathogenesis, and transmission will contribute to the development of novel drugs and safe vaccines.
Collapse
Affiliation(s)
- Yujiao Han
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Qian Pu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Ting Fan
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Tianqi Wei
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Yankun Xu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Lu Zhao
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| | - Shiping Liu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, 400716, China
| |
Collapse
|
46
|
Windon A, Al Assaad M, Hadi K, Mendelson N, Hissong E, Deshpande A, Tranquille M, Mclee J, Levine MF, Patel M, Medina-Martínez JS, Chiu K, Manohar J, Sigouros M, Ocean AJ, Sboner A, Jessurun J, Elemento O, Shah M, Mosquera JM. Emerging molecular phenotypes and potential therapeutic targets in esophageal and gastric adenocarcinoma unearthed by whole genome and transcriptome analyses. Pathol Res Pract 2025; 266:155788. [PMID: 39708521 DOI: 10.1016/j.prp.2024.155788] [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: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Adenocarcinoma of the esophagus and stomach demands a deeper molecular understanding to advance treatment strategies and improve patient outcomes. Here, we profiled the genome and transcriptome landscape of these cancers, explored molecular characteristics that are undetectable by other sequencing platforms, and analyzed their potential clinical ramifications. METHODS Our study employed state-of-the-art integrative analyses of whole genome and transcriptome sequencing on 51 matched tumor and germline samples from 46 patients. Mutations and rearrangements in clinically relevant cancer genes were investigated and correlated with OncoKB, a knowledge-based precision oncology database, to identify treatment implications. Genome-wide signatures and manually curated molecular profiles were also determined. RESULTS The analyses revealed 90 targetable oncogenic mutations and fusions in 63 % of the patients, including novel NTRK, NRG1, ALK, and MET fusions, and structural variants in cancer genes like RAD51B. Also, molecular signatures associated with mismatch repair and homologous recombination deficiency were elucidated. Notably, we identified CDK12-type genomic instability associated with CDK12 fusions. CONCLUSIONS Our findings support the potential of whole genome and transcriptome sequencing analyses as a comprehensive approach to identify treatment targets in adenocarcinoma of the stomach and the esophagus, and their application in precision oncology.
Collapse
Affiliation(s)
- Annika Windon
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Majd Al Assaad
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Nicole Mendelson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Erika Hissong
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Marvel Tranquille
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Justin Mclee
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | | | | | - Kenrry Chiu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jyothi Manohar
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Michael Sigouros
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Allyson J Ocean
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA; Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Andrea Sboner
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - José Jessurun
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Olivier Elemento
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Manish Shah
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA; Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Juan Miguel Mosquera
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
47
|
Zhou H, Clark E, Guan D, Lagarrigue S, Fang L, Cheng H, Tuggle CK, Kapoor M, Wang Y, Giuffra E, Egidy G. Comparative Genomics and Epigenomics of Transcriptional Regulation. Annu Rev Anim Biosci 2025; 13:73-98. [PMID: 39565835 DOI: 10.1146/annurev-animal-111523-102217] [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: 11/22/2024]
Abstract
Transcriptional regulation in response to diverse physiological cues involves complicated biological processes. Recent initiatives that leverage whole genome sequencing and annotation of regulatory elements significantly contribute to our understanding of transcriptional gene regulation. Advances in the data sets available for comparative genomics and epigenomics can identify evolutionarily constrained regulatory variants and shed light on noncoding elements that influence transcription in different tissues and developmental stages across species. Most epigenomic data, however, are generated from healthy subjects at specific developmental stages. To bridge the genotype-phenotype gap, future research should focus on generating multidimensional epigenomic data under diverse physiological conditions. Farm animal species offer advantages in terms of feasibility, cost, and experimental design for such integrative analyses in comparison to humans. Deep learning modeling and cutting-edge technologies in sequencing and functional screening and validation also provide great promise for better understanding transcriptional regulation in this dynamic field.
Collapse
Affiliation(s)
- Huaijun Zhou
- Department of Animal Science, University of California, Davis, California, USA; , , ,
| | - Emily Clark
- The Roslin Institute, University of Edinburgh, Edinburgh, Midlothian, United Kingdom;
| | - Dailu Guan
- Department of Animal Science, University of California, Davis, California, USA; , , ,
| | | | - Lingzhao Fang
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark;
| | - Hao Cheng
- Department of Animal Science, University of California, Davis, California, USA; , , ,
| | | | - Muskan Kapoor
- Department of Animal Science, Iowa State University, Ames, Iowa, USA; ,
| | - Ying Wang
- Department of Animal Science, University of California, Davis, California, USA; , , ,
| | | | - Giorgia Egidy
- GABI, AgroParisTech, INRAE, Jouy-en-Josas, France; ,
| |
Collapse
|
48
|
Kalomeris T, Assaad MA, la Mora JDD, Gundem G, Levine MF, Boyraz B, Manohar J, Sigouros M, Medina-Martínez JS, Sboner A, Elemento O, Scognamiglio T, Mosquera JM. Whole genome profiling of primary and metastatic adrenocortical carcinoma unravels significant molecular events. Pathol Res Pract 2025; 266:155725. [PMID: 39626581 DOI: 10.1016/j.prp.2024.155725] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 04/29/2025]
Abstract
Adrenocortical carcinoma (ACC) is a rare, aggressive malignancy with limited treatment options and poor prognosis, with a 5-year survival rate of about 15 %. This study used whole genome sequencing to characterize the genomic landscape of five patients, one of them with both primary and metastatic samples. Key driver mutations were detected, including APC, JAK1, RFWD3 as well as other genes. Notably, a primary tumor harbored a RAD51 biallelic deleterious translocation, associated with homologous recombination deficiency signature. Large-scale copy neutral loss of heterozygosity (LOH) was identified in four tumors, three had TP53 mutations, with structural variants impacting genes as RB1, CDKN2A, and NF1. A genomic signature specific to mismatch repair was observed in a sample with MHS6 mutation. Two tumors presented novel fusions at TERT locus, including TERT::ZNF521. Comparative analysis between conventional and oncocytic ACC subtypes revealed no significant differences in mutation load, microsatellite instability, or specific gene enrichment. This comprehensive WGS analysis broadens the spectrum of genomic alterations in ACC, highlighting potential molecular targets and differences across subtypes that may inform future therapeutic strategies.
Collapse
Affiliation(s)
- Taylor Kalomeris
- Department of Pathology and Laboratory Medicine, NewYork-Presbyterian Hospital/Weill Cornell Medical Center, 1300 York Ave, New York, NY 10065, USA
| | - Majd Al Assaad
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA
| | - Jesus Delgado-de la Mora
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA
| | - Gunes Gundem
- Isabl Inc., 175 Greenwich Street, Fl 38, New York, NY 10007, USA
| | - Max F Levine
- Isabl Inc., 175 Greenwich Street, Fl 38, New York, NY 10007, USA
| | - Baris Boyraz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, USA
| | - Jyothi Manohar
- Englander Institute for Precision Medicine, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA
| | - Michael Sigouros
- Englander Institute for Precision Medicine, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA
| | | | - Andrea Sboner
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, 1305 York Avenue, New York, NY 10021, USA
| | - Olivier Elemento
- Englander Institute for Precision Medicine, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, 1305 York Avenue, New York, NY 10021, USA
| | - Theresa Scognamiglio
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, USA
| | - Juan Miguel Mosquera
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA.
| |
Collapse
|
49
|
Palma A, Buonaiuto G, Ballarino M, Laneve P. Genome biology of long non-coding RNAs in humans: A virtual karyotype. Comput Struct Biotechnol J 2025; 27:575-584. [PMID: 39989619 PMCID: PMC11847481 DOI: 10.1016/j.csbj.2025.01.026] [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: 09/29/2024] [Revised: 01/29/2025] [Accepted: 01/29/2025] [Indexed: 02/25/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) represent a groundbreaking class of RNA molecules that exert regulatory functions with remarkable tissue and cellular specificity. Although the identification of functionally significant lncRNAs is increasing, a comprehensive profiling of their genomic features remains elusive. Here, we present a detailed overview of the distribution of lncRNA genes across human chromosomes and describe key RNA features-what we refer to as a "virtual lncRNA karyotype"-that provide insights into their biosynthesis and function. To achieve this, we leveraged existing human annotation files to construct a statistical genomic portrait of lncRNAs in comparison with protein-coding genes (PCGs). We found that lncRNAs are unevenly distributed across chromosomes and identified regions of high lncRNA density on chromosomes 18, 13, and X, which overlap with PCG-rich regions. Additionally, we observed that lncRNAs generally exhibit shorter gene lengths and fewer splicing variants compared to protein-coding transcripts, with a subset displaying pronounced clustering patterns that may indicate functional relevance. Finally, we identified several clinically associated and experimentally validated SNPs impacting lncRNA genes (lncGs). Overall, this study provides a foundational reference for exploring the non-coding genome, offering new insights into the genomic characteristics of lncRNAs. These findings may enhance our understanding of their biological significance and potential roles in disease.
Collapse
Affiliation(s)
- Alessandro Palma
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome 00185, Italy
| | - Giulia Buonaiuto
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome 00185, Italy
| | - Monica Ballarino
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome 00185, Italy
| | - Pietro Laneve
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome 00185, Italy
- Institute of Molecular Biology and Pathology, National Research Council of Italy, Piazzale Aldo Moro 7, Rome 00185, Italy
| |
Collapse
|
50
|
Basu S, Nadhan R, Dhanasekaran DN. Long Non-Coding RNAs in Ovarian Cancer: Mechanistic Insights and Clinical Applications. Cancers (Basel) 2025; 17:472. [PMID: 39941838 PMCID: PMC11815776 DOI: 10.3390/cancers17030472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Ovarian cancer is a leading cause of gynecological cancer mortality worldwide, often diagnosed at advanced stages due to vague symptoms and the lack of effective early detection methods. Long non-coding RNAs (lncRNAs) have emerged as key regulators in cancer biology, influencing cellular processes such as proliferation, apoptosis, and chemoresistance. This review explores the multifaceted roles of lncRNAs in ovarian cancer pathogenesis and their potential as biomarkers and therapeutic targets. Methods: A comprehensive literature review was conducted to analyze the structural and functional characteristics of lncRNAs and their contributions to ovarian cancer biology. This includes their regulatory mechanisms, interactions with signaling pathways, and implications for therapeutic resistance. Advanced bioinformatics and omics approaches were also evaluated for their potential in lncRNA research. Results: The review highlights the dual role of lncRNAs as oncogenes and tumor suppressors, modulating processes such as cell proliferation, invasion, and angiogenesis. Specific lncRNAs, such as HOTAIR and GAS5, demonstrate significant potential as diagnostic biomarkers and therapeutic targets. Emerging technologies, such as single-cell sequencing, provide valuable insights into the tumor microenvironment and the heterogeneity of lncRNA expression. Conclusions: LncRNAs hold transformative potential in advancing ovarian cancer diagnosis, prognosis, and treatment. Targeting lncRNAs or their associated pathways offers promising strategies to overcome therapy resistance and enhance personalized medicine. Continued research integrating omics and bioinformatics will be essential to unlock the full clinical potential of lncRNAs in ovarian cancer management.
Collapse
Affiliation(s)
- Sneha Basu
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.B.); (R.N.)
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Revathy Nadhan
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.B.); (R.N.)
| | - Danny N. Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (S.B.); (R.N.)
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|