1
|
Hashemi M, Gholamrezaie H, Ziyaei F, Asadi S, Naeini ZY, Salimian N, Enayat G, Sharifi N, Aliahmadi M, Rezaie YS, Khoushab S, Rahimzadeh P, Miri H, Abedi M, Farahani N, Taheriazam A, Nabavi N, Entezari M. Role of lncRNA PVT1 in the progression of urological cancers: Novel insights into signaling pathways and clinical opportunities. Cell Signal 2025; 131:111736. [PMID: 40081549 DOI: 10.1016/j.cellsig.2025.111736] [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: 07/31/2024] [Revised: 01/02/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Urologic malignancies, encompassing cancers of the kidney, bladder, and prostate, represent approximately 25 % of all cancer cases. Recent advances have enhanced our understanding of PVT1's crucial functions. Long noncoding RNAs influence both the onset and development of cancer, as well as epigenetic alterations. Recent findings have focused on PVT1's mechanism of action across several malignancies, particularly urologic cancers. Understanding the various functions of PVT1 linked to cancer is necessary for the development of cancer detection and treatment when PVT1 is dysregulated. Furthermore, recent advancements in genomic and epigenetic research have elucidated the complex regulatory networks that control PVT1 expression. Comprehending the intricate role of PVT1 Understanding the complex function of PVT1 in urologic cancers has substantial clinical implications. Here, we summarize some of the most recent findings about the carcinogenic effects of PVT1 signaling pathways and the possible treatment strategies for urological malignancies that target these pathways.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hamidreza Gholamrezaie
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Faezeh Ziyaei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Yousefian Naeini
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Cellular and Molecular Biology,Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Niloufar Salimian
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Golnaz Enayat
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nafiseh Sharifi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Melika Aliahmadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Yasamin Soofi Rezaie
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saloomeh Khoushab
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hossein Miri
- Faculty of Medicine, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Abedi
- Department of Pathology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran,Iran.
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
2
|
Saadh MJ, Muhammad FA, Albadr RJ, Sanghvi G, Ballal S, Pathak PK, Bareja L, Aminov Z, Taher WM, Alwan M, Jawad MJ, Al-Nuaimi AMA. Exosomal non-coding RNAs: key regulators of inflammation-related cardiovascular disorders. Eur J Med Res 2025; 30:395. [PMID: 40390035 PMCID: PMC12087048 DOI: 10.1186/s40001-025-02649-6] [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: 02/02/2025] [Accepted: 04/30/2025] [Indexed: 05/21/2025] Open
Abstract
Inflammation is a complex, tightly regulated process involving biochemical and cellular reactions to harmful stimuli. Often termed "the internal fire", it is crucial for protecting the body and facilitating tissue healing. While inflammation is essential for survival, chronic inflammation can be detrimental, leading to tissue damage and reduced survival. The innate immune system triggers inflammation, closely linked to the development of heart diseases, with significant consequences for individuals. Inflammation in arterial walls or the body substantially contributes to atherosclerotic disease progression, affecting the cardiovascular system. Altered lipoproteins increase the risk of excessive blood clotting, a hallmark of atherosclerotic cardiovascular disease and its complications. Integrating inflammatory biomarkers with established risk assessment techniques can enhance our ability to identify at-risk individuals, assess their risk severity, and recommend appropriate CVD prevention strategies. Exosomes, a type of extracellular vesicle, are released by various cells and mediate cell communication locally and systemically. In the past decade, exosomes have been increasingly studied for their vital roles in health maintenance and disease processes. They can transport substances like non-coding RNAs, lipids, and proteins between cells, influencing immune responses and inflammation to elicit harmful or healing effects. This study focuses on the critical role of inflammation in heart disease progression and how non-coding RNAs in exosomes modulate the inflammatory process, either exacerbating or alleviating inflammation-related damage in the cardiovascular system.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat, 360003, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Piyus Kumar Pathak
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Lakshay Bareja
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - Zafar Aminov
- Department of Public Health and Healthcare Management, 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
|
3
|
Bono H. Recent Advances in Genome Editing and Bioinformatics: Addressing Challenges in Genome Editing Implementation and Genome Sequencing. Int J Mol Sci 2025; 26:3442. [PMID: 40244417 PMCID: PMC11989416 DOI: 10.3390/ijms26073442] [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: 03/02/2025] [Revised: 03/30/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
Genome-editing technology has advanced significantly since the 2020 Nobel Prize in Chemistry was awarded for the development of clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (Cas9). While CRISPR-Cas9 has become widely used in academic research, its social implementation has lagged due to unresolved patent disputes and slower progress in gene function analysis. To address this, new approaches bypassing direct gene function analysis are needed, with bioinformatics and next-generation sequencing (NGS) playing crucial roles. NGS is essential for sequencing the genome of target species, but challenges such as data quality, genome heterogeneity, ploidy, and small individual sizes persist. Despite these issues, advancements in sequencing technologies, like PacBio high-fidelity (HiFi) long reads and high-throughput chromosome conformation capture (Hi-C), have improved genome sequencing. Bioinformatics contributes to genome editing through off-target prediction and target gene selection, both of which require accurate genome sequence information. In this review, I will give updates on the development of genome editing and bioinformatics technologies with a focus on the rapid progress in genome sequencing.
Collapse
Affiliation(s)
- Hidemasa Bono
- Graduate School of Integrated Sciences for Life, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima 739-0046, Japan; ; Tel.: +81-82-424-4013
- Department of Biological Science, School of Science, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima 739-0046, Japan
- Genome Editing Innovation Center, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima 739-0046, Japan
| |
Collapse
|
4
|
Jiang R, Chouchane M, Chen KS, Moey C, Bunt J, Li J, Alkuraya F, Alobeid E, Richards L, Ullian E, Sherr E. C12ORF57: a novel principal regulator of synaptic AMPA currents and excitatory neuronal homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.632037. [PMID: 39974932 PMCID: PMC11838199 DOI: 10.1101/2025.01.08.632037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Objective Excitatory neuronal homeostasis is crucial for neuronal survival, circuit function, and plasticity. Disruptions in this form of homeostasis are believed to underpin a variety of neuronal conditions including intellectual disability, epilepsy, and autism. However, the underlying genetic and molecular mechanisms maintaining this homeostasis remain poorly understood. Biallelic recurrent loss of function mutations in C12ORF57 , an evolutionarily conserved X amino acid novel open reading frame, underlie Temtamy syndrome (TS)-a neurodevelopmental disorder characterized by epilepsy, dysgenesis of the corpus callosum, and severe intellectual disability. Methods Through multiple lines of inquiry, we establish that C12ORF57/GRCC10 plays an unexpected central role in synaptic homeostatic downscaling in response to elevated activity, uncovering a novel mechanism for neuronal excitatory homeostasis. To probe these mechanisms, we developed a new knockout (KO) mouse model of the gene's murine ortholog, Grcc10 as well as cellular and in vitro assays. Results Grcc10 KO mice exhibit the characteristic phenotypic features seen in human TS patients, including increased epileptiform activity. Corresponding with the enhanced seizure susceptibility, hippocampal neurons in these mice exhibited significantly increased AMPA receptor expression levels and higher amplitude of miniature excitatory postsynaptic currents (mEPSCs). We further found that GRCC10/C12ORF57 modulates the activity of calcium/calmodulin dependent kinase 4 (CAMK4) and thereby regulates the expression of CREB and ARC. Interpretation Our study suggests through this novel mechanism, deletion of Grcc10 disrupts the characteristic synaptic AMPA receptor downscaling that accompanies increased activity in glutamatergic neurons.
Collapse
|
5
|
Mao Y, Wu J, Weng J, Li M, Xiong Y, Gu W, Jiang R, Pang R, Lin X, Tang D. Inter-view contrastive learning and miRNA fusion for lncRNA-protein interaction prediction in heterogeneous graphs. Brief Bioinform 2025; 26:bbaf148. [PMID: 40194558 PMCID: PMC11975365 DOI: 10.1093/bib/bbaf148] [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: 12/06/2024] [Revised: 02/19/2025] [Accepted: 03/16/2025] [Indexed: 04/09/2025] Open
Abstract
Predicting long non-coding RNA (lncRNA)-protein interactions is essential for understanding biological processes and discovering new therapeutic targets. In this study, we propose a novel model based on inter-view contrastive learning and miRNA fusion for lncRNA-protein interaction (LPI) prediction, called ICMF-LPI, which utilizes a heterogeneous information network to enhance LPI prediction. The model integrates miRNA as a mediator, constructing an lncRNA-miRNA-protein network, and employs metapath to extract diverse relationships from heterogeneous graphs. By fusing miRNA-related information and leveraging contrastive learning across inter-views, ICMF-LPI effectively captures potential interactions. Experimental results, including five-fold cross-validation, demonstrate the model's superior performance compared to several state-of-the-art methods, with significant improvements in the area under the receiver operating characteristic curve and the area under the precision-recall curve metrics. Notably, even when direct LPI connections are excluded, ICMF-LPI still achieves competitive predictive accuracy, performing comparably or better than some existing models. This demonstrates that the proposed model is effective in scenarios where direct interaction data are unavailable. This approach offers a promising direction for developing predictive models in bioinformatics, particularly in challenging conditions.
Collapse
Affiliation(s)
- Yijun Mao
- College of Mathematics and Informatics, South China Agricultural University, 483 Wushan Road, Tianhe District, GuangZhou 510642, China
- National Key Laboratory of Data Space Technology and System, 3 Minzhuang Road, Haidian District, Beijing 100195, China
| | - Jiale Wu
- College of Mathematics and Informatics, South China Agricultural University, 483 Wushan Road, Tianhe District, GuangZhou 510642, China
| | - Jian Weng
- College of Cyber Security, Jinan University, 601 West Huangpu Avenue, Tianhe District, GuangZhou 510632, China
| | - Ming Li
- College of Cyber Security, Jinan University, 601 West Huangpu Avenue, Tianhe District, GuangZhou 510632, China
| | - Yunyan Xiong
- School of Computer and Infomation Engineering, Guangdong Polytechnic of Industry and Commerce, 1098 North Guangzhou Avenue, Tianhe District, GuangZhou 510510, China
| | - Wanrong Gu
- College of Mathematics and Informatics, South China Agricultural University, 483 Wushan Road, Tianhe District, GuangZhou 510642, China
| | - Rongjin Jiang
- Department of Digital process, Wens Foodstuff Group Co., Ltd, 9 Dongdi North Road, Xinxing County, YunFu 527400, China
| | - Rui Pang
- State Key Laboratory of Green Pesticide, College of Plant Protection, South China Agricultural University, 483 Wushan Road, Tianhe District, GuangZhou 510642, China
| | - Xudong Lin
- College of Mathematics and Informatics, South China Agricultural University, 483 Wushan Road, Tianhe District, GuangZhou 510642, China
| | - Deyu Tang
- College of Mathematics and Informatics, South China Agricultural University, 483 Wushan Road, Tianhe District, GuangZhou 510642, China
| |
Collapse
|
6
|
Cornelissen FMG, He Z, Ciputra E, de Haas RR, Beumer‐Chuwonpad A, Noske D, Vandertop WP, Piersma SR, Jiménez CR, Murre C, Westerman BA. The translatome of glioblastoma. Mol Oncol 2025; 19:716-740. [PMID: 39417309 PMCID: PMC11887679 DOI: 10.1002/1878-0261.13743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 10/19/2024] Open
Abstract
Glioblastoma (GB), the most common and aggressive brain tumor, demonstrates intrinsic resistance to current therapies, resulting in poor clinical outcomes. Cancer progression can be partially attributed to the deregulation of protein translation mechanisms that drive cancer cell growth. In this study, we present the translatome landscape of GB as a valuable data resource. Eight patient-derived GB sphere cultures (GSCs) were analyzed using ribosome profiling and messenger RNA (mRNA) sequencing. We investigated inter-cell-line differences through differential expression analysis at both the translatome and transcriptome levels. Translational changes post-radiotherapy were assessed at 30 and 60 min. The translation of non-coding RNAs (ncRNAs) was validated using in-house and public mass spectrometry (MS) data, whereas RNA expression was confirmed by quantitative PCR (qPCR). Our findings demonstrate that ribosome sequencing provides more detailed information than MS or transcriptional analyses. Transcriptional similarities among GSCs correlate with translational similarities, aligning with previously defined subtypes such as proneural and mesenchymal. Additionally, we identified a broad spectrum of open reading frame types in both coding and non-coding mRNA regions, including long non-coding RNAs (lncRNAs) and pseudogenes undergoing active translation. Translation of ncRNAs into peptides was independently confirmed by in-house data and external MS data. We also observed that translational regulation of histones (downregulated) and splicing factors (upregulated) occurs in response to radiotherapy. These data offer new insights into genome-wide protein synthesis, identifying translationally regulated genes and alternative translation initiation sites in GB under normal and radiotherapeutic conditions, providing a rich resource for GB research. Further functional validation of differentially expressed genes after radiotherapy is needed. Understanding translational control in GB can reveal mechanistic insights and identify currently unknown biomarkers, ultimately enhancing the diagnosis and treatment of this aggressive brain cancer.
Collapse
Affiliation(s)
- Fleur M. G. Cornelissen
- Department of Molecular BiologyUniversity of California, San DiegoLa JollaCAUSA
- Department of NeurosurgeryAmsterdam UMC, Location VUMC, Cancer CenterAmsterdamThe Netherlands
| | - Zhaoren He
- Department of Molecular BiologyUniversity of California, San DiegoLa JollaCAUSA
| | - Edward Ciputra
- Department of NeurosurgeryAmsterdam UMC, Location VUMC, Cancer CenterAmsterdamThe Netherlands
| | - Richard R. de Haas
- OncoProteomics Laboratory, Cancer Center AmsterdamAmsterdam UMCThe Netherlands
| | | | - David Noske
- Department of NeurosurgeryAmsterdam UMC, Location VUMC, Cancer CenterAmsterdamThe Netherlands
| | - W. Peter Vandertop
- Department of NeurosurgeryAmsterdam UMC, Location VUMC, Cancer CenterAmsterdamThe Netherlands
| | - Sander R. Piersma
- OncoProteomics Laboratory, Cancer Center AmsterdamAmsterdam UMCThe Netherlands
| | - Connie R. Jiménez
- OncoProteomics Laboratory, Cancer Center AmsterdamAmsterdam UMCThe Netherlands
| | - Cornelis Murre
- Department of Molecular BiologyUniversity of California, San DiegoLa JollaCAUSA
| | - Bart A. Westerman
- Department of NeurosurgeryAmsterdam UMC, Location VUMC, Cancer CenterAmsterdamThe Netherlands
| |
Collapse
|
7
|
Suzuki A. The early days of transcriptome sequencing and functional genomics. Nat Rev Genet 2025; 26:155. [PMID: 39875645 DOI: 10.1038/s41576-025-00814-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Affiliation(s)
- Ayako Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan.
| |
Collapse
|
8
|
Hassan FU, Safdar M, Younus M, Arain MA. Regulation of energy metabolism by non-coding RNAs in livestock species: a review. J Comp Physiol B 2025; 195:1-12. [PMID: 39638953 DOI: 10.1007/s00360-024-01596-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 10/17/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024]
Abstract
The optimisation of livestock production relies on efficient energy metabolism. This review focused on elaborate regulatory processes governed by non-coding RNAs (ncRNAs), such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). It explores the complex energy metabolism processes in livestock, elucidating the functions of ncRNAs in the expression of genes and pathways. miRNAs have been identified as significant regulators of glycolysis and glucose metabolism, whereas lncRNAs are known to affect adipogenesis and mitochondrial activity. Moreover, circRNAs have a substantial influence on the regulation of energy. In addition, this is not only enriching non-coding RNA-mediated energy control but also sheds light on possible applications. It is derived from its ability to condense complex molecular systems, thereby offering crucial insights to researchers. Through a comprehensive analysis of the intricate relationship between ncRNAs and energy metabolism, the information of this review provides a valuable framework for the implementation of focused interventions that hold the potential to significantly enhance the efficiency of livestock production.
Collapse
Affiliation(s)
- Faiz-Ul Hassan
- Department of Breeding and Genetics, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, 63000, Pakistan.
| | - Muhammad Safdar
- Department of Breeding and Genetics, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, 63000, Pakistan
| | - Muhammad Younus
- Department of Zoology, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, 63000, Pakistan
| | - Muhammad Asif Arain
- Faculty of Veterinary and Animal Sciences, Water and Marine Sciences, Lasbela University of Agriculture, Uthal, 90150, Balochistan, Pakistan
| |
Collapse
|
9
|
Yokoo T, Watanabe K, Iida K, Nakachi Y, Suzuki H, Shimano H, Takashima S, Okazaki Y, Yamada N, Toyoshima H. Betagenin ameliorates diabetes by inducing insulin secretion and β-cell proliferation. J Biol Chem 2025; 301:108202. [PMID: 39826690 PMCID: PMC11870162 DOI: 10.1016/j.jbc.2025.108202] [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: 08/15/2024] [Revised: 12/22/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025] Open
Abstract
Recent success with the use of glucagon-like peptide-1 (GLP-1) receptor analogs and dipeptidyl peptidase-4 inhibitors for the treatment of patients with diabetes has highlighted the role of the intestine as an endocrine organ. Gut-derived hormones, including glucagon-like peptide-1, glucose-dependent insulinotropic polypeptide, and ghrelin, have important roles in the control of energy metabolism and food intake, and are associated with the metabolic syndrome. In this study, we isolated and identified a new intestine-derived hormone, betagenin, and showed that it stimulates insulin secretion and β-cell proliferation and suppresses β-cell apoptosis. Adenovirus-mediated expression of betagenin restored the blood glucose concentrations and hemoglobin A1c (HbA1c) levels of mice with streptozotocin-induced diabetes to normal and increased their β-cell mass. Transgenic mice overexpressing betagenin exhibited more than three-fold higher β-cell mass than WT mass, whereas that of KO mice was four-fold lower. A synthetic peptide representing the sequence of purified and secreted betagenin enhanced glucose-dependent insulin secretion in human and mouse pancreatic islets and stimulated the proliferation of the pancreatic β-cell line MIN6 through extracellular signal-regulated kinase 1/2-dependent signaling. The intravenous administration of this peptide to streptozotocin mice stimulated the proliferation of pancreatic β-cells in vivo, and the intraperitoneal administration of betagenin ameliorated diabetes and restored β-cell mass. These results indicate that betagenin may reduce blood glucose concentration and induce β-cell regeneration in patients with diabetes.
Collapse
Affiliation(s)
- Tomotaka Yokoo
- Division of Experimental Animal, Hidaka Branch, Biomedical Research Center, Saitama Medical University, Saitama, Japan; Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Kazuhisa Watanabe
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan; Division of Human Genetics, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Kaoruko Iida
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan; Department of Food and Nutrition Science, Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo, Japan
| | - Yutaka Nakachi
- Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan; Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroaki Suzuki
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Hitoshi Shimano
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasushi Okazaki
- Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan; Diagnostics and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Nobuhiro Yamada
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Hideo Toyoshima
- Division of Experimental Animal, Hidaka Branch, Biomedical Research Center, Saitama Medical University, Saitama, Japan; Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan.
| |
Collapse
|
10
|
Gao R, Liu M, Yang H, Shen Y, Xia N. Epigenetic regulation in coronary artery disease: from mechanisms to emerging therapies. Front Mol Biosci 2025; 12:1548355. [PMID: 39959304 PMCID: PMC11825346 DOI: 10.3389/fmolb.2025.1548355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/13/2025] [Indexed: 02/18/2025] Open
Abstract
Atherosclerosis, the primary cause of coronary artery disease (CAD), remains a leading global cause of mortality. It is characterized by the accumulation of cholesterol-rich plaques and inflammation, which narrow the coronary arteries and increase the risk of rupture. To elucidate this complex biological process and improve therapeutic strategies, CAD has been extensively explored from an epigenetic perspective over the past two decades. Epigenetics is a field investigating heritable alterations in gene expression without DNA sequence changes, such as DNA methylation, histone modifications, and non-coding RNAs. Increasing evidence has indicated that the development of CAD is significantly influenced by epigenetic changes. Meanwhile, the impact of epigenetics in CAD is now transitioning from pathophysiology to therapeutics. Focusing on the key epigenetic enzymes and their target genes will help to facilitate the diagnosis and treatment of CAD. This review synthesizes novel epigenetic insights into CAD, addressing the pathological processes, key molecular mechanisms, and potential biomarkers. Furthermore, we discuss emerging therapeutic strategies targeting epigenetic pathways. By focusing on pivotal enzymes and their associated genes, this work aims to advance CAD diagnostics and interventions.
Collapse
Affiliation(s)
- Rui Gao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meilin Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haoyi Yang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuhan Shen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ni Xia
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Wang J, Liu M, Zhao J, Hu P, Gao L, Tian S, Zhang J, Liu H, Xu X, He Z. Oxidative stress and dysregulated long noncoding RNAs in the pathogenesis of Parkinson's disease. Biol Res 2025; 58:7. [PMID: 39871377 PMCID: PMC11770960 DOI: 10.1186/s40659-025-00585-7] [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: 06/15/2023] [Accepted: 01/07/2025] [Indexed: 01/29/2025] Open
Abstract
Parkinson's disease (PD) is a progressive age-related neurodegenerative disease whose annual incidence is increasing as populations continue to age. Although its pathogenesis has not been fully elucidated, oxidative stress has been shown to play an important role in promoting the occurrence and development of the disease. Long noncoding RNAs (lncRNAs), which are more than 200 nucleotides in length, are also involved in the pathogenesis of PD at the transcriptional level via epigenetic regulation, or at the post-transcriptional level by participating in physiological processes, including aggregation of the α-synuclein, mitochondrial dysfunction, oxidative stress, calcium stabilization, and neuroinflammation. LncRNAs and oxidative stress are correlated during neurodegenerative processes: oxidative stress affects the expression of multiple lncRNAs, while lncRNAs regulate many genes involved in oxidative stress responses. Oxidative stress and lncRNAs also affect other processes associated with neurodegeneration, including mitochondrial dysfunction and increased neuroinflammation that lead to neuronal death. Therefore, modulating the levels of specific lncRNAs may alleviate pathological oxidative damage and have neuroprotective effects. This review discusses the general mechanisms of oxidative stress, pathological mechanism underlying the role of oxidative stress in the pathogenesis of PD, and teases out the mechanisms through which lncRNAs regulate oxidative stress during PD pathogenesis, as well as identifies the possible neuroprotective mechanisms of lncRNAs. Reviewing published studies will help us further understand the mechanisms underlying the role of lncRNAs in the oxidative stress process in PD and to identify potential therapeutic strategies for PD.
Collapse
Affiliation(s)
- Jialu Wang
- Department of Neurology, First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Meitong Liu
- Department of Neurology, Fourth Affiliated Hospital of China Medical University, No.4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China
| | - Jiuhan Zhao
- Department of Neurology, First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Pan Hu
- Department of Neurology, First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Lianbo Gao
- Department of Neurology, Fourth Affiliated Hospital of China Medical University, No.4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China
| | - Shen Tian
- Department of Neurology, Fourth Affiliated Hospital of China Medical University, No.4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China
| | - Jin Zhang
- Department of Neurology, Fourth Affiliated Hospital of China Medical University, No.4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China
| | - Huayan Liu
- Department of Neurology, First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Xiaoxue Xu
- Department of Neurology, First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.
| | - Zhenwei He
- Department of Neurology, Fourth Affiliated Hospital of China Medical University, No.4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China.
| |
Collapse
|
12
|
Sarkar N, Kumar A. Paradigm shift: microRNAs interact with target gene promoters to cause transcriptional gene activation or silencing. Exp Cell Res 2025; 444:114372. [PMID: 39662662 DOI: 10.1016/j.yexcr.2024.114372] [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: 08/18/2024] [Revised: 11/29/2024] [Accepted: 12/08/2024] [Indexed: 12/13/2024]
Abstract
MicroRNAs (miRNAs/miRs) are small (18-25 nucleotides in length), endogenous, non-coding RNAs that typically repress gene expression by interacting with the 3'untranslated regions (3'UTRs) of target mRNAs in the cytoplasm. While most of the scientific community still views miRNAs as repressors of gene expression, this review highlights their non-canonical novel role in the nucleus as activators or silencers of target gene transcription through miRNA-promoter interaction. The mechanistic details of the transcriptional role of miRNAs are yet to be elucidated, however, they can be explained by prospective models. In this review, we aim to discuss the different examples of transcriptional regulation by miRNAs and their possible mechanism of action, thereby offering a comprehensive perspective on the role of miRNAs in gene regulation and their importance in health and diseases.
Collapse
Affiliation(s)
- Neelanjana Sarkar
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India.
| | - Arun Kumar
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
13
|
Nobusada T, Yip C, Agrawal S, Severin J, Abugessaisa I, Hasegawa A, Hon C, Ide S, Koido M, Kondo A, Masuya H, Oki S, Tagami M, Takada T, Terao C, Thalhath N, Walker S, Yasuzawa K, Shin J, de Hoon ML, Carninci P, Kawaji H, Kasukawa T. Update of the FANTOM web resource: enhancement for studying noncoding genomes. Nucleic Acids Res 2025; 53:D419-D424. [PMID: 39592010 PMCID: PMC11701582 DOI: 10.1093/nar/gkae1047] [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: 09/17/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
The FANTOM web resource (https://fantom.gsc.riken.jp/) has been a unique resource for studying mammalian genomes, which is built on the research activities conducted in the international collaborative project FANTOM (Functional ANnoTation Of the Mammalian genome). In recent updates, we expanded annotations for long non-coding RNAs (lncRNAs) and transcribed cis-regulatory elements (CREs). The former was derived from the large-scale lncRNA perturbations in induced pluripotent stem cells (iPSCs) and integrative analysis of Hi-C data conducted in the sixth iteration of the project (FANTOM6). The resulting annotations of lncRNAs, according to the impact on cellular and molecular phenotypes and the potential RNA-chromatin interactions, are accessible via the interactive ZENBU-Reports framework. The latter involves a new platform, fanta.bio (https://fanta.bio/), which collects transcribed CREs identified via use of an extended dataset of CAGE profiles. The CREs, with their annotations including genetic and epigenetic information, are accessible via a dedicated interface as well as the UCSC Genome Browser Database. These updates offer enhanced opportunities to investigate the functions of non-coding regions within mammalian genomes.
Collapse
Affiliation(s)
- Tomoe Nobusada
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Chi Wai Yip
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Saumya Agrawal
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Jessica Severin
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Imad Abugessaisa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Akira Hasegawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Chung Chau Hon
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Satoru Ide
- Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Masaru Koido
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- Graduate School of Frontier Sciences, The University of Tokyo, Tokyo 277-0882, Japan
| | - Atsushi Kondo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Hiroshi Masuya
- RIKEN BioResource Research Center, Tsukuba, Ibaraki 305-0074, Japan
| | - Shinya Oki
- Kumamoto University, Kumamoto 860-0811, Japan
| | - Michihira Tagami
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Toyoyuki Takada
- RIKEN BioResource Research Center, Tsukuba, Ibaraki 305-0074, Japan
| | - Chikashi Terao
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka 420-8527, Japan
| | - Nishad Thalhath
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Scott Walker
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Kayoko Yasuzawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Jay W Shin
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Michiel J L de Hoon
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Piero Carninci
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Hideya Kawaji
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Takeya Kasukawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| |
Collapse
|
14
|
Ma R, Wang M, Ma Q, Zhang Y, Shang F, Wang R, Zhang Y. LncRNA MSTRG.14227.1 regulates the morphogenesis of secondary hair follicles in Inner Mongolia cashmere goats via targeting ADAMTS3 by sponging chi-miR-433. J Anim Sci 2025; 103:skae382. [PMID: 39715333 PMCID: PMC11808577 DOI: 10.1093/jas/skae382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 12/22/2024] [Indexed: 12/25/2024] Open
Abstract
The cashmere goat is a type of livestock primarily known for its cashmere. Cashmere has a soft hand feel and good luster. It is a vital raw material in the textile industry, possessing significant economic value. Improving the yield and quality of cashmere can accelerate the development of the cashmere industry and increase the incomes of farmers and herdsmen. The embryonic stage is the main stage of the formation of hair follicle structure, which directly affects the yield and quality of cashmere. With the rapid advancements in modern molecular technology and high-throughput sequencing, many signaling molecules have been identified as playing critical roles in hair follicle development. Long non-coding RNA (lncRNA), which lacks protein-coding ability and exceeds 200 nucleotides in length, has been discovered to play a role in hair follicle development. In this study, the lncRNA MSTRG.14227.1, which is associated with the morphogenesis of secondary hair follicles, was screened and identified based on previously established lncRNA expression profiles derived from skin tissues of cashmere goats at different embryonic stages. This lncRNA has been shown to inhibit the proliferation and migration of dermal fibroblasts. Furthermore, we confirmed through bioinformatics analysis and dual-luciferase reporter assays that lncRNA MSTRG.14227.1 can function as a sponge for chi-miR-433, thereby alleviating the inhibitory effect of chi-miR-433 on its target gene ADAMTS3. In conclusion, the results of this study suggest that lncRNA MSTRG.14227.1 can inhibit the morphogenesis of secondary hair follicles through the chi-miR-433/ADAMTS3 signaling axis.
Collapse
Affiliation(s)
- Rong Ma
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Min Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Qing Ma
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Yiming Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Fangzheng Shang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Ruijun Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Yanjun Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Mutton Sheep Genetics and Breeding, Ministry of Agriculture, Hohhot, China
- Laboratory of Goat and Sheep Genetics, Breeding and Reproduction in Inner Mongolia Autonomous Region, Hohhot, China
| |
Collapse
|
15
|
Yuan L, Yin L, Lin X, Li J, Liang P, Jiang B. Revealing the Complex Interaction of Noncoding RNAs, Sirtuin Family, and Mitochondrial Function. J Gene Med 2025; 27:e70007. [PMID: 39842441 DOI: 10.1002/jgm.70007] [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: 04/23/2024] [Revised: 10/23/2024] [Accepted: 12/04/2024] [Indexed: 01/24/2025] Open
Abstract
Mitochondria are key organelles that perform and coordinate various metabolic processes in the cell, and their homeostasis is essential for the maintenance of eukaryotic life. To maintain mitochondrial homeostasis and cellular health, close communication between noncoding RNAs (ncRNAs) and proteins is required. For example, there are numerous crosstalk between ncRNAs and the sirtuin (SIRT1-7) family, which is a group of nicotinamide adenine dinucleotides (NAD(+))-dependent Type III deacetylases. NcRNAs are involved in the regulation of gene expression of sirtuin family members, and deacetylation of sirtuin family members can also influence the generation of ncRNAs. This review focuses on the relationship between the two mentioned above and summarizes the impact of their interactions on mitochondrial metabolism, oxidative stress, mitochondrial apoptotic pathways, mitochondrial biogenesis, mitochondrial dynamics, and other mitochondria-related pathophysiological processes. Finally, the review also describes targeted and appropriate treatment strategies. In conclusion, we provide an overview of the ncRNA-sirtuins/mitochondria relationship that could provide a reference for related research in the mitochondrial field and help the future development of new biomedical applications in this area.
Collapse
Affiliation(s)
- Ludong Yuan
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Leijing Yin
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Xiaofang Lin
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Jing Li
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Pengfei Liang
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bimei Jiang
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| |
Collapse
|
16
|
Kotey SK, Tan X, Kinser AL, Liu L, Cheng Y. Host Long Noncoding RNAs as Key Players in Mycobacteria-Host Interactions. Microorganisms 2024; 12:2656. [PMID: 39770858 PMCID: PMC11728548 DOI: 10.3390/microorganisms12122656] [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: 11/23/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Mycobacterial infections, caused by various species within the Mycobacterium genus, remain one of the main challenges to global health across the world. Understanding the complex interplay between the host and mycobacterial pathogens is essential for developing effective diagnostic and therapeutic strategies. Host long noncoding RNAs (lncRNAs) have emerged as key regulators in cellular response to bacterial infections within host cells. This review provides an overview of the intricate relationship between mycobacterial infections and host lncRNAs in the context of Mycobacterium tuberculosis and non-tuberculous mycobacterium (NTM) infections. Accumulation of evidence indicates that host lncRNAs play a critical role in regulating cellular response to mycobacterial infection within host cells, such as macrophages, the primary host cells for mycobacterial intracellular survival. The expression of specific host lncRNAs has been implicated in the pathogenesis of mycobacterial infections, providing potential targets for the development of novel host-directed therapies and biomarkers for TB diagnosis. In summary, this review aims to highlight the current state of knowledge regarding the involvement of host lncRNAs in mycobacterial infections. It also emphasizes their potential application as novel diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Stephen K. Kotey
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA; (S.K.K.); (X.T.); (A.L.K.)
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Xuejuan Tan
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA; (S.K.K.); (X.T.); (A.L.K.)
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Audrey L. Kinser
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA; (S.K.K.); (X.T.); (A.L.K.)
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Lin Liu
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078, USA;
- Department of Physiological Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Yong Cheng
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA; (S.K.K.); (X.T.); (A.L.K.)
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078, USA;
| |
Collapse
|
17
|
Ismail M, Fadul MM, Taha R, Siddig O, Elhafiz M, Yousef BA, Jiang Z, Zhang L, Sun L. Dynamic role of exosomal long non-coding RNA in liver diseases: pathogenesis and diagnostic aspects. Hepatol Int 2024; 18:1715-1730. [PMID: 39306594 DOI: 10.1007/s12072-024-10722-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 08/15/2024] [Indexed: 12/11/2024]
Abstract
BACKGROUND Liver disease has emerged as a significant health concern, characterized by high rates of morbidity and mortality. Circulating exosomes have garnered attention as important mediators of intercellular communication, harboring protein and stable mRNAs, microRNAs, and long non-coding RNAs (lncRNA). This review highlights the involvement of exosomal lncRNA in the pathogenesis and diagnosis of various liver diseases. Notably, exosomal lncRNAs exhibit therapeutic potential as targets for conditions including hepatic carcinoma, hepatic fibrosis, and hepatic viral infections. METHOD An online screening process was employed to identify studies investigating the association between exosomal lncRNA and various liver diseases. RESULT Our study revealed a diverse array of lncRNAs carried by exosomes, including H19, Linc-ROR, VLDLR, MALAT1, DANCR, HEIH, ENSG00000248932.1, ENST00000457302.2, ZSCAN16-AS1, and others, exhibiting varied levels across different liver diseases compared to normal liver tissue. These exosomal-derived lncRNAs are increasingly recognized as pivotal biomarkers for diagnosing and prognosticating liver diseases, supported by emerging evidence. However, the precise mechanisms underlying the involvement of certain exosomal lncRNAs remain incompletely understood. Furthermore, the combined analysis of serum exosomes using ENSG00000258332.1, LINC00635, and serum AFP may serve as novel and valuable biomarker for HCC. Clinically, exosomal ATB expression is upregulated in HCC, while exosomal HEIH and RP11-513I15.6 have shown potential for distinguishing HCC related to HCV infection. CONCLUSION The lack of reliable biomarkers for liver diseases, coupled with the high specificity and sensitivity of exosomal lncRNA and its non-invasive detection, promotes exploring their role in pathogenesis and biomarker for diagnosis, prognosis, and response to treatment liver diseases.
Collapse
Affiliation(s)
- Mohammed Ismail
- Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
- Department of Pharmacology, Faculty of Medicine and Health Science, Dongola University, Dongola, Sudan
| | - Missaa M Fadul
- Department of Pharmacology, Faculty of Medicine and Health Science, Dongola University, Dongola, Sudan
| | - Reham Taha
- Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Orwa Siddig
- Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Muhanad Elhafiz
- Department of Pharmacology, Faculty of Pharmacy, Omdurman Islamic University, Khartoum, Sudan
| | - Bashir A Yousef
- Department of Pharmacology, Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | - Zhenzhou Jiang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Luyong Zhang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.
- Centre for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Lixin Sun
- Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
18
|
Chen LL, Kim VN. Small and long non-coding RNAs: Past, present, and future. Cell 2024; 187:6451-6485. [PMID: 39547208 DOI: 10.1016/j.cell.2024.10.024] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/13/2024] [Accepted: 10/15/2024] [Indexed: 11/17/2024]
Abstract
Since the introduction of the central dogma of molecular biology in 1958, various RNA species have been discovered. Messenger RNAs transmit genetic instructions from DNA to make proteins, a process facilitated by housekeeping non-coding RNAs (ncRNAs) such as small nuclear RNAs (snRNAs), ribosomal RNAs (rRNAs), and transfer RNAs (tRNAs). Over the past four decades, a wide array of regulatory ncRNAs have emerged as crucial players in gene regulation. In celebration of Cell's 50th anniversary, this Review explores our current understanding of the most extensively studied regulatory ncRNAs-small RNAs and long non-coding RNAs (lncRNAs)-which have profoundly shaped the field of RNA biology and beyond. While small RNA pathways have been well documented with clearly defined mechanisms, lncRNAs exhibit a greater diversity of mechanisms, many of which remain unknown. This Review covers pivotal events in their discovery, biogenesis pathways, evolutionary traits, action mechanisms, functions, and crosstalks among ncRNAs. We also highlight their roles in pathophysiological contexts and propose future research directions to decipher the unknowns of lncRNAs by leveraging lessons from small RNAs.
Collapse
Affiliation(s)
- Ling-Ling Chen
- Key Laboratory of RNA Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China; School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; New Cornerstone Science Laboratory, Shenzhen, China.
| | - V Narry Kim
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Korea; School of Biological Sciences, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
19
|
Wang M, Ma R, Ma Q, Ma B, Shang F, Lv Q, Wang Z, Wang R, Su R, Zhao Y, Zhang Y. Role of LncRNA MSTRG.20890.1 in Hair Follicle Development of Cashmere Goats. Genes (Basel) 2024; 15:1392. [PMID: 39596592 PMCID: PMC11593464 DOI: 10.3390/genes15111392] [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: 09/17/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The cashmere goat is a biological resource that mainly produces cashmere. Cashmere has a soft hand feel and good luster, with high economic value. The quality and yield of cashmere are determined by the process of hair follicle development during the embryonic period. METHODS In this study, the skin of the Inner Mongolia cashmere goat at different embryonic stages (45, 55, 65, and 75d) was collected, and the differentially expressed lncRNA MSTRG.20890.1 at 75d was obtained by screening. Dual luciferase reporter gene system, qRT-PCR, and EDU experiments were used to verify further the regulatory role and molecular mechanism of the lncRNA in dermal fibroblasts. RESULTS Based on the transcriptome database of Inner Mongolia cashmere goat skin at different embryonic stages, which was previously constructed by our group, according to the characteristics of hair follicle development in the embryonic stage, we screened out the lncRNA MSTRG.20890.1 that was down-expressed on the 75-SHFINI day of the embryonic stage. We found that lncRNA MSTRG.20890.1 was mainly located in the cytoplasm of cells, and it could inhibit the proliferation and directional migration of dermal fibroblasts through the chi-miR-24-3p/ADAMTS3 signaling axis, thereby inhibiting the formation of dermal papilla structure at embryonic stage. CONCLUSIONS This study revealed that lncRNA MSTRG.20890.1 regulated secondary hair follicle morphogenesis and development in cashmere goats through the chi-miR-24-3p/ADAMTS3 signaling axis.
Collapse
Affiliation(s)
- Min Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.W.); (R.M.)
| | - Rong Ma
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.W.); (R.M.)
| | - Qing Ma
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.W.); (R.M.)
| | - Bingjie Ma
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.W.); (R.M.)
| | - Fangzheng Shang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.W.); (R.M.)
| | - Qi Lv
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.W.); (R.M.)
| | - Zhiying Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.W.); (R.M.)
| | - Ruijun Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.W.); (R.M.)
| | - Rui Su
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.W.); (R.M.)
| | - Yanhong Zhao
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.W.); (R.M.)
| | - Yanjun Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.W.); (R.M.)
- Key Laboratory of Mutton Sheep Genetics and Breeding, Ministry of Agriculture, Hohhot 010018, China
- Laboratory of Goat and Sheep Genetics, Breeding and Reproduction in Inner Mongolia Autonomous Region, Hohhot 010018, China
| |
Collapse
|
20
|
Oguntoyinbo IO, Goyal R. The Role of Long Intergenic Noncoding RNA in Fetal Development. Int J Mol Sci 2024; 25:11453. [PMID: 39519006 PMCID: PMC11546696 DOI: 10.3390/ijms252111453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The role of long intergenic noncoding RNAs (lincRNAs) in fetal development has emerged as a significant area of study, challenging the traditional protein-centric view of gene expression. While messenger RNAs (mRNAs) have long been recognized for their role in encoding proteins, recent advances have illuminated the critical functions of lincRNAs in various biological processes. Initially identified through high-throughput sequencing technologies, lincRNAs are transcribed from intergenic regions between protein-coding genes and exhibit unique regulatory functions. Unlike mRNAs, lincRNAs are involved in complex interactions with chromatin and chromatin-modifying complexes, influencing gene expression and chromatin structure. LincRNAs are pivotal in regulating tissue-specific development and embryogenesis. For example, they are crucial for proper cardiac, neural, and reproductive system development, with specific lincRNAs being associated with organogenesis and differentiation processes. Their roles in embryonic development include regulating transcription factors and modulating chromatin states, which are essential for maintaining developmental programs and cellular identity. Studies using RNA sequencing and genetic knockout models have highlighted the importance of lincRNAs in processes such as cell differentiation, tissue patterning, and organ development. Despite their functional significance, the comprehensive annotation and understanding of lincRNAs remain limited. Ongoing research aims to elucidate their mechanisms of action and potential applications in disease diagnostics and therapeutics. This review summarizes current knowledge on the functional roles of lincRNAs in fetal development, emphasizing their contributions to tissue-specific gene regulation and developmental processes.
Collapse
Affiliation(s)
- Ifetoluwani Oluwadunsin Oguntoyinbo
- School of Animal and Comparative Biomedical Sciences, College of Agriculture, Life & Environmental Sciences, University of Arizona, Tucson, AZ 85721, USA;
| | - Ravi Goyal
- Department of Obstetrics and Gynecology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
21
|
Yu C, Qiu M, Xiong X, Peng H, Han S, Song X, Hu C, Zhang Z, Xia B, Chen J, Zhu S, Yang L, Li W, Yin H, Zhao J, Lin Z, Liu Y, Yang C. Integrative analysis of RNA-seq and Ribo-seq reveals that lncRNA-GRN regulates chicken follicular atresia through miR-103-3p/FBXW7 axis and encoding peptide. Int J Biol Macromol 2024; 278:135051. [PMID: 39182874 DOI: 10.1016/j.ijbiomac.2024.135051] [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: 05/21/2024] [Revised: 07/30/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
Follicular atresia in chickens seriously reduced the egg production and economic benefits of chickens. LncRNA plays a key role in the process of follicular atresia. In this study, RNA-seq and Ribo-seq were performed on normal and atretic follicles of Dahen broilers to screen out lncRNAs that may regulate follicle atresia, and to study the molecular mechanisms of their regulation. GRN granulin precursor (lncGRN, ID: 101748909) was highly expressed in atretic follicles with translational ability. A molecular regulatory network of lncGRN/miR-103-3p/FBXW7 was constructed through bioinformatics analysis and dual luciferase reporting. LncGRN promoted the expression of FBXW7 by adsorption of miR-103-3p, thereby inhibiting the proliferation of chicken granulosa cells (GCs), promoting apoptosis of chicken GCs and inhibiting steroid hormone synthesis thus induced follicular atresia. Meanwhile, we also found a micropeptide named GRN-122aa derived by lncGRN which can promote follicular atresia. In conclusion, our study found that lncGRN promoted follicular atresia through the lncGRN/miR-103-3p/FBXW7 axis and the translation micropeptide GRN-122aa. This study provided new insight into the post-transcriptional regulation mechanism of lncGRN suggesting that lncGRN may act as a potential to regulate chicken follicle development, and provided a theoretical argument for further improving the egg production of chickens through molecular breeding.
Collapse
Affiliation(s)
- Chunlin Yu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Mohan Qiu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Xia Xiong
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Han Peng
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Shunshun Han
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Xiaoyan Song
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Chenming Hu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Zengrong Zhang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Bo Xia
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Jialei Chen
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Shiliang Zhu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Li Yang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Wen Li
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Huadong Yin
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Jing Zhao
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Zhongzhen Lin
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Yiping Liu
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Chaowu Yang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China.
| |
Collapse
|
22
|
Das D, Podder S. Microscale marvels: unveiling the macroscopic significance of micropeptides in human health. Brief Funct Genomics 2024; 23:624-638. [PMID: 38706311 DOI: 10.1093/bfgp/elae018] [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: 01/04/2024] [Revised: 04/07/2024] [Accepted: 04/15/2024] [Indexed: 05/07/2024] Open
Abstract
Non-coding RNA encodes micropeptides from small open reading frames located within the RNA. Interestingly, these micropeptides are involved in a variety of functions within the body. They are emerging as the resolving piece of the puzzle for complex biomolecular signaling pathways within the body. Recent studies highlight the pivotal role of small peptides in regulating important biological processes like DNA repair, gene expression, muscle regeneration, immune responses, etc. On the contrary, altered expression of micropeptides also plays a pivotal role in the progression of various diseases like cardiovascular diseases, neurological disorders and several types of cancer, including colorectal cancer, hepatocellular cancer, lung cancer, etc. This review delves into the dual impact of micropeptides on health and pathology, exploring their pivotal role in preserving normal physiological homeostasis and probing their involvement in the triggering and progression of diseases.
Collapse
Affiliation(s)
- Deepyaman Das
- Computational and Systems Biology Laboratory, Department of Microbiology, Raiganj University, Raiganj, Uttar Dinajpur, West Bengal-733134, India
| | - Soumita Podder
- Computational and Systems Biology Laboratory, Department of Microbiology, Raiganj University, Raiganj, Uttar Dinajpur, West Bengal-733134, India
| |
Collapse
|
23
|
Cheng Y, Liang Y, Tan X, Liu L. Host long noncoding RNAs in bacterial infections. Front Immunol 2024; 15:1419782. [PMID: 39295861 PMCID: PMC11408731 DOI: 10.3389/fimmu.2024.1419782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/15/2024] [Indexed: 09/21/2024] Open
Abstract
Bacterial infections remain a significant global health concern, necessitating a comprehensive understanding of the intricate host-pathogen interactions that play a critical role in the outcome of infectious diseases. Recent investigations have revealed that noncoding RNAs (ncRNAs) are key regulators of these complex interactions. Among them, long noncoding RNAs (lncRNAs) have gained significant attention because of their diverse regulatory roles in gene expression, cellular processes and the production of cytokines and chemokines in response to bacterial infections. The host utilizes lncRNAs as a defense mechanism to limit microbial pathogen invasion and replication. On the other hand, some host lncRNAs contribute to the establishment and maintenance of bacterial pathogen reservoirs within the host by promoting bacterial pathogen survival, replication, and dissemination. However, our understanding of host lncRNAs in the context of bacterial infections remains limited. This review focuses on the impact of host lncRNAs in shaping host-pathogen interactions, shedding light on their multifaceted functions in both host defense and bacterial survival, and paving the way for future research aimed at harnessing their regulatory potential for clinical applications.
Collapse
Affiliation(s)
- Yong Cheng
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, United States
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, United States
| | - Yurong Liang
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, United States
- Department of Physiological Sciences, Oklahoma State University, Stillwater, OK, United States
| | - Xuejuan Tan
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, United States
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, United States
| | - Lin Liu
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK, United States
- Department of Physiological Sciences, Oklahoma State University, Stillwater, OK, United States
| |
Collapse
|
24
|
Yang Y, Pu J, Yang Y. Glycolysis and chemoresistance in acute myeloid leukemia. Heliyon 2024; 10:e35721. [PMID: 39170140 PMCID: PMC11336864 DOI: 10.1016/j.heliyon.2024.e35721] [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: 10/17/2023] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
While traditional high-dose chemotherapy can effectively prolong the overall survival of acute myeloid leukemia (AML) patients and contribute to better prognostic outcomes, the advent of chemoresistance is a persistent challenge to effective AML management in the clinic. The therapeutic resistance is thought to emerge owing to the heterogeneous and adaptable nature of tumor cells when exposed to exogenous stimuli. Recent studies have focused on exploring metabolic changes that may afford novel opportunities to treat AML, with a particular focus on glycolytic metabolism. The Warburg effect, a hallmark of cancer, refers to metabolism of glucose through glycolysis under normoxic conditions, which contributes to the development of chemoresistance. Despite the key significance of this metabolic process in the context of malignant transformation, the underlying molecular mechanisms linking glycolysis to chemoresistance in AML remain incompletely understood. This review offers an overview of the current status of research focused on the relationship between glycolytic metabolism and AML resistance to chemotherapy, with a particular focus on the contributions of glucose transporters, key glycolytic enzymes, signaling pathways, non-coding RNAs, and the tumor microenvironment to this relationship. Together, this article will provide a foundation for the selection of novel therapeutic targets and the formulation of new approaches to treating AML.
Collapse
Affiliation(s)
- Yan Yang
- Department of Neonatology, Zigong Maternity and Child Health Care Hospital, Zigong, Sichuan, 643000, China
| | - Jianlin Pu
- Department of Psychiatry, The Zigong Affiliated Hospital of Southwest Medical University, Zigong mental health Center, Zigong Institute of Brain Science, Zigong, Sichuan, 643000, China
| | - You Yang
- Department of Pediatrics (Children Hematological Oncology), Birth Defects and Childhood Hematological Oncology Laboratory, The Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, 646000, China
- The Second Hospital, Centre for Reproductive Medicine, Advanced Medical Research Institute, Key Laboratory for Experimental Teratology of the Ministry of Education, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, China
| |
Collapse
|
25
|
Li A, Zhou H, Xiong S, Li J, Mallik S, Fei R, Liu Y, Zhou H, Wang X, Hei X, Wang L. PLEKv2: predicting lncRNAs and mRNAs based on intrinsic sequence features and the coding-net model. BMC Genomics 2024; 25:756. [PMID: 39095710 PMCID: PMC11295476 DOI: 10.1186/s12864-024-10662-y] [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: 04/02/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) are RNA transcripts of more than 200 nucleotides that do not encode canonical proteins. Their biological structure is similar to messenger RNAs (mRNAs). To distinguish between lncRNA and mRNA transcripts quickly and accurately, we upgraded the PLEK alignment-free tool to its next version, PLEKv2, and constructed models tailored for both animals and plants. RESULTS PLEKv2 can achieve 98.7% prediction accuracy for human datasets. Compared with classical tools and deep learning-based models, this is 8.1%, 3.7%, 16.6%, 1.4%, 4.9%, and 48.9% higher than CPC2, CNCI, Wen et al.'s CNN, LncADeep, PLEK, and NcResNet, respectively. The accuracy of PLEKv2 was > 90% for cross-species prediction. PLEKv2 is more effective and robust than CPC2, CNCI, LncADeep, PLEK, and NcResNet for primate datasets (including chimpanzees, macaques, and gorillas). Moreover, PLEKv2 is not only suitable for non-human primates that are closely related to humans, but can also predict the coding ability of RNA sequences in plants such as Arabidopsis. CONCLUSIONS The experimental results illustrate that the model constructed by PLEKv2 can distinguish lncRNAs and mRNAs better than PLEK. The PLEKv2 software is freely available at https://sourceforge.net/projects/plek2/ .
Collapse
Affiliation(s)
- Aimin Li
- Shaanxi Key Laboratory for Network Computing and Security Technology, School of Computer Science and Engineering, Xi'an University of Technology, Xi'an, Shaanxi, 710048, China.
| | - Haotian Zhou
- Shaanxi Key Laboratory for Network Computing and Security Technology, School of Computer Science and Engineering, Xi'an University of Technology, Xi'an, Shaanxi, 710048, China
| | - Siqi Xiong
- Department of Information Engineering, College of Technology, Hubei Engineering University, Xiaogan, Hubei, 432000, China.
| | - Junhuai Li
- Shaanxi Key Laboratory for Network Computing and Security Technology, School of Computer Science and Engineering, Xi'an University of Technology, Xi'an, Shaanxi, 710048, China
| | - Saurav Mallik
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Rong Fei
- Shaanxi Key Laboratory for Network Computing and Security Technology, School of Computer Science and Engineering, Xi'an University of Technology, Xi'an, Shaanxi, 710048, China
| | - Yajun Liu
- Shaanxi Key Laboratory for Network Computing and Security Technology, School of Computer Science and Engineering, Xi'an University of Technology, Xi'an, Shaanxi, 710048, China
| | - Hongfang Zhou
- Shaanxi Key Laboratory for Network Computing and Security Technology, School of Computer Science and Engineering, Xi'an University of Technology, Xi'an, Shaanxi, 710048, China
| | - Xiaofan Wang
- Shaanxi Key Laboratory for Network Computing and Security Technology, School of Computer Science and Engineering, Xi'an University of Technology, Xi'an, Shaanxi, 710048, China
| | - Xinhong Hei
- Shaanxi Key Laboratory for Network Computing and Security Technology, School of Computer Science and Engineering, Xi'an University of Technology, Xi'an, Shaanxi, 710048, China
| | - Lei Wang
- Shaanxi Key Laboratory for Network Computing and Security Technology, School of Computer Science and Engineering, Xi'an University of Technology, Xi'an, Shaanxi, 710048, China
| |
Collapse
|
26
|
Ritz NL, Bastiaanssen TFS, Cowan CSM, Smith L, Theune N, Brocka M, Myers EM, Moloney RD, Moloney GM, Shkoporov AN, Draper LA, Hill C, Dinan TG, Slattery DA, Cryan JF. Social fear extinction susceptibility is associated with Microbiota-Gut-Brain axis alterations. Brain Behav Immun 2024; 120:315-326. [PMID: 38852762 DOI: 10.1016/j.bbi.2024.06.009] [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: 11/21/2023] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024] Open
Abstract
Social anxiety disorder is a common psychiatric condition that severely affects quality of life of individuals and is a significant societal burden. Although many risk factors for social anxiety exist, it is currently unknown how social fear sensitivity manifests biologically. Furthermore, since some individuals are resilient and others are susceptible to social fear, it is important to interrogate the mechanisms underpinning individual response to social fear situations. The microbiota-gut-brain axis has been associated with social behaviour, has recently been linked with social anxiety disorder, and may serve as a therapeutic target for modulation. Here, we assess the potential of this axis to be linked with social fear extinction processes in a murine model of social anxiety disorder. To this end, we correlated differential social fear responses with microbiota composition, central gene expression, and immune responses. Our data provide evidence that microbiota variability is strongly correlated with alterations in social fear behaviour. Moreover, we identified altered gene candidates by amygdalar transcriptomics that are linked with social fear sensitivity. These include genes associated with social behaviour (Armcx1, Fam69b, Kcnj9, Maoa, Serinc5, Slc6a17, Spata2, and Syngr1), inflammation and immunity (Cars, Ckmt1, Klf5, Maoa, Map3k12, Pex5, Serinc5, Sidt1, Spata2), and microbe-host interaction (Klf5, Map3k12, Serinc5, Sidt1). Together, these data provide further evidence for a role of the microbiota-gut-brain axis in social fear responses.
Collapse
Affiliation(s)
- Nathaniel L Ritz
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Thomaz F S Bastiaanssen
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Caitlin S M Cowan
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Linda Smith
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; School of Microbiology, University College Cork, Cork, T12K8AF, Ireland
| | - Nigel Theune
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Marta Brocka
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Eibhlís M Myers
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Rachel D Moloney
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Gerard M Moloney
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland
| | - Andrey N Shkoporov
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; School of Microbiology, University College Cork, Cork, T12K8AF, Ireland
| | - Lorraine A Draper
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; School of Microbiology, University College Cork, Cork, T12K8AF, Ireland
| | - Colin Hill
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; School of Microbiology, University College Cork, Cork, T12K8AF, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Psychiatry and Neurobehavioural Science, University College Cork, Cork T12YT20, Ireland
| | - David A Slattery
- Dept. of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, Frankfurt 60528, Germany
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork T12YT20, Ireland; Dept. of Anatomy and Neuroscience, University College Cork, Cork T12YT20, Ireland.
| |
Collapse
|
27
|
Yang H, Xuan L, Wang S, Luo H, Duan X, Guo J, Cui S, Xin J, Hao J, Li X, Chen J, Sun F, Hu X, Li S, Zhang Y, Jiao L, Yang B, Sun L. LncRNA CCRR maintains Ca 2+ homeostasis against myocardial infarction through the FTO-SERCA2a pathway. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1601-1619. [PMID: 38761356 DOI: 10.1007/s11427-023-2527-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/11/2024] [Indexed: 05/20/2024]
Abstract
Cardiac conduction regulatory RNA (CCRR) has been documented as an antiarrhythmic lncRNA in our earlier investigation. This study aimed to evaluate the effects of CCRR on SERCA2a and the associated Ca2+ homeostasis in myocardial infarction (MI). Overexpression of CCRR via AAV9-mediated delivery not only partially reversed ischemia-induced contractile dysfunction but also alleviated abnormal Ca2+ homeostasis and reduced the heightened methylation level of SERCA2a following MI. These effects were also observed in CCRR over-expressing transgenic mice. A conserved sequence domain of CCRR mimicked the protective function observed with the full length. Furthermore, silencing CCRR in healthy mice led to intracellular Ca2+ overloading of cardiomyocytes. CCRR increased SERCA2a protein stability by upregulating FTO expression. The direct interaction between CCRR and FTO protein was characterized by RNA-binding protein immunoprecipitation (RIP) analysis and RNA pulldown experiments. Activation of NFATc3 was identified as an upstream mechanism responsible for CCRR downregulation in MI. This study demonstrates that CCRR is a protective lncRNA that acts by maintaining the function of FTO, thereby reducing the m6A RNA methylation level of SERCA2a, ultimately preserving calcium homeostasis for myocardial contractile function in MI. Therefore, CCRR may be considered a promising therapeutic strategy with a beneficial role in cardiac pathology.
Collapse
Affiliation(s)
- Hua Yang
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Lina Xuan
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Shengjie Wang
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Huishan Luo
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xiaomeng Duan
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jianjun Guo
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Shijia Cui
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jieru Xin
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Junwei Hao
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xiufang Li
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jun Chen
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Feihan Sun
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xiaolin Hu
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Siyun Li
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ying Zhang
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Lei Jiao
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Baofeng Yang
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| | - Lihua Sun
- Department of Pharmacology, Harbin Medical University (State Key Laboratory of Frigid Zone Cardiovascular Disease, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Joint International Research Laboratory of Cardiovascular Medicine Research, Ministry of Education, China), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
28
|
Kazmi SSUH, Tayyab M, Pastorino P, Barcelò D, Yaseen ZM, Grossart HP, Khan ZH, Li G. Decoding the molecular concerto: Toxicotranscriptomic evaluation of microplastic and nanoplastic impacts on aquatic organisms. JOURNAL OF HAZARDOUS MATERIALS 2024; 472:134574. [PMID: 38739959 DOI: 10.1016/j.jhazmat.2024.134574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/30/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
The pervasive and steadily increasing presence of microplastics/nanoplastics (MPs/NPs) in aquatic environments has raised significant concerns regarding their potential adverse effects on aquatic organisms and their integration into trophic dynamics. This emerging issue has garnered the attention of (eco)toxicologists, promoting the utilization of toxicotranscriptomics to unravel the responses of aquatic organisms not only to MPs/NPs but also to a wide spectrum of environmental pollutants. This review aims to systematically explore the broad repertoire of predicted molecular responses by aquatic organisms, providing valuable intuitions into complex interactions between plastic pollutants and aquatic biota. By synthesizing the latest literature, present analysis sheds light on transcriptomic signatures like gene expression, interconnected pathways and overall molecular mechanisms influenced by various plasticizers. Harmful effects of these contaminants on key genes/protein transcripts associated with crucial pathways lead to abnormal immune response, metabolic response, neural response, apoptosis and DNA damage, growth, development, reproductive abnormalities, detoxification, and oxidative stress in aquatic organisms. However, unique challenge lies in enhancing the fingerprint of MPs/NPs, presenting complicated enigma that requires decoding their specific impact at molecular levels. The exploration endeavors, not only to consolidate existing knowledge, but also to identify critical gaps in understanding, push forward the frontiers of knowledge about transcriptomic signatures of plastic contaminants. Moreover, this appraisal emphasizes the imperative to monitor and mitigate the contamination of commercially important aquatic species by MPs/NPs, highlighting the pivotal role that regulatory frameworks must play in protecting all aquatic ecosystems. This commitment aligns with the broader goal of ensuring the sustainability of aquatic resources and the resilience of ecosystems facing the growing threat of plastic pollutants.
Collapse
Affiliation(s)
- Syed Shabi Ul Hassan Kazmi
- Key Laboratory of Urban Environment and Health, Ningbo Urban Environment Observation and Research Station, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China; Zhejiang Key Laboratory of Urban Environmental Processes and Pollution Control, CAS Haixi Industrial Technology Innovation Center in Beilun, Ningbo 315830, PR China
| | - Muhammad Tayyab
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, PR China
| | - Paolo Pastorino
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, 10154 Torino, Italy
| | - Damià Barcelò
- Institute of Environmental Assessment and Water Research (IDAEA-CSIC), 08034 Barcelona, Spain
| | - Zaher Mundher Yaseen
- Civil and Environmental Engineering Department, King Fahd University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia; Interdisciplinary Research Center for Membranes and Water Security, King Fahd University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia
| | - Hans-Peter Grossart
- Plankton and Microbial Ecology, Leibniz Institute for Freshwater Ecology and Inland Fisheries, (IGB), Alte Fischerhuette 2, Neuglobsow, D-16775, Germany; Institute of Biochemistry and Biology, Potsdam University, Maulbeerallee 2, D-14469 Potsdam, Germany
| | - Zulqarnain Haider Khan
- Key Laboratory of Urban Environment and Health, Ningbo Urban Environment Observation and Research Station, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China; Zhejiang Key Laboratory of Urban Environmental Processes and Pollution Control, CAS Haixi Industrial Technology Innovation Center in Beilun, Ningbo 315830, PR China
| | - Gang Li
- Key Laboratory of Urban Environment and Health, Ningbo Urban Environment Observation and Research Station, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China; Zhejiang Key Laboratory of Urban Environmental Processes and Pollution Control, CAS Haixi Industrial Technology Innovation Center in Beilun, Ningbo 315830, PR China.
| |
Collapse
|
29
|
Hu S, Han X, Liu G, Wang S. LncRNAs as potential prognosis/diagnosis markers and factors driving drug resistance of osteosarcoma, a review. Front Endocrinol (Lausanne) 2024; 15:1415722. [PMID: 39015175 PMCID: PMC11249743 DOI: 10.3389/fendo.2024.1415722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Osteosarcoma is a common malignancy that often occurs in children, teenagers and young adults. Although the treatment strategy has improved, the results are still poor for most patients with metastatic or recurrent osteosarcomas. Therefore, it is necessary to identify new and effective prognostic biomarkers and therapeutic targets for diseases. Human genomes contain lncRNAs, transcripts with limited or insufficient capacity to encode proteins. They have been implicated in tumorigenesis, particularly regarding the onset, advancement, resistance to treatment, recurrence and remote dissemination of malignancies. Aberrant lncRNA expression in osteosarcomas has been reported by numerous researchers; lncRNAs have the potential to exhibit either oncogenic or tumor-suppressing behaviors and thus, to govern the advancement of this skeletal cancer. They are suspected to influence osteosarcoma cell growth, replication, invasion, migration, remote dissemination and programmed cell death. Additionally, they have been recognized as clinical markers, and may participate in the development of multidrug resistance. Therefore, the study of lncRNAs in the growth, metastasis, treatment and prognosis of osteosarcoma is very important for the active prevention and treatment of osteosarcoma. Consequently, this work reviews the functions of lncRNAs.
Collapse
Affiliation(s)
- Siwang Hu
- The Orthopedic Center, Wenling First People’s Hospital (The Affiliated Wenling Hospital of Wenzhou Medical University), Wenling, China
| | - Xuebing Han
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, China
| | - Gang Liu
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, China
| | - Shuangshuang Wang
- Department of Cardiology, Wenling First People’s Hospital (The Affiliated Wenling Hospital of Wenzhou Medical University), Wenling, China
| |
Collapse
|
30
|
Das S, Zea Rojas MP, Tran EJ. Novel insights on the positive correlation between sense and antisense pairs on gene expression. WILEY INTERDISCIPLINARY REVIEWS. RNA 2024; 15:e1864. [PMID: 39087253 PMCID: PMC11626863 DOI: 10.1002/wrna.1864] [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: 11/30/2023] [Revised: 05/14/2024] [Accepted: 05/19/2024] [Indexed: 08/02/2024]
Abstract
A considerable proportion of the eukaryotic genome undergoes transcription, leading to the generation of noncoding RNA molecules that lack protein-coding information and are not subjected to translation. These noncoding RNAs (ncRNAs) are well recognized to have essential roles in several biological processes. Long noncoding RNAs (lncRNAs) represent the most extensive category of ncRNAs found in the human genome. Much research has focused on investigating the roles of cis-acting lncRNAs in the regulation of specific target gene expression. In the majority of instances, the regulation of sense gene expression by its corresponding antisense pair occurs in a negative (discordant) manner, resulting in the suppression of the target genes. The notion that a negative correlation exists between sense and antisense pairings is, however, not universally valid. In fact, several recent studies have reported a positive relationship between corresponding cis antisense pairs within plants, budding yeast, and mammalian cancer cells. The positive (concordant) correlation between anti-sense and sense transcripts leads to an increase in the level of the sense transcript within the same genomic loci. In addition, mechanisms such as altering chromatin structure, the formation of R loops, and the recruitment of transcription factors can either enhance transcription or stabilize sense transcripts through their antisense pairs. The primary objective of this work is to provide a comprehensive understanding of both aspects of antisense regulation, specifically focusing on the positive correlation between sense and antisense transcripts in the context of eukaryotic gene expression, including its implications towards cancer progression. This article is categorized under: RNA Processing > 3' End Processing Regulatory RNAs/RNAi/Riboswitches > Regulatory RNAs.
Collapse
Affiliation(s)
- Subhadeep Das
- Department of BiochemistryPurdue UniversityWest LafayetteIndianaUSA
- Purdue University Institute for Cancer Research, Purdue UniversityWest LafayetteIndianaUSA
| | | | - Elizabeth J. Tran
- Department of BiochemistryPurdue UniversityWest LafayetteIndianaUSA
- Purdue University Institute for Cancer Research, Purdue UniversityWest LafayetteIndianaUSA
| |
Collapse
|
31
|
Naseer QA, Malik A, Zhang F, Chen S. Exploring the enigma: history, present, and future of long non-coding RNAs in cancer. Discov Oncol 2024; 15:214. [PMID: 38847897 PMCID: PMC11161455 DOI: 10.1007/s12672-024-01077-y] [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: 02/23/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Long noncoding RNAs (lncRNAs), which are more than 200 nucleotides in length and do not encode proteins, play crucial roles in governing gene expression at both the transcriptional and posttranscriptional levels. These molecules demonstrate specific expression patterns in various tissues and developmental stages, suggesting their involvement in numerous developmental processes and diseases, notably cancer. Despite their widespread acknowledgment and the growing enthusiasm surrounding their potential as diagnostic and prognostic biomarkers, the precise mechanisms through which lncRNAs function remain inadequately understood. A few lncRNAs have been studied in depth, providing valuable insights into their biological activities and suggesting emerging functional themes and mechanistic models. However, the extent to which the mammalian genome is transcribed into functional noncoding transcripts is still a matter of debate. This review synthesizes our current understanding of lncRNA biogenesis, their genomic contexts, and their multifaceted roles in tumorigenesis, highlighting their potential in cancer-targeted therapy. By exploring historical perspectives alongside recent breakthroughs, we aim to illuminate the diverse roles of lncRNA and reflect on the broader implications of their study for understanding genome evolution and function, as well as for advancing clinical applications.
Collapse
Affiliation(s)
- Qais Ahmad Naseer
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, China
| | - Abdul Malik
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, China
| | - Fengyuan Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, China
| | - Shengxia Chen
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, China.
| |
Collapse
|
32
|
Wei P, Tian K, Liu H, Li K, Alam N, Cheng D, Li M, He X, Guo J, Wang R, Wang W, Bai L, Liu E, Xu B, Li Y, Zhao S. Urotensin II receptor deficiency ameliorates ligation-induced carotid intimal hyperplasia partially through the RhoA-YAP1 pathway. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167170. [PMID: 38631407 DOI: 10.1016/j.bbadis.2024.167170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
Intimal hyperplasia (IH) is a common pathological feature of vascular proliferative diseases, such as atherosclerosis and restenosis after angioplasty. Urotensin II (UII) and its receptor (UTR) are widely expressed in cardiovascular tissues. However, it remains unclear whether the UII/UTR system is involved in IH. Right unilateral common carotid artery ligation was performed and maintained for 21 days to induce IH in UTR knockout (UTR-/-) and wild-type (WT) mice. Histological analysis revealed that compared with WT mice, UTR-deficient mice exhibited a decreased neointimal area, angiostenosis and intima-media ratio. Immunostaining revealed fewer smooth muscle cells (SMCs), endothelial cells and macrophages in the lesions of UTR-/- mice than in those of WT mice. Protein interaction analysis suggested that the UTR may affect cell proliferation by regulating YAP and its downstream target genes. In vitro experiments revealed that UII can promote the proliferation and migration of SMCs, and western blotting also revealed that UII increased the protein expression of RhoA, CTGF, Cyclin D1 and PCNA and downregulated p-YAP protein expression, while these effects could be partly reversed by urantide. To evaluate the translational value of UTRs in IH management, WT mice were also treated with two doses of urantide, a UTR antagonist, to confirm the benefit of UTR blockade in IH progression. A high dose of urantide (600 μg/kg/day), rather than a low dose (60 μg/kg/day), successfully improved ligation-induced IH compared with that in mice receiving vehicle. The results of the present study suggested that the UII/UTR system may regulate IH partly through the RhoA-YAP signaling pathway.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Movement
- Cell Proliferation
- Hyperplasia/metabolism
- Hyperplasia/pathology
- Ligation
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima/metabolism
- Neointima/pathology
- Neointima/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- rhoA GTP-Binding Protein/metabolism
- rhoA GTP-Binding Protein/genetics
- Signal Transduction
- Tunica Intima/pathology
- Tunica Intima/metabolism
- Urotensins/metabolism
- Urotensins/genetics
- Urotensins/pharmacology
- YAP-Signaling Proteins/metabolism
Collapse
Affiliation(s)
- Panpan Wei
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Kangli Tian
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Haole Liu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Kexin Li
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Naqash Alam
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Daxin Cheng
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Meng Li
- Department of Vascular Surgery, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xue He
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jia Guo
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rong Wang
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Weirong Wang
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Liang Bai
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Enqi Liu
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China; Laboratory Animal Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yankui Li
- Department of Vascular Surgery, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Sihai Zhao
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China; Laboratory Animal Center, Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
33
|
Duffy EE, Assad EG, Kalish BT, Greenberg ME. Small but mighty: the rise of microprotein biology in neuroscience. Front Mol Neurosci 2024; 17:1386219. [PMID: 38807924 PMCID: PMC11130481 DOI: 10.3389/fnmol.2024.1386219] [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: 02/14/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
The mammalian central nervous system coordinates a network of signaling pathways and cellular interactions, which enable a myriad of complex cognitive and physiological functions. While traditional efforts to understand the molecular basis of brain function have focused on well-characterized proteins, recent advances in high-throughput translatome profiling have revealed a staggering number of proteins translated from non-canonical open reading frames (ncORFs) such as 5' and 3' untranslated regions of annotated proteins, out-of-frame internal ORFs, and previously annotated non-coding RNAs. Of note, microproteins < 100 amino acids (AA) that are translated from such ncORFs have often been neglected due to computational and biochemical challenges. Thousands of putative microproteins have been identified in cell lines and tissues including the brain, with some serving critical biological functions. In this perspective, we highlight the recent discovery of microproteins in the brain and describe several hypotheses that have emerged concerning microprotein function in the developing and mature nervous system.
Collapse
Affiliation(s)
- Erin E. Duffy
- Department of Neurobiology, Harvard Medical School, Boston, MA, United States
| | - Elena G. Assad
- Department of Neurobiology, Harvard Medical School, Boston, MA, United States
| | - Brian T. Kalish
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Division of Neonatology, Department of Paediatrics, Hospital for Sick Children, Toronto, ON, Canada
| | | |
Collapse
|
34
|
Singh AK. Rules and impacts of nonsense-mediated mRNA decay in the degradation of long noncoding RNAs. WILEY INTERDISCIPLINARY REVIEWS. RNA 2024; 15:e1853. [PMID: 38741356 DOI: 10.1002/wrna.1853] [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: 12/30/2023] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 05/16/2024]
Abstract
Nonsense-mediated mRNA decay (NMD) is a quality-control process that selectively degrades mRNAs having premature termination codon, upstream open reading frame, or unusually long 3'UTR. NMD detects such mRNAs and rapidly degrades them during initial rounds of translation in the eukaryotic cells. Since NMD is a translation-dependent cytoplasmic mRNA surveillance process, the noncoding RNAs were initially believed to be NMD-resistant. The sequence feature-based analysis has revealed that many putative long noncoding RNAs (lncRNAs) have short open reading frames, most of which have translation potential. Subsequent transcriptome-based molecular studies showed an association of a large set of such putative lncRNAs with translating ribosomes, and some of them produce stable and functionally active micropeptides. The translationally active lncRNAs typically have relatively longer and unprotected 3'UTR, which can induce their NMD-dependent degradation. This review defines the mechanism and regulation of NMD-dependent degradation of lncRNAs and its impact on biological processes related to the functions of lncRNAs or their encoded micropeptides. This article is categorized under: RNA Turnover and Surveillance > Turnover/Surveillance Mechanisms RNA Turnover and Surveillance > Regulation of RNA Stability RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Anand Kumar Singh
- Department of Biology, Indian Institute of Science Education and Research Tirupati, Tirupati, Andhra Pradesh, India
| |
Collapse
|
35
|
Biferali B, Mocciaro E, Runfola V, Gabellini D. Long non-coding RNAs and their role in muscle regeneration. Curr Top Dev Biol 2024; 158:433-465. [PMID: 38670715 DOI: 10.1016/bs.ctdb.2024.02.010] [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: 04/28/2024]
Abstract
In mammals, most of the genome is transcribed to generate a large and heterogeneous variety of non-protein coding RNAs, that are broadly grouped according to their size. Long noncoding RNAs include a very large and versatile group of molecules. Despite only a minority of them has been functionally characterized, there is emerging evidence indicating long noncoding RNAs as important regulators of expression at multiple levels. Several of them have been shown to be modulated during myogenic differentiation, playing important roles in the regulation of skeletal muscle development, differentiation and homeostasis, and contributing to neuromuscular diseases. In this chapter, we have summarized the current knowledge about long noncoding RNAs in skeletal muscle and discussed specific examples of long noncoding RNAs (lncRNAs and circRNAs) regulating muscle stem cell biology. We have also discussed selected long noncoding RNAs involved in the most common neuromuscular diseases.
Collapse
Affiliation(s)
- Beatrice Biferali
- Gene Expression Regulation Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Emanuele Mocciaro
- Gene Expression Regulation Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valeria Runfola
- Gene Expression Regulation Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Davide Gabellini
- Gene Expression Regulation Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
36
|
Shin YJ, Kwon KS, Suh Y, Lee KP. The role of non-coding RNAs in muscle aging: regulatory mechanisms and therapeutic potential. Front Mol Biosci 2024; 10:1308274. [PMID: 38264571 PMCID: PMC10803457 DOI: 10.3389/fmolb.2023.1308274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/26/2023] [Indexed: 01/25/2024] Open
Abstract
Muscle aging is a complex physiological process that leads to the progressive decline in muscle mass and function, contributing to debilitating conditions in the elderly such as sarcopenia. In recent years, non-coding RNAs (ncRNAs) have been increasingly recognized as major regulators of muscle aging and related cellular processes. Here, we comprehensively review the emerging role of ncRNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), in the regulation of muscle aging. We also discuss how targeting these ncRNAs can be explored for the development of novel interventions to combat age-related muscle decline. The insights provided in this review offer a promising avenue for future research and therapeutic strategies aimed at improving muscle health during aging.
Collapse
Affiliation(s)
- Yeo Jin Shin
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Ki-Sun Kwon
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Bioscience, KRIBB School, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
- Aventi Inc., Daejeon, Republic of Korea
| | - Yousin Suh
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, United States
- Department of Genetics and Development, Columbia University, New York, NY, United States
| | - Kwang-Pyo Lee
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Bioscience, KRIBB School, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
37
|
Sabikunnahar B, Caldwell S, Varnum S, Hogan T, Lahue KG, Rathkolb B, Gerlini R, Dragano NRV, Aguilar‐Pimentel A, Irmler M, Sanz‐Moreno A, da Silva‐Buttkus P, Beckers J, Wolf E, Gailus‐Durner V, Fuchs H, Hrabe de Angelis M, Ather JL, Poynter ME, Krementsov DN. LncRNA U90926 is dispensable for the development of obesity-associated phenotypes in vivo. Physiol Rep 2024; 12:e15901. [PMID: 38171546 PMCID: PMC10764201 DOI: 10.14814/phy2.15901] [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: 08/26/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024] Open
Abstract
Obesity is a global health problem characterized by excessive fat accumulation, driven by adipogenesis and lipid accumulation. Long non-coding RNAs (lncRNAs) have recently been implicated in regulating adipogenesis and adipose tissue function. Mouse lncRNA U90926 was previously identified as a repressor of in vitro adipogenesis in 3T3-L1 preadipocytes. Consequently, we hypothesized that, in vivo, U90926 may repress adipogenesis, and hence its deletion would increase weight gain and adiposity. We tested the hypothesis by applying U90926-deficient (U9-KO) mice to a high-throughput phenotyping pipeline. Compared with WT, U9-KO mice showed no major differences across a wide range of behavioral, neurological, and other physiological parameters. In mice fed a standard diet, we have found no differences in obesity-related phenotypes, including weight gain, fat mass, and plasma concentrations of glucose, insulin, triglycerides, and free fatty acids, in U9-KO mice compared to WT. U90926 deficiency lacked a major effect on white adipose tissue morphology and gene expression profile. Furthermore, in mice fed a high-fat diet, we found increased expression of U90926 in adipose tissue stromal vascular cell fraction, yet observed no effect of U90926 deficiency on weight gain, fat mass, adipogenesis marker expression, and immune cell infiltration into the adipose tissue. These data suggest that the U90926 lacks an essential role in obesity-related phenotypes and adipose tissue biology in vivo.
Collapse
Affiliation(s)
- Bristy Sabikunnahar
- Department of Biomedical and Health SciencesUniversity of VermontBurlingtonVermontUSA
| | - Sydney Caldwell
- Department of Biomedical and Health SciencesUniversity of VermontBurlingtonVermontUSA
| | - Stella Varnum
- Department of Biomedical and Health SciencesUniversity of VermontBurlingtonVermontUSA
| | - Tyler Hogan
- Department of Biomedical and Health SciencesUniversity of VermontBurlingtonVermontUSA
| | - Karolyn G. Lahue
- Department of Biomedical and Health SciencesUniversity of VermontBurlingtonVermontUSA
| | - Birgit Rathkolb
- Institute of Experimental Genetics and German Mouse ClinicHelmholtz Zentrum MünchenNeuherbergGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
- Institute of Molecular Animal Breeding and Biotechnology, Gene CenterLudwig‐Maximilians‐University MünchenMunichGermany
| | - Raffaele Gerlini
- Institute of Experimental Genetics and German Mouse ClinicHelmholtz Zentrum MünchenNeuherbergGermany
| | - Nathalia R. V. Dragano
- Institute of Experimental Genetics and German Mouse ClinicHelmholtz Zentrum MünchenNeuherbergGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| | - Antonio Aguilar‐Pimentel
- Institute of Experimental Genetics and German Mouse ClinicHelmholtz Zentrum MünchenNeuherbergGermany
| | - Martin Irmler
- Institute of Experimental Genetics and German Mouse ClinicHelmholtz Zentrum MünchenNeuherbergGermany
| | - Adrián Sanz‐Moreno
- Institute of Experimental Genetics and German Mouse ClinicHelmholtz Zentrum MünchenNeuherbergGermany
| | - Patricia da Silva‐Buttkus
- Institute of Experimental Genetics and German Mouse ClinicHelmholtz Zentrum MünchenNeuherbergGermany
| | - Johannes Beckers
- Institute of Experimental Genetics and German Mouse ClinicHelmholtz Zentrum MünchenNeuherbergGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
- TUM School of Life SciencesTechnische Universität MünchenFreisingGermany
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene CenterLudwig‐Maximilians‐University MünchenMunichGermany
| | - Valerie Gailus‐Durner
- Institute of Experimental Genetics and German Mouse ClinicHelmholtz Zentrum MünchenNeuherbergGermany
| | - Helmut Fuchs
- Institute of Experimental Genetics and German Mouse ClinicHelmholtz Zentrum MünchenNeuherbergGermany
| | - Martin Hrabe de Angelis
- Institute of Experimental Genetics and German Mouse ClinicHelmholtz Zentrum MünchenNeuherbergGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
- TUM School of Life SciencesTechnische Universität MünchenFreisingGermany
| | | | | | - Dimitry N. Krementsov
- Department of Biomedical and Health SciencesUniversity of VermontBurlingtonVermontUSA
| |
Collapse
|
38
|
Liu J, Hu X, Xin W, Wang X. Exosomal Non-coding RNAs: A New Approach to Melanoma Diagnosis and Therapeutic Strategy. Curr Med Chem 2024; 31:6084-6109. [PMID: 37877505 DOI: 10.2174/0109298673267553231017053329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/03/2023] [Accepted: 09/22/2023] [Indexed: 10/26/2023]
Abstract
Malignant melanoma (MM) is a highly aggressive cancer with a poor prognosis. Currently, although a variety of therapies are available for treating melanoma, MM is still a serious threat to the patient's life due to numerous factors, such as the recurrence of tumors, the emergence of drug resistance, and the lack of effective therapeutic agents. Exosomes are biologically active lipid-bilayer extracellular vesicles secreted by diverse cell types that mediate intercellular signal communication. Studies found that exosomes are involved in cancer by carrying multiple bioactive molecules, including non-- coding RNAs (ncRNAs). The ncRNAs have been reported to play an important role in regulating proliferation, angiogenesis, immune regulation, invasion, metastasis, and treatment resistance of tumors. However, the functional role of exosomal ncRNAs in MM remains unknown. Therefore, this review summarizes the current state of melanoma diagnosis, treatment, and the application of exosomal ncRNAs in MM patients, which may provide new insights into the mechanisms involved in melanoma progression and serve as biomarkers for diagnosis and therapeutic targets.
Collapse
Affiliation(s)
- Jie Liu
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| | - Xiaoping Hu
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| | - Wenqiang Xin
- Department of Neurology, University Medical Center Göttingen, Göttingen 37075, Germany
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China, 300052
| | - Xianbin Wang
- Department of Emergency Medicine, The Second Affiliated Hospital of Baotou Medical College, Baotou 014030, China
| |
Collapse
|
39
|
Fontana F, Esser AK, Egbulefu C, Karmakar P, Su X, Allen JS, Xu Y, Davis JL, Gabay A, Xiang J, Kwakwa KA, Manion B, Bakewell S, Li S, Park H, Lanza GM, Achilefu S, Weilbaecher KN. Transferrin receptor in primary and metastatic breast cancer: Evaluation of expression and experimental modulation to improve molecular targeting. PLoS One 2023; 18:e0293700. [PMID: 38117806 PMCID: PMC10732420 DOI: 10.1371/journal.pone.0293700] [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: 08/09/2023] [Accepted: 10/17/2023] [Indexed: 12/22/2023] Open
Abstract
BACKGROUND Conjugation of transferrin (Tf) to imaging or nanotherapeutic agents is a promising strategy to target breast cancer. Since the efficacy of these biomaterials often depends on the overexpression of the targeted receptor, we set out to survey expression of transferrin receptor (TfR) in primary and metastatic breast cancer samples, including metastases and relapse, and investigate its modulation in experimental models. METHODS Gene expression was investigated by datamining in twelve publicly-available datasets. Dedicated Tissue microarrays (TMAs) were generated to evaluate matched primary and bone metastases as well as and pre and post chemotherapy tumors from the same patient. TMA were stained with the FDA-approved MRQ-48 antibody against TfR and graded by staining intensity (H-score). Patient-derived xenografts (PDX) and isogenic metastatic mouse models were used to study in vivo TfR expression and uptake of transferrin. RESULTS TFRC gene and protein expression were high in breast cancer of all subtypes and stages, and in 60-85% of bone metastases. TfR was detectable after neoadjuvant chemotherapy, albeit with some variability. Fluorophore-conjugated transferrin iron chelator deferoxamine (DFO) enhanced TfR uptake in human breast cancer cells in vitro and proved transferrin localization at metastatic sites and correlation of tumor burden relative to untreated tumor mice. CONCLUSIONS TfR is expressed in breast cancer, primary, metastatic, and after neoadjuvant chemotherapy. Variability in expression of TfR suggests that evaluation of the expression of TfR in individual patients could identify the best candidates for targeting. Further, systemic iron chelation with DFO may upregulate receptor expression and improve uptake of therapeutics or tracers that use transferrin as a homing ligand.
Collapse
Affiliation(s)
- Francesca Fontana
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Alison K. Esser
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Christopher Egbulefu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Partha Karmakar
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Xinming Su
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - John S. Allen
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Yalin Xu
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Jennifer L. Davis
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Ariel Gabay
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Jingyu Xiang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Kristin A. Kwakwa
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Brad Manion
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Suzanne Bakewell
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Shunqiang Li
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Haeseong Park
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Gregory M. Lanza
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Samuel Achilefu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Katherine N. Weilbaecher
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States of America
| |
Collapse
|
40
|
Ma M, Chen M, Wu X, Sooranna SR, Liu Q, Shi D, Wang J, Li H. A newly identified lncRNA lnc000100 regulates proliferation and differentiation of cattle skeletal muscle cells. Epigenetics 2023; 18:2270864. [PMID: 37910666 PMCID: PMC10768731 DOI: 10.1080/15592294.2023.2270864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 09/01/2023] [Indexed: 11/03/2023] Open
Abstract
Cattle skeletal muscle development is a complex and highly coordinated biological process mediated by a series of myogenic regulators, which plays a critical role in beef yield and quality. Long non-coding RNAs (lncRNAs) have been shown to regulate skeletal muscle development. However, the molecular mechanism by which lncRNAs regulate skeletal muscle development is largely unknown. We performed transcriptome analysis of muscle tissues of adult and embryo Angus cattle to investigate the mechanism by which lncRNA regulates skeletal muscle development between adult and embryo cattle. A total of 37,115 candidate lncRNAs were detected, and a total of 1,998 lncRNAs were differentially expressed between the muscle tissue libraries of adult and embryo cattle, including 1,229 up-regulated lncRNAs and 769 down-regulated lncRNAs (adult cattle were the control group). We verified the expression of 7 differentially expressed lncRNAs by quantitative real-time PCR (RT-qPCR), and analysed the tissue expression profile of lnc000100, which is down-regulated in the longest dorsal muscle during foetal life and which is highly specifically expressed in muscle tissue. We found that the interference of lnc000100 significantly inhibited cell proliferation and promoted cell differentiation. Lnc000100 was located in the nucleus by RNA-FISH. Our research provides certain resources for the analysis of lncRNA regulating cattle skeletal muscle development, and may also provide new insights for improving beef production and breed selection.
Collapse
Affiliation(s)
- Mengke Ma
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
| | - Mengjie Chen
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
| | - Xiaoyun Wu
- Key Laboratory of Yak Breeding Engineering of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Suren R. Sooranna
- Institute of Reproductive and Developmental Biology, Imperial College London, London, UK
| | - Qingyou Liu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Deshun Shi
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
| | - Jian Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
| | - Hui Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
| |
Collapse
|
41
|
Özdemir S, Aydın Ş, Laçin BB, Arslan H. Identification and characterization of long non-coding RNA (lncRNA) in cypermethrin and chlorpyrifos exposed zebrafish (Danio rerio) brain. CHEMOSPHERE 2023; 344:140324. [PMID: 37778644 DOI: 10.1016/j.chemosphere.2023.140324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/03/2023]
Abstract
Pesticides, such as cypermethrin (CYP) and chlorpyrifos (CPF), are widely used around the world and are known to cause toxicological effects in the brains of fish and other non-target organisms. Long non-coding RNAs (LncRNAs) are a new class of non-coding RNAs that are highly expressed in the brain and play crucial roles in brain function by regulating gene expression. Many studies have investigated the toxic effects of CYP and CPF on the brain. However, no study has been conducted on the relationship between LncRNAs and the toxicity caused by these chemicals. Therefore, this study aimed to determine changes in the lncRNA expression profile in the brains of fish exposed to CYP and CPF. Out of a total of 482 lncRNAs that were differentially expressed between control and CPF groups, 53 were found to be up-regulated, and 429 were down-regulated. Similarly, among the 200 lncRNAs differentially expressed between the control and CYP groups, 71 were up-regulated, and 129 were down-regulated. Additionally, 268 differentially expressed lncRNAs were identified between CYP and CPF groups, with 240 being up-regulated and the rest being down-regulated. In addition, LncRNAs expressed from fish brains exposed to CYP and CPF were found to regulate multiple signaling pathways, including MAPK, FoxO, PPAR, TGF-β, and Wnt signaling pathways.
Collapse
Affiliation(s)
- Selçuk Özdemir
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey; German Center for Neurodegenerative Diseases, DZNE, Bonn, Germany.
| | - Şeyma Aydın
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Burak Batuhan Laçin
- Department of Physiology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Harun Arslan
- Atatürk University, Faculty of Fisheries, Department of Basic Science, Erzurum, Turkey
| |
Collapse
|
42
|
Kumari N, Das K, Sharma S, Dahal S, Desai SS, Roy U, Sharma A, Manjunath M, Gopalakrishnan V, Retheesh ST, Javadekar SM, Choudhary B, Raghavan SC. Evaluation of potential role of R-loop and G-quadruplex DNA in the fragility of c-MYC during chromosomal translocation associated with Burkitt's lymphoma. J Biol Chem 2023; 299:105431. [PMID: 37926284 PMCID: PMC10704377 DOI: 10.1016/j.jbc.2023.105431] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/07/2023] Open
Abstract
t(8;14) translocation is the hallmark of Burkitt's lymphoma and results in c-MYC deregulation. During the translocation, c-MYC gene on chromosome 8 gets juxtaposed to the Ig switch regions on chromosome 14. Although the promoter of c-MYC has been investigated for its mechanism of fragility, little is known about other c-MYC breakpoint regions. We have analyzed the translocation break points at the exon 1/intron 1 of c-MYC locus from patients with Burkitt's lymphoma. Results showed that the breakpoint region, when present on a plasmid, could fold into an R-loop confirmation in a transcription-dependent manner. Sodium bisulfite modification assay revealed significant single-strandedness on chromosomal DNA of Burkitt's lymphoma cell line, Raji, and normal lymphocytes, revealing distinct R-loops covering up to 100 bp region. Besides, ChIP-DRIP analysis reveals that the R-loop antibody can bind to the breakpoint region. Further, we show the formation of stable parallel intramolecular G-quadruplex on non-template strand of the genome. Finally, incubation of purified AID in vitro or overexpression of AID within the cells led to enhanced mutation frequency at the c-MYC breakpoint region. Interestingly, anti-γH2AX can bind to DSBs generated at the c-MYC breakpoint region within the cells. The formation of R-loop and G-quadruplex was found to be mutually exclusive. Therefore, our results suggest that AID can bind to the single-stranded region of the R-loop and G4 DNA, leading to the deamination of cytosines to uracil and induction of DNA breaks in one of the DNA strands, leading to double-strand break, which could culminate in t(8;14) chromosomal translocation.
Collapse
Affiliation(s)
- Nitu Kumari
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Kohal Das
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Shivangi Sharma
- Department of Biochemistry, Indian Institute of Science, Bangalore, India; Institute of Bioinformatics and Applied Biotechnology, Bangalore, India
| | - Sumedha Dahal
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | | | - Urbi Roy
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Anju Sharma
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Meghana Manjunath
- Institute of Bioinformatics and Applied Biotechnology, Bangalore, India
| | - Vidya Gopalakrishnan
- Department of Biochemistry, Indian Institute of Science, Bangalore, India; Department of Zoology, St Joseph's College, Irinjalakuda, Kerala, India
| | - S T Retheesh
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Saniya M Javadekar
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Bibha Choudhary
- Institute of Bioinformatics and Applied Biotechnology, Bangalore, India
| | - Sathees C Raghavan
- Department of Biochemistry, Indian Institute of Science, Bangalore, India.
| |
Collapse
|
43
|
Mohsen JJ, Martel AA, Slavoff SA. Microproteins-Discovery, structure, and function. Proteomics 2023; 23:e2100211. [PMID: 37603371 PMCID: PMC10841188 DOI: 10.1002/pmic.202100211] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/03/2023] [Accepted: 08/10/2023] [Indexed: 08/22/2023]
Abstract
Advances in proteogenomic technologies have revealed hundreds to thousands of translated small open reading frames (sORFs) that encode microproteins in genomes across evolutionary space. While many microproteins have now been shown to play critical roles in biology and human disease, a majority of recently identified microproteins have little or no experimental evidence regarding their functionality. Computational tools have some limitations for analysis of short, poorly conserved microprotein sequences, so additional approaches are needed to determine the role of each member of this recently discovered polypeptide class. A currently underexplored avenue in the study of microproteins is structure prediction and determination, which delivers a depth of functional information. In this review, we provide a brief overview of microprotein discovery methods, then examine examples of microprotein structures (and, conversely, intrinsic disorder) that have been experimentally determined using crystallography, cryo-electron microscopy, and NMR, which provide insight into their molecular functions and mechanisms. Additionally, we discuss examples of predicted microprotein structures that have provided insight or context regarding their function. Analysis of microprotein structure at the angstrom level, and confirmation of predicted structures, therefore, has potential to identify translated microproteins that are of biological importance and to provide molecular mechanism for their in vivo roles.
Collapse
Affiliation(s)
- Jessica J. Mohsen
- Department of Chemistry, Yale University, New Haven, CT, USA
- Institute of Biomolecular Design and Discovery, Yale University, West Haven, CT, USA
| | - Alina A. Martel
- Institute of Biomolecular Design and Discovery, Yale University, West Haven, CT, USA
| | - Sarah A. Slavoff
- Department of Chemistry, Yale University, New Haven, CT, USA
- Institute of Biomolecular Design and Discovery, Yale University, West Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| |
Collapse
|
44
|
Li P, Ma X, Gu X. LncRNA MAFG-AS1 is involved in human cancer progression. Eur J Med Res 2023; 28:497. [PMID: 37941063 PMCID: PMC10631199 DOI: 10.1186/s40001-023-01486-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) refer to a type of non-protein-coding transcript of more than 200 nucleotides. LncRNAs play fundamental roles in disease development and progression, and lncRNAs are dysregulated in many pathophysiological processes. Thus, lncRNAs may have potential value in clinical applications. The lncRNA, MAF BZIP Transcription Factor G (MAFG)-AS1, is dysregulated in several cancer, including breast cancer, lung cancer, liver cancer, bladder cancer, colorectal cancer, gastric cancer, esophagus cancer, prostate cancer, pancreatic cancer, ovarian cancer, and glioma. Altered MAFG-AS1 levels are also associated with diverse clinical characteristics and patient outcomes. Mechanistically, MAFG-AS1 mediates a variety of cellular processes via the regulation of target gene expression. Therefore, the diagnostic, prognostic, and therapeutic aspects of MAFG-AS1 have been widely explored. In this review, we discuss the expression, major roles, and molecular mechanisms of MAFG-AS1, the relationship between MAFG-AS1 and clinical features of diseases, and the clinical applications of MAFG-AS1.
Collapse
Affiliation(s)
- Penghui Li
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Xiao Ma
- Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China.
| |
Collapse
|
45
|
Kratz A, Ranganathan S. Christian Schönbach 1965-2023. BIOINFORMATICS ADVANCES 2023; 3:vbad147. [PMID: 37886713 PMCID: PMC10599964 DOI: 10.1093/bioadv/vbad147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023]
Affiliation(s)
- Anton Kratz
- The Systems Biology Institute, Tokyo 141-0022, Japan
| | - Shoba Ranganathan
- Applied Biosciences, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
46
|
Sun L, Ding M, Chen F, Zhu D, Xie X. Long non‑coding RNA L13Rik promotes high glucose-induced mesangial cell hypertrophy and matrix protein expression by regulating miR-2861/CDKN1B axis. PeerJ 2023; 11:e16170. [PMID: 37868060 PMCID: PMC10586299 DOI: 10.7717/peerj.16170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 09/03/2023] [Indexed: 10/24/2023] Open
Abstract
Background Diabetic nephropathy (DN) is a frequent microvascular complication of diabetes. Glomerular mesangial cell (MC) hypertrophy occurs at the initial phase of DN and plays a critical role in the pathogenesis of DN. Given the role of long non coding RNA (lncRNA) in regulating MC hypertrophy and extracellular matrix (ECM) accumulation, our aim was to identify functional lncRNAs during MC hypertrophy. Methods Here, an lncRNA, C920021L13Rik (L13Rik for short), was identified to be up-regulated in DN progression. The expression of L13Rik in DN patients and diabetic mice was assessed using quantitative real-time PCR (qRT-PCR), and the function of L13Rik in regulating HG-induced MC hypertrophy and ECM accumulation was assessed through flow cytometry and western blotting analysis. Results The L13Rik levels were significantly increased while the miR-2861 levels were decreased in the peripheral blood of DN patients, the renal tissues of diabetic mice, and HG-treated MCs. Functionally, both L13Rik depletion and miR-2861 overexpression effectively reduced HG-induced cell hypertrophy and ECM accumulation. Mechanistically, L13Rik functioned as a competing endogenous RNA (ceRNA) to sponge miR-2861, resulting in the de-repression of cyclin-dependent kinase inhibitor 1B (CDKN1B), a gene known to regulate cell cycle and MC hypertrophy. Conclusions Collectively, the current results demonstrate that up-regulated L13Rik is correlated with DN and may be a hopeful therapeutic target for DN.
Collapse
Affiliation(s)
- Linlin Sun
- Nephrology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Miao Ding
- Nephrology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fuhua Chen
- Nephrology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dingyu Zhu
- Nephrology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinmiao Xie
- Nephrology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
47
|
Tang X, Li Q, Feng X, Yang B, Zhong X, Zhou Y, Wang Q, Mao Y, Xie W, Liu T, Tang Q, Guo W, Wu F, Feng X, Wang Q, Lu Y, Xu J. Identification and Functional Analysis of Drought-Responsive Long Noncoding RNAs in Maize Roots. Int J Mol Sci 2023; 24:15039. [PMID: 37894720 PMCID: PMC10606207 DOI: 10.3390/ijms242015039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/28/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are transcripts with lengths of more than 200 nt and limited protein-coding potential. They were found to play important roles in plant stress responses. In this study, the maize drought-tolerant inbred line AC7643 and drought-sensitive inbred line AC7729/TZSRW, as well as their recombinant inbred lines (RILs) were selected to identify drought-responsive lncRNAs in roots. Compared with non-responsive lncRNAs, drought-responsive lncRNAs had different sequence characteristics in length of genes and number of exons. The ratio of down-regulated lncRNAs induced by drought was significantly higher than that of coding genes; and lncRNAs were more widespread expressed in recombination sites in the RILs. Additionally, by integration of the modifications of DNA 5-methylcytidine (5mC), histones, and RNA N6-methyladenosine (m6A), it was found that the enrichment of histone modifications associated with transcriptional activation in the genes generated lncRNAs was lower that coding genes. The lncRNAs-mRNAs co-expression network, containing 15,340 coding genes and 953 lncRNAs, was constructed to investigate the molecular functions of lncRNAs. There are 13 modules found to be associated with survival rate under drought. We found nine SNPs located in lncRNAs among the modules associated with plant survival under drought. In conclusion, we revealed the characteristics of lncRNAs responding to drought in maize roots based on multiomics studies. These findings enrich our understanding of lncRNAs under drought and shed light on the complex regulatory networks that are orchestrated by the noncoding RNAs in response to drought stress.
Collapse
Affiliation(s)
- Xin Tang
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| | - Qimeng Li
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaoju Feng
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| | - Bo Yang
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiu Zhong
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| | - Yang Zhou
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| | - Qi Wang
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Mao
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| | - Wubin Xie
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| | - Tianhong Liu
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| | - Qi Tang
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| | - Wei Guo
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| | - Fengkai Wu
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| | - Xuanjun Feng
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| | - Qingjun Wang
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| | - Yanli Lu
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| | - Jie Xu
- Maize Research Institute, Sichuan Agricultural University, Chengdu 611130, China; (X.T.); (Q.L.); (X.F.); (B.Y.); (X.Z.); (Y.Z.); (Q.W.); (Y.M.); (W.X.); (T.L.); (Q.T.); (W.G.); (F.W.); (X.F.); (Q.W.)
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
48
|
Amaral P, Carbonell-Sala S, De La Vega FM, Faial T, Frankish A, Gingeras T, Guigo R, Harrow JL, Hatzigeorgiou AG, Johnson R, Murphy TD, Pertea M, Pruitt KD, Pujar S, Takahashi H, Ulitsky I, Varabyou A, Wells CA, Yandell M, Carninci P, Salzberg SL. The status of the human gene catalogue. Nature 2023; 622:41-47. [PMID: 37794265 PMCID: PMC10575709 DOI: 10.1038/s41586-023-06490-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/27/2023] [Indexed: 10/06/2023]
Abstract
Scientists have been trying to identify every gene in the human genome since the initial draft was published in 2001. In the years since, much progress has been made in identifying protein-coding genes, currently estimated to number fewer than 20,000, with an ever-expanding number of distinct protein-coding isoforms. Here we review the status of the human gene catalogue and the efforts to complete it in recent years. Beside the ongoing annotation of protein-coding genes, their isoforms and pseudogenes, the invention of high-throughput RNA sequencing and other technological breakthroughs have led to a rapid growth in the number of reported non-coding RNA genes. For most of these non-coding RNAs, the functional relevance is currently unclear; we look at recent advances that offer paths forward to identifying their functions and towards eventually completing the human gene catalogue. Finally, we examine the need for a universal annotation standard that includes all medically significant genes and maintains their relationships with different reference genomes for the use of the human gene catalogue in clinical settings.
Collapse
Affiliation(s)
- Paulo Amaral
- INSPER Institute of Education and Research, Sao Paulo, Brazil
| | | | - Francisco M De La Vega
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Tempus Labs, Chicago, IL, USA
| | | | - Adam Frankish
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, UK
| | - Thomas Gingeras
- Department of Functional Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Roderic Guigo
- Centre for Genomic Regulation (CRG), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jennifer L Harrow
- Centre for Genomics Research, Discovery Sciences, AstraZeneca, Royston, UK
| | - Artemis G Hatzigeorgiou
- Department of Computer Science and Biomedical Informatics, Universithy of Thessaly, Lamia, Greece
- Hellenic Pasteur Institute, Athens, Greece
| | - Rory Johnson
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland
- Conway Institute of Biomedical and Biomolecular Research, University College Dublin, Dublin, Ireland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Terence D Murphy
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Mihaela Pertea
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
| | - Kim D Pruitt
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Shashikant Pujar
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Hazuki Takahashi
- Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Igor Ulitsky
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Ales Varabyou
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
| | - Christine A Wells
- Stem Cell Systems, Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Mark Yandell
- Departent of Human Genetics, Utah Center for Genetic Discovery, University of Utah, Salt Lake City, UT, USA
| | - Piero Carninci
- Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Human Technopole, Milan, Italy.
| | - Steven L Salzberg
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA.
- Department of Biostatistics, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
49
|
Wei M, Tang W, Lv D, Liu M, Wang G, Liu Q, Qin L, Huang B, Zhang D. Long-chain noncoding RNA sequencing analysis reveals the molecular profiles of chemically induced mammary epithelial cells. Front Genet 2023; 14:1189487. [PMID: 37745843 PMCID: PMC10514351 DOI: 10.3389/fgene.2023.1189487] [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: 03/19/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) were important regulators affecting the cellular reprogramming process. Previous studies from our group have demonstrated that small molecule compounds can induce goat ear fibroblasts to reprogram into mammary epithelial cells with lactation function. In this study, we used lncRNA-Sequencing (lncRNA-seq) to analyze the lncRNA expression profile of cells before and after reprogramming (CK vs. 5i8 d). The results showed that a total of 3,970 candidate differential lncRNAs were detected, 1,170 annotated and 2,800 new lncRNAs. Compared to 0 d cells, 738 lncRNAs were significantly upregulated and 550 were significantly downregulated in 8 d cells. Heat maps of lncrnas and target genes with significant differences showed that the fate of cell lineages changed. Functional enrichment analysis revealed that these differently expressed (DE) lncRNAs target genes were mainly involved in signaling pathways related to reprogramming and mammary gland development, such as the Wnt signaling pathway, PI3K-Akt signaling pathway, arginine and proline metabolism, ECM-receptor interaction, and MAPK signaling pathway. The accuracy of sequencing was verified by real-time fluorescence quantification (RT-qPCR) of lncRNAs and key candidate genes, and it was also demonstrated that the phenotype and genes of the cells were changed. Therefore, this study offers a foundation for explaining the molecular mechanisms of lncRNAs in chemically induced mammary epithelial cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ben Huang
- Guangxi Key Laboratory of Eye Health, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Dandan Zhang
- Guangxi Key Laboratory of Eye Health, Guangxi Academy of Medical Sciences, People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
50
|
Polina I, Mishra J, Cypress MW, Landherr M, Valkov N, Chaput I, Nieto B, Mende U, Zhang P, Jhun BS, O-Uchi J. Mitochondrial Ca 2+ uniporter (MCU) variants form plasma-membrane channels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.31.551242. [PMID: 37577584 PMCID: PMC10418069 DOI: 10.1101/2023.07.31.551242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
MCU is widely recognized as a responsible gene for encoding a pore-forming subunit of highly mitochondrial-specific and Ca 2+ -selective channel, mitochondrial Ca 2+ uniporter complex (mtCUC). Here, we report a novel short variant derived from the MCU gene (termed MCU-S) which lacks mitochondria-targeted sequence and forms a Ca 2+ - permeable channel outside of mitochondria. MCU-S was ubiquitously expressed in all cell-types/tissues, with particularly high expression in human platelets. MCU-S formed Ca 2+ channels at the plasma membrane, which exhibited similar channel properties to those observed in mtCUC. MCU-S channels at the plasma membrane served as an additional Ca 2+ influx pathway for platelet activation. Our finding is completely distinct from the originally reported MCU gene function and provides novel insights into the molecular basis of MCU variant-dependent cellular Ca 2+ handling.
Collapse
|