1
|
Xie H, Xiong T, Guan J, Han Y, Feng H, Xu F, Chen S, Li J, Xie Z, Liu D, Chen R. Induction of mitochondrial damage via the CREB3L1/miR-34c/COX1 axis by porcine epidemic diarrhea virus infection facilitates pathogenicity. J Virol 2025; 99:e0059124. [PMID: 40071922 PMCID: PMC11998543 DOI: 10.1128/jvi.00591-24] [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: 07/05/2024] [Accepted: 12/23/2024] [Indexed: 03/26/2025] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) is a primary cause of viral diarrhea in neonatal piglets, leading to substantial economic losses in the swine industry globally. It primarily targets epithelial cells of the small intestine, compromising intestinal function and resulting in the death of affected animals. As mitochondria are essential for maintaining gut health, this study investigates the effects of PEDV infection on mitochondrial function in small intestinal epithelial cells and its subsequent impacts. Using small RNA sequencing, fluorescence in situ hybridization, dual luciferase reporter assay, gene overexpression, and silencing experiments, we investigated the mitochondrial structural and functional impairments induced by PEDV infection in jejunum epithelial cells of piglets and characterized the regulatory pattern of miRNAs in mitochondria of jejunum epithelial cells during PEDV infection. The results indicate that PEDV infection leads to the upregulation and mitochondrial localization of the nuclear-encoded microRNA, miR-34c, which in turn suppresses COX1 expression. The activation of the miR-34c/COX1 axis diminishes mitochondrial complex III, IV, and V activities, depletes ATP, lowers mitochondrial oxygen consumption, induces mitochondrial depolarization, increases the accumulation of mitochondrial reactive oxygen species (mtROS), and stimulates mitophagy. Furthermore, we confirm that CREB3L1 acts as an upstream transcription factor regulating the miR-34c/COX1 axis during PEDV infection, modulating mitochondrial damage in the epithelial cells of the jejunum. These findings demonstrate for the first time that PEDV infection activates the miR-34c/COX1 axis via the transcription factor CREB3L1 and regulates the nuclear-mitochondrial communication and mitochondrial fate, providing a new perspective on the pathogenesis of PEDV.IMPORTANCEThis study reveals the mechanism by which the porcine epidemic diarrhea virus (PEDV) disrupts mitochondrial function in piglets, enhancing viral pathogenicity. By demonstrating how PEDV infection upregulates miR-34c, leading to COX1 suppression and subsequent mitochondrial dysfunction, the research highlights a novel aspect of viral manipulation of host cellular mechanisms. These findings provide a deeper understanding of the PEDV pathogenesis and identify potential targets for therapeutic intervention, advancing efforts to mitigate the economic impact of PEDV on the swine industry.
Collapse
Affiliation(s)
- Hangao Xie
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, China
| | - Ting Xiong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, China
| | - Jinlian Guan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, China
| | - Yin Han
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, China
| | - Haixia Feng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Fei Xu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Sixuan Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jiahui Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ziwei Xie
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, China
| | - Dingxiang Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, China
- Integrative Microbiology Research Centre, South China Agricultural University Integrative Microbiology Research Centre, Guangzhou, China
| | - Ruiai Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, China
- Key Laboratory of Manufacture Technology of Veterinary Bioproducts, Ministry of Agriculture and Rural Affairs, Beijing, China
- Guangdong Enterprise Key Laboratory of Biotechnology R&D of Veterinary Biologics, Zhaoqing, China
- Zhaoqing Dahuanong Biology Medicine Co. Ltd., Zhaoqing, China
| |
Collapse
|
2
|
Altan Z, Johnson R. Targeting Wnt signalling through LINC02418: insights from CRISPR screens. Gut 2025; 74:513-515. [PMID: 39978931 DOI: 10.1136/gutjnl-2024-334266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 01/31/2025] [Indexed: 02/22/2025]
Affiliation(s)
- Zekiye Altan
- School of Biology and Environmental Science, University College Dublin, Dublin, Leinster, Ireland
- School of Medicine, University College Dublin, Dublin, Leinster, Ireland
| | - Rory Johnson
- School of Biology and Environmental Science, University College Dublin, Dublin, Leinster, Ireland
- University College Dublin Conway Institute of Biomolecular and Biomedical Research, Dublin, Leinster, Ireland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Medical Oncology, University of Bern, Bern, Switzerland
| |
Collapse
|
3
|
Schwarzmueller LJ, Adam RS, Moreno LF, Nijman LE, Logiantara A, Eleonora S, Bril O, Vromans S, de Groot NE, Giugliano FP, Stepanova E, Muncan V, Elbers CC, Lenos KJ, Zwijnenburg DA, van Eijndhoven MAJ, Pegtel DM, van Neerven SM, Loayza-Puch F, Dadali T, Broom WJ, Maier MA, Koster J, Vermeulen L, Léveillé N. Identifying colorectal cancer-specific vulnerabilities in the Wnt-driven long non-coding transcriptome. Gut 2025; 74:571-585. [PMID: 39562049 PMCID: PMC12013597 DOI: 10.1136/gutjnl-2024-332752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/31/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Aberrant Wnt pathway activation is a key driver of colorectal cancer (CRC) and is essential to sustain tumour growth and progression. Although the downstream protein-coding target genes of the Wnt cascade are well known, the long non-coding transcriptome has not yet been fully resolved. OBJECTIVE In this study, we aim to comprehensively reveal the Wnt-regulated long non-coding transcriptome and exploit essential molecules as novel therapeutic targets. DESIGN We used global run-on sequencing to define β-catenin-regulated long non-coding RNAs (lncRNAs) in CRC. CRISPRi dropout screens were subsequently used to establish the functional relevance of a subset of these lncRNAs for long-term expansion of CRC. RESULTS We uncovered that LINC02418 is essential for cancer cell clonogenic outgrowth. Mechanistically, LINC02418 regulates MYC expression levels to promote CRC stem cell functionality and prevent terminal differentiation. Furthermore, we developed effective small interfering RNA (siRNA)-based therapeutics to target LINC02418 RNA in vivo. CONCLUSION We propose that cancer-specific Wnt-regulated lncRNAs provide novel therapeutic opportunities to interfere with the Wnt pathway, which has so far defied effective pharmacological inhibition.
Collapse
Affiliation(s)
- Laura J Schwarzmueller
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Ronja S Adam
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Leandro F Moreno
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Lisanne E Nijman
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Adrian Logiantara
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Steven Eleonora
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Oscar Bril
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Sophie Vromans
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Nina E de Groot
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Francesca Paola Giugliano
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ekaterina Stepanova
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Vanesa Muncan
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Clara C Elbers
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Kristiaan J Lenos
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Danny A Zwijnenburg
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | | | - Dirk Michiel Pegtel
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Sanne M van Neerven
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Fabricio Loayza-Puch
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tulin Dadali
- Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts, USA
| | - Wendy J Broom
- Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts, USA
| | - Martin A Maier
- Alnylam Pharmaceuticals Inc, Cambridge, Massachusetts, USA
| | - Jan Koster
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Nicolas Léveillé
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Sobański D, Sobańska M, Staszkiewicz R, Strojny D, Grabarek BO. Changes in the Expression Profile of Growth-Associated Protein 43 in Degenerative Lumbosacral Stenosis. J Clin Med 2025; 14:1223. [PMID: 40004753 PMCID: PMC11856692 DOI: 10.3390/jcm14041223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Degenerative spinal stenosis is a common condition associated with structural degeneration and pain, yet its molecular underpinnings remain incompletely understood. Growth-associated protein 43 (GAP-43), a key player in neuronal plasticity and regeneration, may serve as a biomarker for disease progression and pain severity. This study investigates the expression of GAP-43 at the mRNA and protein levels in the ligamentum flavum of affected patients. Methods: Samples were collected from 96 patients with degenerative spinal stenosis and 85 controls. GAP-43 mRNA expression was analyzed using reverse transcription-quantitative polymerase chain reaction (RT-qPCR), while protein levels were quantified via enzyme-linked immunosorbent assay (ELISA) and Western blot. Pain severity was assessed using the visual analog scale (VAS), and associations with lifestyle factors were analyzed. Results:GAP-43 mRNA expression was significantly downregulated in the study group compared to the controls (fold change = 0.58 ± 0.12, p < 0.05), with an inverse correlation to VAS pain severity (fold change = 0.76 at VAS 4 vs. 0.36 at VAS 10). Conversely, GAP-43 protein levels were markedly elevated in the study group (5.57 ± 0.21 ng/mL) when compared to controls (0.54 ± 0.87 ng/mL, p < 0.0001). Protein levels were also correlated with lifestyle factors, including smoking and alcohol consumption (p < 0.05). Conclusions: GAP-43 shows potential as a biomarker for pain severity and disease progression in degenerative spinal stenosis, in a manner influenced by lifestyle factors. Further research is needed to explore its diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Dawid Sobański
- Department of Neurosurgery, Szpital sw. Rafala in Cracow, 30-693 Cracow, Poland;
- Collegium Medicum, WSB University, 41-300 Dabrowa Gornicza, Poland; (R.S.); (D.S.); (B.O.G.)
| | - Małgorzata Sobańska
- Department of Neurosurgery, Szpital sw. Rafala in Cracow, 30-693 Cracow, Poland;
- Collegium Medicum, WSB University, 41-300 Dabrowa Gornicza, Poland; (R.S.); (D.S.); (B.O.G.)
| | - Rafał Staszkiewicz
- Collegium Medicum, WSB University, 41-300 Dabrowa Gornicza, Poland; (R.S.); (D.S.); (B.O.G.)
- Department of Neurosurgery, 5th Military Clinical Hospital with the SP ZOZ Polyclinic in Krakow, 30-901 Krakow, Poland
- Department of Neurosurgery, Faculty of Medicine in Zabrze, Academy of Silesia, 40-555 Katowice, Poland
| | - Damian Strojny
- Collegium Medicum, WSB University, 41-300 Dabrowa Gornicza, Poland; (R.S.); (D.S.); (B.O.G.)
- Institute of Health Care, National Academy of Applied Sciences in Przemyśl, 37-700 Przemyśl, Poland
- New Medical Techniques Specialist Hospital of St. Family in Rudna Mała, 36-060 Rzeszów, Poland
| | - Beniamin Oskar Grabarek
- Collegium Medicum, WSB University, 41-300 Dabrowa Gornicza, Poland; (R.S.); (D.S.); (B.O.G.)
| |
Collapse
|
5
|
Farberov S, Ziv O, Lau JY, Ben-Tov Perry R, Lubelsky Y, Miska E, Kudla G, Ulitsky I. Structural features within the NORAD long noncoding RNA underlie efficient repression of Pumilio activity. Nat Struct Mol Biol 2025; 32:287-299. [PMID: 39327473 DOI: 10.1038/s41594-024-01393-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 08/27/2024] [Indexed: 09/28/2024]
Abstract
Long noncoding RNAs (lncRNAs) are increasingly appreciated for their important functions in mammalian cells. However, how their functional capacities are encoded in their sequences and manifested in their structures remains largely unknown. Some lncRNAs bind to and modulate the availability of RNA-binding proteins, but the structural principles that underlie this mode of regulation are unknown. The NORAD lncRNA is a known decoy for Pumilio proteins, which modulate the translation and stability of hundreds of messenger RNAs and, consequently, a regulator of genomic stability and aging. Here we probed the RNA structure and long-range RNA-RNA interactions formed by human NORAD inside cells under different stressful conditions. We discovered a highly modular structure consisting of well-defined domains that contribute independently to NORAD function. Following arsenite stress, most structural domains undergo relaxation and form interactions with other RNAs that are targeted to stress granules. We further revealed a unique structural organization that spatially clusters the multiple Pumilio binding sites along NORAD and consequently contributes to the derepression of Pumilio targets. We then applied these structural principles to design an effective artificial decoy for the let-7 microRNA. Our work demonstrates how the sequence of a lncRNA spatially clusters its function into separated domains and how structural principles can be employed for the rational design of lncRNAs with desired activities.
Collapse
Affiliation(s)
- Svetlana Farberov
- Department of Immunology and Regenerative Biology and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Omer Ziv
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK.
- Eleven Therapeutics, Cambridge, UK.
| | - Jian You Lau
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh, UK
| | - Rotem Ben-Tov Perry
- Department of Immunology and Regenerative Biology and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Yoav Lubelsky
- Department of Immunology and Regenerative Biology and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Eric Miska
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK.
| | - Grzegorz Kudla
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh, UK.
| | - Igor Ulitsky
- Department of Immunology and Regenerative Biology and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
6
|
Thakore P, Karki S, Hrdlicka HC, Garcia-Munoz J, Pereira RC, Delany AM. Decreasing miR-433-3p Activity in the Osteoblast Lineage Blunts Glucocorticoid-mediated Bone Loss. Endocrinology 2025; 166:bqaf008. [PMID: 39820728 PMCID: PMC11791524 DOI: 10.1210/endocr/bqaf008] [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: 08/27/2024] [Revised: 12/05/2024] [Accepted: 01/15/2025] [Indexed: 01/19/2025]
Abstract
Glucocorticoid excess causes bone loss due to decreased bone formation and increased bone resorption; miR-433-3p is a microRNA (miRNA) that negatively regulates bone formation in male mice by targeting Runx2 as well as RNAs involved in Wnt, protein kinase A, and endogenous glucocorticoid signaling. To examine the impact of miR-433-3p on glucocorticoid-mediated bone loss, transgenic mice expressing a miR-433-3p tough decoy inhibitor in the osteoblast lineage were administered prednisolone via slow-release pellets. Bone loss was greater in control mice treated with prednisolone compared with miR-433-3p tough decoy mice due to higher osteoclast activity in the controls. In whole femurs, Rankl was significantly higher in prednisolone-treated controls compared with miR-433-3p tough decoy mice. Surprisingly, negative regulators of Wnt signaling Sost and Dkk1 were higher in miR-433-3p tough decoy mice and were unaffected by prednisolone. Luciferase- 3'-untranslated region reporter assays demonstrated that Sost is a novel miR-433-3p target, whereas Dkk1 is a previously validated miR-433-3p target. miR-433-3p levels are lower in matrix-synthesizing osteoblasts than in more osteocytic cells; thus the impact of miR-433-3p on the osteoblast lineage may be dependent on cell context: it is a negative regulator in matrix-depositing osteoblasts by targeting RNAs important for differentiation and function but a positive regulator in osteocytes, due to its ability to target prominently expressed negative regulators of Wnt signaling, Sost and Dkk1. The mechanisms by which miR-433-3p indirectly regulates glucocorticoid-mediated osteoclastogenesis remain unknown. However, we speculate that this regulation may be mediated by miR-433-3p activity in osteocytes, which play an important role in controlling osteoclastogenesis.
Collapse
Affiliation(s)
- Prachi Thakore
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - Sangita Karki
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - Henry C Hrdlicka
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - John Garcia-Munoz
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - Renata C Pereira
- Division of Pediatric Nephrology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Anne M Delany
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| |
Collapse
|
7
|
Yu TWB. A phenotypic drug discovery approach by latent interaction in deep learning. ROYAL SOCIETY OPEN SCIENCE 2024; 11:240720. [PMID: 40191531 PMCID: PMC11972434 DOI: 10.1098/rsos.240720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 04/09/2025]
Abstract
Contemporary drug discovery paradigms rely heavily on binding assays about the bio-physicochemical processes. However, this dominant approach suffers from overlooked higher-order interactions arising from the intricacies of molecular mechanisms, such as those involving cis-regulatory elements. It introduces potential impairments and restrains the potential development of computational methods. To address this limitation, I developed a deep learning model that leverages an end-to-end approach, relying exclusively on therapeutic information about drugs. By transforming textual representations of drug and virus genetic information into high-dimensional latent representations, this method evades the challenges arising from insufficient information about binding specificities. Its strengths lie in its ability to implicitly consider complexities such as epistasis and chemical-genetic interactions, and to handle the pervasive challenge of data scarcity. Through various modeling skills and data augmentation techniques, the proposed model demonstrates outstanding performance in out-of-sample validations, even in scenarios with unknown complex interactions. Furthermore, the study highlights the importance of chemical diversity for model training. While the method showcases the feasibility of deep learning in data-scarce scenarios, it reveals a promising alternative for drug discovery in situations where knowledge of underlying mechanisms is limited.
Collapse
Affiliation(s)
- Tat Wai Billy Yu
- Macao Polytechnic University, Macau SAR, People’s Republic of China
| |
Collapse
|
8
|
Ma J, Pei J, Zhang X, Bai X, Ding S, Dai D. Colon cancer-associated transcript 1 ( CCAT1 ): A potential novel target in cancer therapy. Chin Med J (Engl) 2024; 137:2128-2130. [PMID: 38945528 PMCID: PMC11374205 DOI: 10.1097/cm9.0000000000003092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Indexed: 07/02/2024] Open
Affiliation(s)
- Jinjie Ma
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Junpeng Pei
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Xueping Zhang
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Xiao Bai
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Siqi Ding
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Dongqiu Dai
- Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
- Cancer Center, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| |
Collapse
|
9
|
Verheyden NA, Klostermann M, Brüggemann M, Steede H, Scholz A, Amr S, Lichtenthaeler C, Münch C, Schmid T, Zarnack K, Krueger A. A high-resolution map of functional miR-181 response elements in the thymus reveals the role of coding sequence targeting and an alternative seed match. Nucleic Acids Res 2024; 52:8515-8533. [PMID: 38783381 PMCID: PMC11317165 DOI: 10.1093/nar/gkae416] [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: 09/14/2023] [Revised: 04/25/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
MicroRNAs (miRNAs) are critical post-transcriptional regulators in many biological processes. They act by guiding RNA-induced silencing complexes to miRNA response elements (MREs) in target mRNAs, inducing translational inhibition and/or mRNA degradation. Functional MREs are expected to predominantly occur in the 3' untranslated region and involve perfect base-pairing of the miRNA seed. Here, we generate a high-resolution map of miR-181a/b-1 (miR-181) MREs to define the targeting rules of miR-181 in developing murine T cells. By combining a multi-omics approach with computational high-resolution analyses, we uncover novel miR-181 targets and demonstrate that miR-181 acts predominantly through RNA destabilization. Importantly, we discover an alternative seed match and identify a distinct set of targets with repeat elements in the coding sequence which are targeted by miR-181 and mediate translational inhibition. In conclusion, deep profiling of MREs in primary cells is critical to expand physiologically relevant targetomes and establish context-dependent miRNA targeting rules.
Collapse
Affiliation(s)
- Nikita A Verheyden
- Molecular Immunology, Justus Liebig University Gießen, 35392 Gießen, Germany
| | - Melina Klostermann
- Buchmann Institute for Molecular Life Sciences & Institute of Molecular Biosciences, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| | - Mirko Brüggemann
- Buchmann Institute for Molecular Life Sciences & Institute of Molecular Biosciences, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| | - Hanna M Steede
- Molecular Immunology, Justus Liebig University Gießen, 35392 Gießen, Germany
| | - Anica Scholz
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Shady Amr
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Chiara Lichtenthaeler
- Institute of Molecular Medicine, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Christian Münch
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Tobias Schmid
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Kathi Zarnack
- Buchmann Institute for Molecular Life Sciences & Institute of Molecular Biosciences, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| | - Andreas Krueger
- Molecular Immunology, Justus Liebig University Gießen, 35392 Gießen, Germany
| |
Collapse
|
10
|
Solovev YV, Evpak AS, Kudriaeva AA, Gabibov AG, Belogurov AA. Evaluation of Clinically Significant miRNAs Level by Machine Learning Approaches Utilizing Total Transcriptome Data. DOKL BIOCHEM BIOPHYS 2024; 516:98-106. [PMID: 38539010 DOI: 10.1134/s1607672924700790] [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: 12/28/2023] [Revised: 01/25/2024] [Accepted: 02/03/2024] [Indexed: 05/26/2024]
Abstract
Analysis of the mechanisms underlying the occurrence and progression of cancer represents a key objective in contemporary clinical bioinformatics and molecular biology. Utilizing omics data, particularly transcriptomes, enables a detailed characterization of expression patterns and post-transcriptional regulation across various RNA types relative to the entire transcriptome. Here, we assembled a dataset comprising transcriptomic data from approximately 16 000 patients encompassing over 160 types of cancer. We employed state-of-the-art gradient boosting algorithms to discern intricate correlations in the expression levels of four clinically significant microRNAs, specifically, hsa-mir-21, hsa-let-7a-1, hsa-let-7b, and hsa-let-7i, with the expression levels of the remaining 60 660 unique RNAs. Our analysis revealed a dependence of the expression levels of the studied microRNAs on the concentrations of several small nucleolar RNAs and regulatory long noncoding RNAs. Notably, the roles of these RNAs in the development of specific cancer types had been previously established through experimental evidence. Subsequent evaluation of the created database will facilitate the identification of a broader spectrum of overarching dependencies related to changes in the expression levels of various RNA classes in diverse cancers. In future, it will make possible to discover unique alterations specific to certain types of malignant transformations.
Collapse
Affiliation(s)
- Ya V Solovev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.
| | - A S Evpak
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.
| | - A A Kudriaeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - A G Gabibov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Moscow State University, Moscow, Russia
| | - A A Belogurov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Moscow State University of Medicine and Dentistry, 127473, Moscow, Russia
| |
Collapse
|
11
|
Xing W, Li S. LncRNA ENSGALG00000021686 regulates fat metabolism in chicken hepatocytes via miR-146b/AGPAT2 pathway. Anim Genet 2024; 55:420-429. [PMID: 38369771 DOI: 10.1111/age.13405] [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: 12/03/2023] [Revised: 01/10/2024] [Accepted: 01/31/2024] [Indexed: 02/20/2024]
Abstract
The liver contributes to lipid metabolism as the hub of fat synthesis. Long non-coding RNAs (lncRNAs) are considered the regulators of cellular processes. Since LncRNA ENSGALG00000021686 (lncRNA 21 686) has been described as a regulator of lipid metabolism, the present study aimed to clarify the role of lncRNA 21 686 in chicken hepatocytes' lipid metabolism. Thirty-two chickens were divided into four groups and were treated with diets containing different amounts of fat, and the hepatic expression of lncRNA 21 686 and miR-146b along with the levels of proteins involved in the regulation of fat metabolism, lipid indices and oxidative stress were measured. Moreover, primary chicken hepatocytes were transfected with lncRNA 21 686 small interfering RNA or microRNA (miRNA, miR)-146b mimics to measure the consequences of suppressing lncRNA or inducing miRNA expression on the levels of proteins involved in fat metabolism and stress markers. The results showed that the high-fat diet modulated the expression of lncRNA 21 686 and miR-146b (p-value < 0.001). Moreover, there was a significant increase in 1-acyl-sn-glycerol-3-phosphate acyltransferase 2 (AGPAT2) gene expression and protein levels and modulated fat-related markers. Furthermore, the results showed that lncRNA 21 686 suppression reduced the expression of AGPAT2 and its downstream proteins (p-value < 0.05). Overexpression of miR-146b regulated fat metabolism indicator expression. Transfection experiments revealed that lncRNA 21 686 suppression increased miR-146b expression. The findings suggested a novel mechanism containing lncRNA 21 686/miR-146b/AGPAT2 in the regulation of fat metabolism in chicken hepatocytes.
Collapse
Affiliation(s)
- Wenhao Xing
- State Key Laboratory of Animal Nutrition, Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Shijie Li
- Dongying Jintengsheng Medical Device Sales Co., Ltd., Dongying, Shandong Province, China
| |
Collapse
|
12
|
Fuchs Wightman F, Lukin J, Giusti S, Soutschek M, Bragado L, Pozzi B, Pierelli M, González P, Fededa J, Schratt G, Fujiwara R, Wilusz J, Refojo D, de la Mata M. Influence of RNA circularity on Target RNA-Directed MicroRNA Degradation. Nucleic Acids Res 2024; 52:3358-3374. [PMID: 38381063 PMCID: PMC11014252 DOI: 10.1093/nar/gkae094] [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: 08/12/2023] [Revised: 01/03/2024] [Accepted: 01/30/2024] [Indexed: 02/22/2024] Open
Abstract
A subset of circular RNAs (circRNAs) and linear RNAs have been proposed to 'sponge' or block microRNA activity. Additionally, certain RNAs induce microRNA destruction through the process of Target RNA-Directed MicroRNA Degradation (TDMD), but whether both linear and circular transcripts are equivalent in driving TDMD is unknown. Here, we studied whether circular/linear topology of endogenous and artificial RNA targets affects TDMD. Consistent with previous knowledge that Cdr1as (ciRS-7) circular RNA protects miR-7 from Cyrano-mediated TDMD, we demonstrate that depletion of Cdr1as reduces miR-7 abundance. In contrast, overexpression of an artificial linear version of Cdr1as drives miR-7 degradation. Using plasmids that express a circRNA with minimal co-expressed cognate linear RNA, we show differential effects on TDMD that cannot be attributed to the nucleotide sequence, as the TDMD properties of a sequence often differ when in a circular versus linear form. By analysing RNA sequencing data of a neuron differentiation system, we further detect potential effects of circRNAs on microRNA stability. Our results support the view that RNA circularity influences TDMD, either enhancing or inhibiting it on specific microRNAs.
Collapse
Affiliation(s)
- Federico Fuchs Wightman
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires 1428, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Buenos Aires 1428, Argentina
| | - Jerónimo Lukin
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Godoy Cruz 2390, C1425FQD Buenos Aires 1425, Argentina
| | - Sebastián A Giusti
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Godoy Cruz 2390, C1425FQD Buenos Aires 1425, Argentina
| | - Michael Soutschek
- Lab of Systems Neuroscience, D-HEST Institute for Neuroscience, ETH Zürich 8092, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, ETH Zürich 8092, Switzerland
| | - Laureano Bragado
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires 1428, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Buenos Aires 1428, Argentina
| | - Berta Pozzi
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires 1428, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Buenos Aires 1428, Argentina
- Institute of Cell Biology, University of Bern, Bern 3012, Switzerland
| | - María L Pierelli
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires 1428, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Buenos Aires 1428, Argentina
| | - Paula González
- Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo A. Ugalde”, IIB-UNSAM, IIBIO-CONICET, Universidad Nacional de San Martín, Buenos Aires 1650, Argentina
| | - Juan P Fededa
- Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo A. Ugalde”, IIB-UNSAM, IIBIO-CONICET, Universidad Nacional de San Martín, Buenos Aires 1650, Argentina
| | - Gerhard Schratt
- Lab of Systems Neuroscience, D-HEST Institute for Neuroscience, ETH Zürich 8092, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, ETH Zürich 8092, Switzerland
| | - Rina Fujiwara
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Therapeutic Innovation Center, Baylor College of Medicine, Houston, TX77030, USA
| | - Jeremy E Wilusz
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Therapeutic Innovation Center, Baylor College of Medicine, Houston, TX77030, USA
| | - Damián Refojo
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Godoy Cruz 2390, C1425FQD Buenos Aires 1425, Argentina
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Manuel de la Mata
- CONICET-Universidad de Buenos Aires, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Buenos Aires 1428, Argentina
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales. Departamento de Fisiología, Biología Molecular y Celular, Buenos Aires 1428, Argentina
| |
Collapse
|
13
|
Joshi R, Sharma A, Kulshreshtha R. Noncoding RNA landscape and their emerging roles as biomarkers and therapeutic targets in meningioma. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200782. [PMID: 38596289 PMCID: PMC10951709 DOI: 10.1016/j.omton.2024.200782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Meningiomas are among the most prevalent primary CNS tumors in adults, accounting for nearly 38% of all brain neoplasms. The World Health Organization (WHO) grade assigned to meningiomas guides medical care in patients and is primarily based on tumor histology and malignancy potential. Although often considered benign, meningiomas with complicated histology, limited accessibility for surgical resection, and/or higher malignancy potential (WHO grade 2 and WHO grade 3) are harder to combat, resulting in significant morbidity. With limited treatment options and no systemic therapies, it is imperative to understand meningioma tumorigenesis at the molecular level and identify novel therapeutic targets. The last decade witnessed considerable progress in understanding the noncoding RNA landscape of meningioma, with microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) emerging as molecular entities of interest. This review aims to highlight the commonly dysregulated miRNAs and lncRNAs in meningioma and their correlation with meningioma progression, malignancy, recurrence, and radioresistance. The role of "key" miRNAs as biomarkers and their therapeutic potential has also been reviewed in detail. Furthermore, current and emerging therapeutic modalities for meningioma have been discussed, with emphasis on the need to identify and subsequently employ clinically relevant miRNAs and lncRNAs as novel therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Ritanksha Joshi
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Anuja Sharma
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi 110016, India
| |
Collapse
|
14
|
Sobański D, Bogdał P, Staszkiewicz R, Sobańska M, Filipowicz M, Czepko RA, Strojny D, Grabarek BO. Evaluation of differences in expression pattern of three isoforms of the transforming growth factor beta in patients with lumbosacral stenosis. Cell Cycle 2024; 23:555-572. [PMID: 38695374 PMCID: PMC11135850 DOI: 10.1080/15384101.2024.2345484] [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: 01/30/2024] [Accepted: 03/30/2024] [Indexed: 05/28/2024] Open
Abstract
The study investigates molecular changes in the lumbosacral (L/S) spine's yellow ligamentum flavum during degenerative stenosis, focusing on the role of transforming growth factor beta 1-3 (TGF-β-1-3). Sixty patients with degenerative stenosis and sixty control participants underwent molecular analysis using real-time quantitative reverse transcription reaction technique (RTqPCR), enzyme-linked immunosorbent assay (ELISA), Western blot, and immunohistochemical analysis (IHC). At the mRNA level, study samples showed reduced expression of TGF-β-1 and TGF-β-3, while TGF-β-2 increased by only 4%. Conversely, at the protein level, the study group exhibited significantly higher concentrations of TGF-β-1, TGF-β-2, and TGF-β-3 compared to controls. On the other hand, at the protein level, a statistically significant higher concentration of TGF-β-1 was observed (2139.33 pg/mL ± 2593.72 pg/mL vs. 252.45 pg/mL ± 83.89 pg/mL; p < 0.0001), TGF-β-2 (3104.34 pg/mL ± 1192.74 pg/mL vs. 258.86 pg/mL ± 82.98 pg/mL; p < 0.0001), TGF-β-3 (512.75 pg/mL ± 107.36 pg/mL vs. 55.06 pg/mL ± 9.83 pg/mL, p < 0.0001) in yellow ligaments obtained from patients of the study group compared to control samples. The study did not establish a significant correlation between TGF-β-1-3 concentrations and pain severity. The findings suggest that molecular therapy aimed at restoring the normal expression pattern of TGF-β-1-3 could be a promising strategy for treating degenerative stenosis of the L/S spine. The study underscores the potential therapeutic significance of addressing molecular changes at the TGF-β isoforms level for better understanding and managing degenerative spinal conditions.
Collapse
Affiliation(s)
- Dawid Sobański
- Department of Neurosurgery, Szpital sw. Rafala in Cracow, Cracow, Poland
- Collegium Medicum, WSB University, Dabrowa Gornicza, Poland
| | - Paweł Bogdał
- Department of Orthopedic, Szpital Powiatowy w Zawierciu, Zawiercie, Poland
| | - Rafał Staszkiewicz
- Collegium Medicum, WSB University, Dabrowa Gornicza, Poland
- Department of Neurosurgery, 5th Military Clinical Hospital with the SP ZOZ Polyclinic in Krakow, Krakow, Poland
- Department of Neurosurgery, Faculty of Medicine in Zabrze, Academy of Silesia, Katowice, Poland
| | | | - Michał Filipowicz
- Department of Neurosurgery, Szpital sw. Rafala in Cracow, Cracow, Poland
| | - Ryszard Adam Czepko
- Department of Neurosurgery, Szpital sw. Rafala in Cracow, Cracow, Poland
- Department of Neurosurgery, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski University in Cracow, Cracow, Poland
| | - Damian Strojny
- Collegium Medicum, WSB University, Dabrowa Gornicza, Poland
- Institute of Health Care, National Academy of Applied Sciences in Przemyśl, Przemyśl, Poland
- Department of Medical Science, New Medical Techniques Specialist Hospital of St. Family in Rudna Mała, Rzeszów, Poland
| | | |
Collapse
|
15
|
Ilker E, Hinczewski M. Bioenergetic costs and the evolution of noise regulation by microRNAs. Proc Natl Acad Sci U S A 2024; 121:e2308796121. [PMID: 38386708 PMCID: PMC10907262 DOI: 10.1073/pnas.2308796121] [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: 05/27/2023] [Accepted: 01/14/2024] [Indexed: 02/24/2024] Open
Abstract
Noise control, together with other regulatory functions facilitated by microRNAs (miRNAs), is believed to have played important roles in the evolution of multicellular eukaryotic organisms. miRNAs can dampen protein fluctuations via enhanced degradation of messenger RNA (mRNA), but this requires compensation by increased mRNA transcription to maintain the same expression levels. The overall mechanism is metabolically expensive, leading to questions about how it might have evolved in the first place. We develop a stochastic model of miRNA noise regulation, coupled with a detailed analysis of the associated metabolic costs. Additionally, we calculate binding free energies for a range of miRNA seeds, the short sequences which govern target recognition. We argue that natural selection may have fine-tuned the Michaelis-Menten constant [Formula: see text] describing miRNA-mRNA affinity and show supporting evidence from analysis of experimental data. [Formula: see text] is constrained by seed length, and optimal noise control (minimum protein variance at a given energy cost) is achievable for seeds of 6 to 7 nucleotides in length, the most commonly observed types. Moreover, at optimality, the degree of noise reduction approaches the theoretical bound set by the Wiener-Kolmogorov linear filter. The results illustrate how selective pressure toward energy efficiency has potentially shaped a crucial regulatory pathway in eukaryotes.
Collapse
Affiliation(s)
- Efe Ilker
- Max Planck Institute for the Physics of Complex Systems, Dresden01187, Germany
| | - Michael Hinczewski
- Department of Physics, Case Western Reserve University, Cleveland, OH44106
| |
Collapse
|
16
|
Staszkiewicz R, Gładysz D, Sobański D, Bolechała F, Golec E, Dammermann W, Grabarek BO. The Impacts of Intervertebral Disc Degeneration of the Spine, Alcohol Consumption, Smoking Tobacco Products, and Glycemic Disorders on the Expression Profiles of Neurotrophins-3 and -4. Biomedicines 2024; 12:427. [PMID: 38398029 PMCID: PMC10886622 DOI: 10.3390/biomedicines12020427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
In the etiology of discogenic pain, attention is paid to the role of neurotrophic factors, which include classic neurotrophins (NTs). This study aimed to assess changes in the concentrations of NT-3 and NT-4 in the intervertebral discs (IVDs) of the lumbosacral (L/S) spine depending on the advancement of degenerative changes, pain severity, habits, and comorbidities. The study group included 113 patients who underwent microdiscectomy due to degenerative IVD disease of the L/S spine. The severity of degenerative IVD changes was assessed using the five-point Pfirrmann scale, and the pain intensity was assessed according to the visual analog scale (VAS). In turn, the control group included 81 participants from whom IVDs of the L/S section of the spine were collected post-mortem during forensic autopsy or organ donation. At the mRNA level, we noted NT-3 overexpression in the test samples compared with the controls (fold change (FC) = 9.12 ± 0.56; p < 0.05), while NT-4 transcriptional activity was decreased in the test samples compared with the controls (FC = 0.33 ± 0.07; p < 0.05). However, at the protein level, the concentrations of NT-3 (134 ± 5.78 pg/mL vs. 6.78 ± 1.17 pg/mL; p < 0.05) and NT-4 (316.77 ± 8.19 pg/mL vs. 76.92 ± 4.82 pg/mL; p < 0.05) were significantly higher in the test samples compared with the control samples. Nevertheless, the concentration of both proteins did not statistically significantly change depending on the advancement of degenerative changes and the pain intensity (p > 0.05). In addition, higher levels of NT-3 and NT-4 were noted in IVD samples from patients who consumed alcohol, smoked tobacco, were overweight/obese, or had comorbid diabetes compared with patients without these risk factors (p < 0.05). Our analysis confirmed that differences in the degenerative process of IVD, energy metabolism, and lifestyle are related to changes in the concentration profiles of NT-3 and NT-4.
Collapse
Affiliation(s)
- Rafał Staszkiewicz
- Collegium Medicum, WSB University, 41-300 Dabrowa Gornicza, Poland;
- Department of Neurosurgery, 5th Military Clinical Hospital, SP ZOZ Polyclinic in Cracow, 30-901 Cracow, Poland;
- Department of Neurosurgery, Faculty of Medicine in Zabrze, Academy of Silesia, 40-555 Katowice, Poland
| | - Dorian Gładysz
- Department of Neurosurgery, 5th Military Clinical Hospital, SP ZOZ Polyclinic in Cracow, 30-901 Cracow, Poland;
- Department of Neurosurgery, Faculty of Medicine in Zabrze, Academy of Silesia, 40-555 Katowice, Poland
| | - Dawid Sobański
- Collegium Medicum, WSB University, 41-300 Dabrowa Gornicza, Poland;
- Department of Neurosurgery, Szpital sw. Rafala in Cracow, 30-693 Cracow, Poland
- Department of Neurosurgery, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski University in Cracow, 30-705 Cracow, Poland
| | - Filip Bolechała
- Department of Forensic Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland;
| | - Edward Golec
- Department of Rehabilitation in Orthopaedics, Faculty of Motor Rehabilitation, Bronisław Czech University of Physical Education, 31-571 Cracow, Poland;
| | - Werner Dammermann
- Center of Internal Medicine II, University Hospital Brandenburg, 03048 Brandenburg, Germany;
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany
| | - Beniamin Oskar Grabarek
- Collegium Medicum, WSB University, 41-300 Dabrowa Gornicza, Poland;
- Gyncentrum, Laboratory of Molecular Biology and Virology, Department of Molecular Biology, 40-851 Katowice, Poland
| |
Collapse
|
17
|
Li J, Huang S, Shi L, Chen G, Liu X, Liu M, Guo G. Interaction between long noncoding RNA and microRNA in lung inflammatory diseases. Immun Inflamm Dis 2024; 12:e1129. [PMID: 38270295 PMCID: PMC10777888 DOI: 10.1002/iid3.1129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 12/04/2023] [Accepted: 12/13/2023] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Non-coding RNAs (ncRNAs) are a group of RNAs that cannot synthesize proteins, but are critical in gene expression regulation. Long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), the two major family members, are intimately involved in controlling immune response, cell proliferation, apoptosis, differentiation and polarization, and cytokine secretion. Their interactions significantly influence lung inflammatory diseases and could be potential therapeutic targets. OBJECTIVES The review aims to elucidate the role of ncRNAs, especially the interactions between lncRNA and miRNA in lung diseases, including acute and chronic lung inflammatory diseases, as well as lung cancer. And provide novel insights into disease mechanisms and potential therapeutic methods. METHODS We conducted a comprehensive review of the latest studies on lncRNA and miRNA in lung inflammatory diseases. Our research involved searching through electronic databases like PubMed, Web of Science, and Scopus. RESULTS We explain the fundamental characteristics and functions of miRNA and lncRNA, their potential interaction mechanisms, and summarize the newly explorations on the role of lncRNA and miRNA interactions in lung inflammatory diseases. CONCLUSIONS Numerous lncRNAs and miRNAs have been found to partipicate in all stages of lung inflammatory diseases. While ncRNA-based therapies have been validated and developed, there remain challenges in developing more stable and effective drugs for clinical use.
Collapse
Affiliation(s)
- Jiaqi Li
- Medical Center of Burn Plastic and Wound RepairThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Shengyu Huang
- Medical Center of Burn Plastic and Wound RepairThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Liangliang Shi
- Medical Center of Burn Plastic and Wound RepairThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Guochang Chen
- Medical Center of Burn Plastic and Wound RepairThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Xiaoxiao Liu
- Medical Center of Burn Plastic and Wound RepairThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Mingzhuo Liu
- Medical Center of Burn Plastic and Wound RepairThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Guanghua Guo
- Medical Center of Burn Plastic and Wound RepairThe First Affiliated Hospital of Nanchang UniversityNanchangChina
| |
Collapse
|
18
|
Burkhart JG, Wu G, Song X, Raimondi F, McWeeney S, Wong MH, Deng Y. Biology-inspired graph neural network encodes reactome and reveals biochemical reactions of disease. PATTERNS (NEW YORK, N.Y.) 2023; 4:100758. [PMID: 37521042 PMCID: PMC10382942 DOI: 10.1016/j.patter.2023.100758] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/20/2022] [Accepted: 05/01/2023] [Indexed: 08/01/2023]
Abstract
Functional heterogeneity of healthy human tissues complicates interpretation of molecular studies, impeding precision therapeutic target identification and treatment. Considering this, we generated a graph neural network with Reactome-based architecture and trained it using 9,115 samples from Genotype-Tissue Expression (GTEx). Our graph neural network (GNN) achieves adjusted Rand index (ARI) = 0.7909, while a Resnet18 control model achieves ARI = 0.7781, on 370 held-out healthy human tissue samples from The Cancer Genome Atlas (TCGA), despite the Resnet18 using over 600 times the parameters. Our GNN also succeeds in separating 83 healthy skin samples from 95 lesional psoriasis samples, revealing that upregulation of 26S- and NUB1-mediated degradation of NEDD8, UBD, and their conjugates is central to the largest perturbed reaction network component in psoriasis. We show that our results are not discoverable using traditional differential expression and hypergeometric pathway enrichment analyses yet are supported by separate human multi-omics and small-molecule mouse studies, suggesting future molecular disease studies may benefit from similar GNN analytical approaches.
Collapse
Affiliation(s)
- Joshua G. Burkhart
- Department of Quantitative Health Sciences, University of Hawaii John A. Burns School of Medicine, Honolulu, HI 96813, USA
| | - Guanming Wu
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Xubo Song
- Department of Computer Science and Electrical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | | | - Shannon McWeeney
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Melissa H. Wong
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR 97201, USA
| | - Youping Deng
- Department of Quantitative Health Sciences, University of Hawaii John A. Burns School of Medicine, Honolulu, HI 96813, USA
| |
Collapse
|
19
|
Danckwardt S, Trégouët DA, Castoldi E. Post-transcriptional control of haemostatic genes: mechanisms and emerging therapeutic concepts in thrombo-inflammatory disorders. Cardiovasc Res 2023; 119:1624-1640. [PMID: 36943786 PMCID: PMC10325701 DOI: 10.1093/cvr/cvad046] [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: 09/26/2022] [Revised: 12/20/2022] [Accepted: 01/05/2023] [Indexed: 03/23/2023] Open
Abstract
The haemostatic system is pivotal to maintaining vascular integrity. Multiple components involved in blood coagulation have central functions in inflammation and immunity. A derailed haemostasis is common in prevalent pathologies such as sepsis, cardiovascular disorders, and lately, COVID-19. Physiological mechanisms limit the deleterious consequences of a hyperactivated haemostatic system through adaptive changes in gene expression. While this is mainly regulated at the level of transcription, co- and posttranscriptional mechanisms are increasingly perceived as central hubs governing multiple facets of the haemostatic system. This layer of regulation modulates the biogenesis of haemostatic components, for example in situations of increased turnover and demand. However, they can also be 'hijacked' in disease processes, thereby perpetuating and even causally entertaining associated pathologies. This review summarizes examples and emerging concepts that illustrate the importance of posttranscriptional mechanisms in haemostatic control and crosstalk with the immune system. It also discusses how such regulatory principles can be used to usher in new therapeutic concepts to combat global medical threats such as sepsis or cardiovascular disorders.
Collapse
Affiliation(s)
- Sven Danckwardt
- Centre for Thrombosis and Hemostasis (CTH), University Medical Centre
Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- German Centre for Cardiovascular Research (DZHK),
Berlin, Germany
- Posttranscriptional Gene Regulation, University Medical Centre
Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, University
Medical Centre Mainz, Langenbeckstr. 1, 55131
Mainz, Germany
- Center for Healthy Aging (CHA), Mainz,
Germany
| | - David-Alexandre Trégouët
- INSERM, Bordeaux Population Health Research Center, UMR 1219, Department of
Molecular Epidemiology of Vascular and Brain Disorders (ELEANOR), University of
Bordeaux, Bordeaux, France
| | - Elisabetta Castoldi
- Department of Biochemistry, Cardiovascular Research Institute Maastricht
(CARIM), Maastricht University, Universiteitsingel 50, 6229
ER Maastricht, The Netherlands
| |
Collapse
|
20
|
Walgrave H, Penning A, Tosoni G, Snoeck S, Davie K, Davis E, Wolfs L, Sierksma A, Mars M, Bu T, Thrupp N, Zhou L, Moechars D, Mancuso R, Fiers M, Howden AJ, De Strooper B, Salta E. microRNA-132 regulates gene expression programs involved in microglial homeostasis. iScience 2023; 26:106829. [PMID: 37250784 PMCID: PMC10213004 DOI: 10.1016/j.isci.2023.106829] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/13/2023] [Accepted: 05/03/2023] [Indexed: 05/31/2023] Open
Abstract
microRNA-132 (miR-132), a known neuronal regulator, is one of the most robustly downregulated microRNAs (miRNAs) in the brain of Alzheimer's disease (AD) patients. Increasing miR-132 in AD mouse brain ameliorates amyloid and Tau pathologies, and also restores adult hippocampal neurogenesis and memory deficits. However, the functional pleiotropy of miRNAs requires in-depth analysis of the effects of miR-132 supplementation before it can be moved forward for AD therapy. We employ here miR-132 loss- and gain-of-function approaches using single-cell transcriptomics, proteomics, and in silico AGO-CLIP datasets to identify molecular pathways targeted by miR-132 in mouse hippocampus. We find that miR-132 modulation significantly affects the transition of microglia from a disease-associated to a homeostatic cell state. We confirm the regulatory role of miR-132 in shifting microglial cell states using human microglial cultures derived from induced pluripotent stem cells.
Collapse
Affiliation(s)
- Hannah Walgrave
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), 3000 Leuven, Belgium
| | - Amber Penning
- Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands
| | - Giorgia Tosoni
- Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands
| | - Sarah Snoeck
- Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands
| | - Kristofer Davie
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Bioinformatics Core Facility, 3000 Leuven, Belgium
| | - Emma Davis
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
| | - Leen Wolfs
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), 3000 Leuven, Belgium
| | - Annerieke Sierksma
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), 3000 Leuven, Belgium
| | - Mayte Mars
- Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands
| | - Taofeng Bu
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), 3000 Leuven, Belgium
| | - Nicola Thrupp
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), 3000 Leuven, Belgium
| | - Lujia Zhou
- Discovery Neuroscience, Janssen Research and Development, Division of Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Diederik Moechars
- Discovery Neuroscience, Janssen Research and Development, Division of Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, 2610 Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Mark Fiers
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), 3000 Leuven, Belgium
| | - Andrew J.M. Howden
- UK Dementia Research Institute, University of Dundee, Dundee DD1 4HN, UK
| | - Bart De Strooper
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute (LBI), 3000 Leuven, Belgium
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
| | - Evgenia Salta
- Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands
| |
Collapse
|
21
|
Hwang H, Chang HR, Baek D. Determinants of Functional MicroRNA Targeting. Mol Cells 2023; 46:21-32. [PMID: 36697234 PMCID: PMC9880601 DOI: 10.14348/molcells.2023.2157] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 01/27/2023] Open
Abstract
MicroRNAs (miRNAs) play cardinal roles in regulating biological pathways and processes, resulting in significant physiological effects. To understand the complex regulatory network of miRNAs, previous studies have utilized massivescale datasets of miRNA targeting and attempted to computationally predict the functional targets of miRNAs. Many miRNA target prediction tools have been developed and are widely used by scientists from various fields of biology and medicine. Most of these tools consider seed pairing between miRNAs and their mRNA targets and additionally consider other determinants to improve prediction accuracy. However, these tools exhibit limited prediction accuracy and high false positive rates. The utilization of additional determinants, such as RNA modifications and RNA-binding protein binding sites, may further improve miRNA target prediction. In this review, we discuss the determinants of functional miRNA targeting that are currently used in miRNA target prediction and the potentially predictive but unappreciated determinants that may improve prediction accuracy.
Collapse
Affiliation(s)
- Hyeonseo Hwang
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Hee Ryung Chang
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Daehyun Baek
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
22
|
van Wijk N, Zohar K, Linial M. Challenging Cellular Homeostasis: Spatial and Temporal Regulation of miRNAs. Int J Mol Sci 2022; 23:16152. [PMID: 36555797 PMCID: PMC9787707 DOI: 10.3390/ijms232416152] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/11/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Mature microRNAs (miRNAs) are single-stranded non-coding RNA (ncRNA) molecules that act in post-transcriptional regulation in animals and plants. A mature miRNA is the end product of consecutive, highly regulated processing steps of the primary miRNA transcript. Following base-paring of the mature miRNA with its mRNA target, translation is inhibited, and the targeted mRNA is degraded. There are hundreds of miRNAs in each cell that work together to regulate cellular key processes, including development, differentiation, cell cycle, apoptosis, inflammation, viral infection, and more. In this review, we present an overlooked layer of cellular regulation that addresses cell dynamics affecting miRNA accessibility. We discuss the regulation of miRNA local storage and translocation among cell compartments. The local amounts of the miRNAs and their targets dictate their actual availability, which determines the ability to fine-tune cell responses to abrupt or chronic changes. We emphasize that changes in miRNA storage and compactization occur under induced stress and changing conditions. Furthermore, we demonstrate shared principles on cell physiology, governed by miRNA under oxidative stress, tumorigenesis, viral infection, or synaptic plasticity. The evidence presented in this review article highlights the importance of spatial and temporal miRNA regulation for cell physiology. We argue that limiting the research to mature miRNAs within the cytosol undermines our understanding of the efficacy of miRNAs to regulate cell fate under stress conditions.
Collapse
Affiliation(s)
| | | | - Michal Linial
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, Faculty of Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| |
Collapse
|
23
|
Liu N, Yang C, Gao A, Sun M, Lv D. MiR-101: An Important Regulator of Gene Expression and Tumor Ecosystem. Cancers (Basel) 2022; 14:cancers14235861. [PMID: 36497343 PMCID: PMC9739992 DOI: 10.3390/cancers14235861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
MiRNAs are small single-stranded non-coding RNAs. MiRNA contributes to the transcriptional and post-transcriptional regulation of mRNA in different cell types, including mRNA transcription inhibition and mRNA decay and phenotypes via the effect of several essential oncogenic processes and tumor microenvironment. MiR-101 is a highly conserved miRNA that was found to alter the expression in various human cancers. MiR-101 has been reported to have tumor oncogenic and suppressive effects to regulate tumorigenesis and tumor progression. In this review, we summarize the new findings about the roles of miR-101 in cancers and the underlying mechanisms of targeting genes degradation and microenvironment regulation, which will improve biological understanding and design of novel therapeutics.
Collapse
Affiliation(s)
- Ning Liu
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
| | - Chunsheng Yang
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
| | - Ang Gao
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
| | - Meili Sun
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
- Correspondence: (M.S.); (D.L.)
| | - Deguan Lv
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA
- Correspondence: (M.S.); (D.L.)
| |
Collapse
|
24
|
Sun X, Kang Y, Li M, Li Y, Song J. The emerging regulatory mechanisms and biological function of circular RNAs in skeletal muscle development. BIOCHIMICA ET BIOPHYSICA ACTA (BBA) - GENE REGULATORY MECHANISMS 2022; 1865:194888. [DOI: 10.1016/j.bbagrm.2022.194888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/13/2022] [Accepted: 10/15/2022] [Indexed: 11/07/2022]
|
25
|
Smirnov A. How global RNA-binding proteins coordinate the behaviour of RNA regulons: an information approach. Comput Struct Biotechnol J 2022; 20:6317-6338. [DOI: 10.1016/j.csbj.2022.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022] Open
|
26
|
Seeler S, Moldovan LI, Bertelsen T, Hager H, Iversen L, Johansen C, Kjems J, Sommer Kristensen L. Global circRNA expression changes predate clinical and histological improvements of psoriasis patients upon secukinumab treatment. PLoS One 2022; 17:e0275219. [PMID: 36174034 PMCID: PMC9522259 DOI: 10.1371/journal.pone.0275219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/13/2022] [Indexed: 11/18/2022] Open
Abstract
Psoriasis is a common chronic inflammatory skin disease accompanied by heterogenous clinical and histological features, including a characteristic keratinocyte hyperproliferation and dermal immunogenic profile. In addition, psoriasis is associated with widespread transcriptomic alterations including changes in microRNA (miRNA) and circular RNA (circRNA) abundance, which constitute non-coding RNA (ncRNA) classes with specific regulatory capacities in diverse physiological and pathological processes. However, the knowledge about the expression dynamics of ncRNA during psoriasis treatment is sparse. To elucidate the dynamics of miRNA and circRNA abundance during secukinumab (anti-IL-17A) treatment, we studied their expression patterns in skin biopsies from 14 patients with severe plaque-type psoriasis before and during an 84-day secukinumab therapy at day 0, 4, 14, 42, and 84 using NanoString nCounter technology. We found a comprehensive downregulation of the majority of investigated circRNAs and specific alterations in the miRNA profile, including an upregulation of miR-203a-3p, miR-93-5p, and miR-378i in lesional compared to non-lesional skin before treatment. During treatment, the circRNAs progressively returned to the expression levels observed in non-lesional skin and already four days after treatment initiation most circRNAs were significantly upregulated. In comparison, for miRNAs, the normalization to baseline during treatment was delayed and limited to a subset of miRNAs. Moreover, we observed a strong correlation between multiple circRNAs, including ciRS-7 and circPTPRA, and the psoriasis area and severity index (PASI). Similar pronounced correlations could, however, not be found for miRNAs. Finally, we did not observe any significant changes in circRNA expression in peripheral blood mononuclear cells during treatment. In conclusion, we uncovered a rapid shift in global circRNA abundance upon anti-IL-17A treatment, which predated clinical and histological improvements, and a strong correlation with PASI, indicating a biomarker potential of individual circRNAs.
Collapse
Affiliation(s)
- Sabine Seeler
- Faculty of Health, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Liviu-Ionut Moldovan
- Faculty of Health, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Trine Bertelsen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Henrik Hager
- Department of Clinical Pathology, Vejle Hospital, Vejle, Denmark
| | - Lars Iversen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Claus Johansen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark
- Department of Molecular Biology and Genetics (MBG), Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
27
|
Xu X, Wang K, Vera O, Verma A, Jasani N, Bok I, Elemento O, Du D, Yu X, Karreth FA. Gain of Chromosome 1q Perturbs a Competitive Endogenous RNA Network to Promote Melanoma Metastasis. Cancer Res 2022; 82:3016-3031. [PMID: 36052492 PMCID: PMC9971359 DOI: 10.1158/0008-5472.can-22-0283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/19/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022]
Abstract
Somatic copy-number alterations (CNA) promote cancer, but the underlying driver genes may not be comprehensively identified if only the functions of the encoded proteins are considered. mRNAs can act as competitive endogenous RNAs (ceRNA), which sponge miRNAs to posttranscriptionally regulate gene expression in a protein coding-independent manner. We investigated the contribution of ceRNAs to the oncogenic effects of CNAs. Chromosome 1q gains promoted melanoma progression and metastasis at least in part through overexpression of three mRNAs with ceRNA activity: CEP170, NUCKS1, and ZC3H11A. These ceRNAs enhanced melanoma metastasis by sequestering tumor suppressor miRNAs. Orthogonal genetic assays with miRNA inhibitors and target site blockers, along with rescue experiments, demonstrated that miRNA sequestration is critical for the oncogenic effects of CEP170, NUCKS1, and ZC3H11A mRNAs. Furthermore, chromosome 1q ceRNA-mediated miRNA sequestration alleviated the repression of several prometastatic target genes. This regulatory RNA network was evident in other cancer types, suggesting chromosome 1q ceRNA deregulation as a common driver of cancer progression. Taken together, this work demonstrates that ceRNAs mediate the oncogenicity of somatic CNAs. SIGNIFICANCE The function of CEP170, NUCKS1, and ZC3H11A mRNAs as competitive endogenous RNAs that sequester tumor suppressor microRNAs underlies the oncogenic activity of chromosome 1q gains.
Collapse
Affiliation(s)
- Xiaonan Xu
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | - Kaizhen Wang
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
- Cancer Biology PhD program, University of South Florida, Tampa, FL 33612, USA
| | - Olga Vera
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | - Akanksha Verma
- Caryl and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Neel Jasani
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
- Cancer Biology PhD program, University of South Florida, Tampa, FL 33612, USA
| | - Ilah Bok
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
- Cancer Biology PhD program, University of South Florida, Tampa, FL 33612, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Dongliang Du
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | - Florian A. Karreth
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
- Correspondence: Florian A. Karreth, PhD, Moffitt Cancer Center, 12902 Magnolia Drive, Stabile Research Building, Rm 23043, Tampa, FL 33612, USA, , Phone: 813-745-1851
| |
Collapse
|
28
|
RNA-Binding Proteins in the Regulation of Adipogenesis and Adipose Function. Cells 2022; 11:cells11152357. [PMID: 35954201 PMCID: PMC9367552 DOI: 10.3390/cells11152357] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 01/27/2023] Open
Abstract
The obesity epidemic represents a critical public health issue worldwide, as it is a vital risk factor for many diseases, including type 2 diabetes (T2D) and cardiovascular disease. Obesity is a complex disease involving excessive fat accumulation. Proper adipose tissue accumulation and function are highly transcriptional and regulated by many genes. Recent studies have discovered that post-transcriptional regulation, mainly mediated by RNA-binding proteins (RBPs), also plays a crucial role. In the lifetime of RNA, it is bound by various RBPs that determine every step of RNA metabolism, from RNA processing to alternative splicing, nucleus export, rate of translation, and finally decay. In humans, it is predicted that RBPs account for more than 10% of proteins based on the presence of RNA-binding domains. However, only very few RBPs have been studied in adipose tissue. The primary aim of this paper is to provide an overview of RBPs in adipogenesis and adipose function. Specifically, the following best-characterized RBPs will be discussed, including HuR, PSPC1, Sam68, RBM4, Ybx1, Ybx2, IGF2BP2, and KSRP. Characterization of these proteins will increase our understanding of the regulatory mechanisms of RBPs in adipogenesis and provide clues for the etiology and pathology of adipose-tissue-related diseases.
Collapse
|
29
|
Górka S, Kubiak D, Ciesińska M, Niedojadło K, Tyburski J, Niedojadło J. Function of Cajal Bodies in Nuclear RNA Retention in A. thaliana Leaves Subjected to Hypoxia. Int J Mol Sci 2022; 23:ijms23147568. [PMID: 35886915 PMCID: PMC9321658 DOI: 10.3390/ijms23147568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 11/29/2022] Open
Abstract
Retention of RNA in the nucleus precisely regulates the time and rate of translation and controls transcriptional bursts that can generate profound variability in mRNA levels among identical cells in tissues. In this study, we investigated the function of Cajal bodies (CBs) in RNA retention in A. thaliana leaf nuclei during hypoxia stress was investigated. It was observed that in ncb-1 mutants with a complete absence of CBs, the accumulation of poly(A+) RNA in the leaf nuclei was lower than that in wt under stress. Moreover, unlike in root cells, CBs store less RNA, and RNA retention in the nuclei is much less intense. Our results reveal that the function of CBs in the accumulation of RNA in nuclei under stress depends on the plant organ. Additionally, in ncb-1, retention of introns of mRNA RPB1 (largest subunit of RNA polymerase II) mRNA was observed. However, this isoform is highly accumulated in the nucleus. It thus follows that intron retention in transcripts is more important than CBs for the accumulation of RNA in nuclei. Accumulated mRNAs with introns in the nucleus could escape transcript degradation by NMD (nonsense-mediated mRNA decay). From non-fully spliced mRNAs in ncb-1 nuclei, whose levels increase during hypoxia, introns are removed during reoxygenation. Then, the mRNA is transferred to the cytoplasm, and the RPB1 protein is translated. Despite the accumulation of isoforms in nuclei with retention of introns in reoxygenation, ncb-1 coped much worse with long hypoxia, and manifested faster yellowing and shrinkage of leaves.
Collapse
Affiliation(s)
- Sylwia Górka
- Department of Cellular and Molecular Biology, Nicolaus Copernicus University, Lwowska 1, 87-100 Toruń, Poland; (S.G.); (D.K.); (M.C.); (K.N.)
- Centre For Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Wileńska 4, 87-100 Toruń, Poland
| | - Dawid Kubiak
- Department of Cellular and Molecular Biology, Nicolaus Copernicus University, Lwowska 1, 87-100 Toruń, Poland; (S.G.); (D.K.); (M.C.); (K.N.)
- Centre For Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Wileńska 4, 87-100 Toruń, Poland
| | - Małgorzata Ciesińska
- Department of Cellular and Molecular Biology, Nicolaus Copernicus University, Lwowska 1, 87-100 Toruń, Poland; (S.G.); (D.K.); (M.C.); (K.N.)
| | - Katarzyna Niedojadło
- Department of Cellular and Molecular Biology, Nicolaus Copernicus University, Lwowska 1, 87-100 Toruń, Poland; (S.G.); (D.K.); (M.C.); (K.N.)
- Centre For Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Wileńska 4, 87-100 Toruń, Poland
| | - Jarosław Tyburski
- Chair of Plant Physiology and Biotechnology, Nicolaus Copernicus University, Lwowska 1, 87-100 Toruń, Poland;
| | - Janusz Niedojadło
- Department of Cellular and Molecular Biology, Nicolaus Copernicus University, Lwowska 1, 87-100 Toruń, Poland; (S.G.); (D.K.); (M.C.); (K.N.)
- Centre For Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Wileńska 4, 87-100 Toruń, Poland
- Correspondence:
| |
Collapse
|
30
|
Krueger A, Łyszkiewicz M, Heissmeyer V. Post-transcriptional control of T-cell development in the thymus. Immunol Lett 2022; 247:1-12. [DOI: 10.1016/j.imlet.2022.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/18/2022] [Accepted: 04/26/2022] [Indexed: 11/05/2022]
|
31
|
Balihodzic A, Prinz F, Dengler MA, Calin GA, Jost PJ, Pichler M. Non-coding RNAs and ferroptosis: potential implications for cancer therapy. Cell Death Differ 2022; 29:1094-1106. [PMID: 35422492 PMCID: PMC9177660 DOI: 10.1038/s41418-022-00998-x] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 12/15/2022] Open
Abstract
Ferroptosis is a recently defined form of regulated cell death, which is biochemically and morphologically distinct from traditional forms of programmed cell death such as apoptosis or necrosis. It is driven by iron, reactive oxygen species, and phospholipids that are oxidatively damaged, ultimately resulting in mitochondrial damage and breakdown of membrane integrity. Numerous cellular signaling pathways and molecules are involved in the regulation of ferroptosis, including enzymes that control the cellular redox status. Alterations in the ferroptosis-regulating network can contribute to the development of various diseases, including cancer. Evidence suggests that ferroptosis is commonly suppressed in cancer cells, allowing them to survive and progress. However, cancer cells which are resistant to common chemotherapeutic drugs seem to be highly susceptible to ferroptosis inducers, highlighting the great potential of pharmacologic modulation of ferroptosis for cancer treatment. Non-coding RNAs (ncRNAs) are considered master regulators of various cellular processes, particularly in cancer where they have been implicated in all hallmarks of cancer. Recent work also demonstrated their involvement in the molecular control of ferroptosis. Hence, ncRNA-based therapeutics represent an exciting alternative to modulate ferroptosis for cancer therapy. This review summarizes the ncRNAs implicated in the regulation of ferroptosis in cancer and highlights their underlying molecular mechanisms in the light of potential therapeutic applications.
Collapse
Affiliation(s)
- Amar Balihodzic
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036, Graz, Austria.,Research Unit "Non-Coding RNAs and Genome Editing in Cancer", Division of Oncology, Medical University of Graz, 8036, Graz, Austria
| | - Felix Prinz
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036, Graz, Austria.,Research Unit "Non-Coding RNAs and Genome Editing in Cancer", Division of Oncology, Medical University of Graz, 8036, Graz, Austria
| | - Michael A Dengler
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036, Graz, Austria
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Philipp J Jost
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036, Graz, Austria.,Medical Department III for Hematology and Oncology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Martin Pichler
- Department of Internal Medicine, Division of Oncology, Medical University of Graz, 8036, Graz, Austria. .,Research Unit "Non-Coding RNAs and Genome Editing in Cancer", Division of Oncology, Medical University of Graz, 8036, Graz, Austria. .,Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
32
|
Yuan C, Lu J, Chen Z, Zhou Q. Circ-GTF2I/miR-590-5p Axis Aggravates Myocardial Ischemia-Reperfusion Injury by Regulating Kelch Repeat and BTB Domain-Containing Protein 7. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2327669. [PMID: 35668777 PMCID: PMC9166968 DOI: 10.1155/2022/2327669] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/27/2022] [Accepted: 05/03/2022] [Indexed: 11/26/2022]
Abstract
Purpose We investigated the effect of the circular RNA (circRNA) general transcription factor IIi (GTF2I) on myocardial ischemia (MI) deterioration and neonatal rat cardiomyocyte damage. Methods The cell experiment was performed by using neonatal rat cardiomyocytes. Moreover, a hypoxia/reoxygenation treatment model was established. Cell Counting Kit-8 assay was conducted, and EdU cell proliferation was detected. Cell apoptosis was detected via flow cytometry and quantitative RT-PCR (RT-qPCR). Binding detection was performed through a double-luciferase reporter assay. Interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and lactate dehydrogenase (LDH) were detected via enzyme-linked immunosorbent assay (ELISA). Results Compared with that in the sham and control groups, circ-GTF2I expression in MIRI and the hypoxia/reoxygenation treatment model was significantly upregulated in vivo and in vitro. The knockdown of circ-GTF2I relieved neonatal rat cardiomyocyte damage and MI. Further detection through the double-luciferase reporter assay confirmed that the binding site of circ-GTF2I to miR-590-5p and miR-590-5p was Kelch repeat and BTB domain-containing protein 7 (KBTBD7). ELISA and RT-qPCR results showed that circ-GTF2I induced the abnormal expressions of IL-6 TNF-α, LDH, Bax, Bcl-2, and Cyt-c in MIRI and the hypoxia/reoxygenation treatment models by regulating miR-590-5p and the heart development transcription factor KBTBD7. Conclusions CircRNA circ-GTF2I aggravated MIRI and neonatal rat cardiomyocyte damage in vivo and in vitro by regulating miR-590-5p and the heart development transcription factor KBTBD7.
Collapse
Affiliation(s)
- Chunju Yuan
- Department of Cardiovascular Medicine, Zhongda Hospital Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Jing Lu
- Department of Cardiovascular Medicine, Zhongda Hospital Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Zhongpu Chen
- Department of Cardiovascular Medicine, Zhongda Hospital Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Qianxing Zhou
- Department of Cardiovascular Medicine, Zhongda Hospital Southeast University, Nanjing 210009, Jiangsu Province, China
| |
Collapse
|
33
|
Tong KL, Tan KE, Lim YY, Tien XY, Wong PF. CircRNA-miRNA interactions in atherogenesis. Mol Cell Biochem 2022; 477:2703-2733. [PMID: 35604519 DOI: 10.1007/s11010-022-04455-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 04/27/2022] [Indexed: 11/30/2022]
Abstract
Atherosclerosis is the major cause of coronary artery disease (CAD) which includes unstable angina, myocardial infarction, and heart failure. The onset of atherogenesis, a process of atherosclerotic lesion formation in the intima of arteries, is driven by lipid accumulation, a vicious cycle of reactive oxygen species (ROS)-induced oxidative stress and inflammatory reactions leading to endothelial cell (EC) dysfunction, vascular smooth muscle cell (VSMC) activation, and foam cell formation which further fuel plaque formation and destabilization. In recent years, there is a surge in the number of publications reporting the involvement of circular RNAs (circRNAs) in the pathogenesis of cardiovascular diseases, cancers, and metabolic syndromes. These studies have advanced our understanding on the biological functions of circRNAs. One of the most common mechanism of action of circRNAs reported is the sponging of microRNAs (miRNAs) by binding to the miRNAs response element (MRE), thereby indirectly increases the transcription of their target messenger RNAs (mRNAs). Individual networks of circRNA-miRNA-mRNA associated with atherogenesis have been extensively reported, however, there is a need to connect these findings for a complete overview. This review aims to provide an update on atherogenesis-related circRNAs and analyze the circRNA-miRNA-mRNA interactions in atherogenesis. The atherogenic mechanisms and clinical relevance of each atherogenesis-related circRNA were systematically discussed for better understanding of the knowledge gap in this area.
Collapse
Affiliation(s)
- Kind-Leng Tong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Ke-En Tan
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Yat-Yuen Lim
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Xin-Yi Tien
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Pooi-Fong Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
34
|
Gao F, Zhao Y, Zhang B, Xiao C, Sun Z, Gao Y, Dou X. Suppression of lncRNA Gm47283 attenuates myocardial infarction via miR-706/ Ptgs2/ferroptosis axis. Bioengineered 2022; 13:10786-10802. [PMID: 35485136 PMCID: PMC9208485 DOI: 10.1080/21655979.2022.2065743] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Myocardial infarction (MI) is the leading cause of sudden death. Long non-doing RNAs (lncRNAs) were demonstrated to play crucial roles in multiple diseases, including cancer and cardiovascular diseases. Nevertheless, the molecular mechanism of lncNRAs in MI is unclear. In this study, we integrated bioinformatics and molecular biological experiments to identify the novel lncRNA transcripts and elucidated its regulatory mechanism in MI. First, we identified 10 dysregualted lncRNAs and found that lncRNA Gm47283 was the top risk factor in MI. Bioinformatics analysis predicted that lncRNA Gm47283 exerted function via targeting miR-706 and Ptgs2. Ptgs2 was also the known regulator of ferroptosis. Inhibition or overexpression of lncRNA Gm47283 could regulate Ptgs2 expression and downstream ferroptosis activity. Overexpression of miR-706 could inhibit the expression of Ptgs2 and the activity of ferroptosis, thereby attenuated cellular injury. Mechanically, co-transfection experiments showed that overexpression of miR-706 could reverse the damage effect that was caused by lncRNA Gm47283 overexpression, via inhibiting Ptgs2 and ferroptosis. Additionally, inhibition of lncRNA Gm47283 by stem cell membrane coated siRNA could attenuate MI in vivo. Our study elucidated a novel mechanism containing lncRNA Gm47283/miR-706/Ptgs2/ferroptosis in MI, which provided a potential therapeutic for MI. Graphical Abstract. Stem cell membrane coated siRNA of lncRNA Gm47283 inhibits cardiomyocyte ferroptosis in myocardial infarction rat. Stem cell membrane-coated siRNA of lncRNA Gm47283 increases miR-706, and then miR-706 suppresses the expression of Ptgs2 to reduce lipid peroxidation toxicity, and then inhibits cardiomyocyte ferroptosis. PUFA: polyunsaturated fatty acid.
Collapse
Affiliation(s)
- Feng Gao
- Department Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou City, Jiangsu, China
| | - Yongcheng Zhao
- Department Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou City, Jiangsu, China
| | - Bin Zhang
- Department Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou City, Jiangsu, China
| | - Chunwei Xiao
- Department Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou City, Jiangsu, China
| | - Zhanfa Sun
- Department Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou City, Jiangsu, China
| | - Yuan Gao
- Department Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou City, Jiangsu, China
| | - Xueyong Dou
- Department Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou City, Jiangsu, China
| |
Collapse
|
35
|
Lin W, Wen X, Li X, Chen L, Wei W, Zhang L, Chen J. MiR-144 regulates adipogenesis by mediating formation of C/EBPα-FOXO1 protein complex. Biochem Biophys Res Commun 2022; 612:126-133. [PMID: 35525196 DOI: 10.1016/j.bbrc.2022.04.093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 11/17/2022]
Abstract
CeRNA effect was an important regulation mode of miRNA mediated bio-activities, however, most of the researches of ceRNA were on ncRNAs synergetic with mRNAs, the exploration of ceRNA effect regulated mRNA interaction was still lack of. Besides, C/EBPα was one of the most crucial adipogenic regulators, which has been demonstrated to form a protein complex with FOXO1 to mediate AdipoQ expression. So that, we try to explore whether the ceRNA effect mediated the interaction of C/EBPα and FOXO1, and identified the key miRNAs of their ceRNA effect. In this paper, we found the ceRNA effect of C/EBPα and FOXO1 mediated their protein complex formation, furthermore regulated its transcriptional role for AdipoQ, thereby influencing pre-adipocytes adipogenesis. More importantly, we demonstrated that the miR-144 was the decisive factor that mediated the ceRNA effect of C/EBPα and FOXO1 to influence AdipoQ, thus regulated pre-adipocytes adipogenesis. This research will provide a new supplementary idea of the miRNA role in mediating coding RNA interaction that regulates pre-adipocyte adipogenesis.
Collapse
Affiliation(s)
- Weimin Lin
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Xianyu Wen
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Xuexin Li
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Lei Chen
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Wei Wei
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Lifan Zhang
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Jie Chen
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China.
| |
Collapse
|
36
|
Arthurs AL, Jankovic-Karasoulos T, Smith MD, Roberts CT. Circular RNAs in Pregnancy and the Placenta. Int J Mol Sci 2022; 23:ijms23094551. [PMID: 35562943 PMCID: PMC9100345 DOI: 10.3390/ijms23094551] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 01/27/2023] Open
Abstract
The emerging field of circular RNAs (circRNAs) has identified their novel roles in the development and function of many cancers and inspired the interest of many researchers. circRNAs are also found throughout the healthy body, as well as in other pathological states, but while research into the function and abundance of circRNAs has progressed, our overall understanding of these molecules remains primitive. Importantly, recent studies are elucidating new roles for circRNAs in pregnancy, particularly in the placenta. Given that many of the genes responsible for circRNA production in cancer are also highly expressed in the placenta, it is likely that the same genes act in the production of circRNAs in the placenta. Furthermore, placental development can be referred to as ‘controlled cancer’, as it shares many key signalling pathways and hallmarks with tumour growth and metastasis. Hence, the roles of circRNAs in this field are important to study with respect to pregnancy success but also may provide novel insights for cancer progression. This review illuminates the known roles of circRNAs in pregnancy and the placenta, as well as demonstrating differential placental expressions of circRNAs between complicated and uncomplicated pregnancies.
Collapse
|
37
|
Trophectoderm Transcriptome Analysis in LIN28 Knockdown Ovine Conceptuses Suggests Diverse Roles of the LIN28-let-7 Axis in Placental and Fetal Development. Cells 2022; 11:cells11071234. [PMID: 35406798 PMCID: PMC8997724 DOI: 10.3390/cells11071234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/26/2022] [Accepted: 03/31/2022] [Indexed: 02/07/2023] Open
Abstract
The proper conceptus elongation in ruminants is critical for the successful placentation and establishment of pregnancy. We have previously shown that the trophectoderm-specific knockdown of LIN28A/B in day 9 ovine blastocysts resulted in increased let-7 miRNAs and reduced conceptus elongation at day 16 of gestation. In this current study, by transcriptome analysis of LIN28A knockdown (AKD) or LIN28B knockdown (BKD) trophectoderm (TE), we explored the downstream target genes of the LIN28-let-7 axis and their roles in the placental and fetal development. We identified 449 differentially expressed genes (DEGs) in AKD TE and 1214 DEGs in BKD TE compared to non-targeting control (NTC). Our analysis further revealed that 210 downregulated genes in AKD TE and 562 downregulated genes in BKD TE were the potential targets of let-7 miRNAs. Moreover, 16 downregulated genes in AKD TE and 57 downregulated and 7 upregulated genes in BKD TE were transcription factors. The DEGs in AKD and BKD TE showed enrichment in the biological processes and pathways critical for placental development and function, and fetal development and growth. The results of this study suggest the potential roles of the LIN28-let-7 axis in placental and fetal development beyond its involvement in trophoblast proliferation and conceptus elongation.
Collapse
|
38
|
Shi P, Li M, Song C, Qi H, Ba L, Cao Y, Zhang M, Xie Y, Ren J, Wu J, Ren P, Sun H. Neutrophil-like cell membrane-coated siRNA of lncRNA AABR07017145.1 therapy for cardiac hypertrophy via inhibiting ferroptosis of CMECs. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:16-36. [PMID: 34938604 PMCID: PMC8646082 DOI: 10.1016/j.omtn.2021.10.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/28/2021] [Accepted: 10/28/2021] [Indexed: 12/20/2022]
Abstract
Cardiac microvascular dysfunction is associated with cardiac hypertrophy and can eventually lead to heart failure. Dysregulation of long non-coding RNAs (lncRNAs) has recently been recognized as one of the key mechanisms involved in cardiac hypertrophy. However, the potential roles and underlying mechanisms of lncRNAs in cardiac microvascular dysfunction have not been explicitly delineated. Our results confirmed that cardiac microvascular dysfunction was related to cardiac hypertrophy and ferroptosis of cardiac microvascular endothelial cells (CMECs) occurred during cardiac hypertrophy. Using a combination of in vivo and in vitro studies, we identified a lncRNA AABR07017145.1, named as lncRNA AAB for short, and revealed that lncRNA AAB was upregulated in the hearts of cardiac hypertrophy rats as well as in the Ang II-induced CMECs. Importantly, we found that lncRNA AAB sponged and sequestered miR-30b-5p to induce the imbalance of MMP9/TIMP1, which enhanced the activation of transferrin receptor 1 (TFR-1) and then eventually led to the ferroptosis of CMECs. Moreover, we have developed a delivery system based on neutrophil membrane (NM)-camouflaged mesoporous silica nanocomplex (MSN) for inhibition of cardiac hypertrophy, indicating the potential role of silenced lncRNA AAB (si-AAB) and overexpressed miR-30b-5p as the novel therapy for cardiac hypertrophy.
Collapse
Affiliation(s)
- Pilong Shi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Minghui Li
- Department of Pharmacy, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Chao Song
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Hanping Qi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Lina Ba
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Yonggang Cao
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Meitian Zhang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Yawen Xie
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Jing Ren
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Jiabi Wu
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Ping Ren
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Hongli Sun
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| |
Collapse
|
39
|
Chirichella M, Bianchi N, Džafo E, Foli E, Gualdrini F, Kenyon A, Natoli G, Monticelli S. RFX transcription factors control a miR-150/PDAP1 axis that restrains the proliferation of human T cells. PLoS Biol 2022; 20:e3001538. [PMID: 35143476 PMCID: PMC8865640 DOI: 10.1371/journal.pbio.3001538] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 02/23/2022] [Accepted: 01/13/2022] [Indexed: 01/11/2023] Open
Abstract
Within the immune system, microRNAs (miRNAs) exert key regulatory functions. However, what are the mRNA targets regulated by miRNAs and how miRNAs are transcriptionally regulated themselves remain for the most part unknown. We found that in primary human memory T helper lymphocytes, miR-150 was the most abundantly expressed miRNA, and its expression decreased drastically upon activation, suggesting regulatory roles. Constitutive MIR150 gene expression required the RFX family of transcription factors, and its activation-induced down-regulation was linked to their reduced expression. By performing miRNA pull-down and sequencing experiments, we identified PDGFA-associated protein 1 (PDAP1) as one main target of miR-150 in human T lymphocytes. PDAP1 acted as an RNA-binding protein (RBP), and its CRISPR/Cas-9–mediated deletion revealed that it prominently contributed to the regulation of T-cell proliferation. Overall, using an integrated approach involving quantitative analysis, unbiased genomics, and genome editing, we identified RFX factors, miR-150, and the PDAP1 RBP as the components of a regulatory axis that restrains proliferation of primary human T lymphocytes. MicroRNAs exert key regulatory functions in the immune system, but their targets are largely unknown. This study shows that the ability of primary human T lymphocytes to proliferate in response to T cell receptor activation is modulated by a network comprising miR-150, transcription factors of the RFX family, and the RNA-binding protein PDAP1.
Collapse
Affiliation(s)
- Michele Chirichella
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana (USI), Bellinzona, Switzerland
| | - Niccolò Bianchi
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana (USI), Bellinzona, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Emina Džafo
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana (USI), Bellinzona, Switzerland
| | - Elena Foli
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana (USI), Bellinzona, Switzerland
| | - Francesco Gualdrini
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Milan, Italy
- Humanitas University, Milan, Italy
| | - Amy Kenyon
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Milan, Italy
| | - Gioacchino Natoli
- IEO, European Institute of Oncology IRCCS, Department of Experimental Oncology, Milan, Italy
- Humanitas University, Milan, Italy
| | - Silvia Monticelli
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana (USI), Bellinzona, Switzerland
| |
Collapse
|
40
|
Abstract
Most of the transcribed human genome codes for noncoding RNAs (ncRNAs), and long noncoding RNAs (lncRNAs) make for the lion's share of the human ncRNA space. Despite growing interest in lncRNAs, because there are so many of them, and because of their tissue specialization and, often, lower abundance, their catalog remains incomplete and there are multiple ongoing efforts to improve it. Consequently, the number of human lncRNA genes may be lower than 10,000 or higher than 200,000. A key open challenge for lncRNA research, now that so many lncRNA species have been identified, is the characterization of lncRNA function and the interpretation of the roles of genetic and epigenetic alterations at their loci. After all, the most important human genes to catalog and study are those that contribute to important cellular functions-that affect development or cell differentiation and whose dysregulation may play a role in the genesis and progression of human diseases. Multiple efforts have used screens based on RNA-mediated interference (RNAi), antisense oligonucleotide (ASO), and CRISPR screens to identify the consequences of lncRNA dysregulation and predict lncRNA function in select contexts, but these approaches have unresolved scalability and accuracy challenges. Instead-as was the case for better-studied ncRNAs in the past-researchers often focus on characterizing lncRNA interactions and investigating their effects on genes and pathways with known functions. Here, we focus most of our review on computational methods to identify lncRNA interactions and to predict the effects of their alterations and dysregulation on human disease pathways.
Collapse
|
41
|
Tang K, Wu Z, Sun M, Huang X, Sun J, Shi J, Wang X, Miao Z, Gao P, Song Y, Wang Z. Elevated MMP10/13 mediated barrier disruption and NF-κB activation aggravate colitis and colon tumorigenesis in both individual or full miR-148/152 family knockout mice. Cancer Lett 2022; 529:53-69. [PMID: 34979166 DOI: 10.1016/j.canlet.2021.12.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/07/2021] [Accepted: 12/28/2021] [Indexed: 12/13/2022]
Abstract
Dynamic miRNA alteration is known to occur in colitis-associated colon cancer (CAC), while the molecular mechanisms underpinning how miRNAs modulate the development from chronic inflammation to CAC is lacking. For the first time, we constructed knockout (KO) mice for individual miR-148/152 family members and entire miR-148/152 family. Based on these KO mice, we conduct the first comprehensive analysis of miR-148/152 family, demonstrating that deficiency of any member of miR-148/152 family aggravate colitis and CAC. Loss of individual miR-148/152 family members or full-family enhance MMP10 and MMP13 expression, causing disruption of intestinal barrier and cleaving pro-TNF-α into bioactive TNF-α fragments to activate NF-κB signaling, thereby aggravating colitis. Individual and full-family deletion also increase accumulation of IKKα and IKKβ, resulting in further hyperactivation of NF-κB signaling, exacerbating colitis and CAC. Moreover, blocking NF-κB signaling exerts a restorative effect on colitis and CAC models only in KO mice. Taken together, these findings demonstrate deleting the full miR-148/152 family or individual members exhibit similar effects in colitis and CAC. Mechanically, miR-148/152 family members deficiency in mice elevates MMP10 and MMP13 to accelerate colitis and CAC via disrupting intestinal barrier function and activating NF-κB signaling, suggesting a potential therapeutic strategy for colitis and CAC.
Collapse
Affiliation(s)
- Kaiwen Tang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Zhonghua Wu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Mingwei Sun
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Xuanzhang Huang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Jingxu Sun
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Jinxin Shi
- Department of Gastrointestinal Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Xin Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Zhifeng Miao
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Peng Gao
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China.
| | - Yongxi Song
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China.
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| |
Collapse
|
42
|
Bioinformatic Analysis Identified Potentially Prognostic Long Noncoding RNAs and MicroRNAs for Gastric Cancer. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6683136. [PMID: 34926687 PMCID: PMC8683174 DOI: 10.1155/2021/6683136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 07/24/2021] [Accepted: 10/26/2021] [Indexed: 11/17/2022]
Abstract
Gastric cancer (GC) is the fifth most common malignant tumor in the world. The present study was performed to discover the potential diagnostic and therapeutic long noncoding RNAs (lncRNAs) and microRNAs (miRNAs) of GC. Data used in this study to identify differentially expressed lncRNAs (DElncRNAs) and miRNAs (DEmiRNAs) were obtained from 187 GC tissues and 32 adjacent nontumor tissues. The total clinical data on GC included 187 cases. The above data were from the TCGA database. RStudio/Bioconductor software was used to conduct univariate analysis, the least absolute shrinkage and selection operator (LASSO) Cox, and multivariate Cox proportional risk regression for the DElncRNAs and DEmiRNAs. Clinical information was analyzed through univariate and multivariate Cox analysis. Results: five lncRNAs (AC007785.3, AC079385.3, LINC00392, LINC01729, and U95743.1) and two miRNAs (hsa-miR-3174, hsa-miR-605) were proven to be independent prognostic indicators of GC. Results of the Kaplan-Meier survival analysis showed that AC007785.3, AC079385.3, LINC01729, miR-3174, and miR-605 were significantly correlated with OS of GC. The target genes of AC079385.3, miR-3174, and miR-605 were obtained and clustered mainly on MAPK and cGMP-PKG signaling pathways. The clinical data showed that age and clinicopathologic stage were correlated with the prognosis of GC. Furthermore, AC007785.3 was associated with metastasis of GC, and miR-3174 was associated with the primary tumor condition of GC. We identified three lncRNAs (AC007785.3, AC079385.3, LINC01729), two miRNAs (miR-3174, miR-605), and clinical factors related to the pathogenesis and prognosis of GC. Our predicted results provide a possible entry point for the study of prognostic markers for GC.
Collapse
|
43
|
Yang C, Zhang Y, Yang B. MIAT, a potent CVD-promoting lncRNA. Cell Mol Life Sci 2021; 79:43. [PMID: 34921634 PMCID: PMC11072732 DOI: 10.1007/s00018-021-04046-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 10/30/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022]
Abstract
The initial identification of long non-coding RNA myocardial infarction associated transcript (MIAT) as a genetic risk factor of myocardial infarction has made this lncRNA (designated as lncR-MIAT here) a focus of intensive studies worldwide. Emerging evidence supports that lncR-MIAT is susceptible in its expression to multiple deleterious factors like angiotensin II, isoproterenol, hypoxia, and infection and is anomaly overexpressed in serum, plasma, blood cells and myocardial tissues under a variety of cardiovascular conditions including myocardial infarction, cardiac hypertrophy, diabetic cardiomyopathy, dilated cardiomyopathy, sepsis cardiomyopathy, atrial fibrillation and microvascular dysfunction. Experimental results consistently demonstrated that upregulation of lncR-MIAT plays active roles in the pathological processes of the cardiovascular system and knockdown of this lncRNA effectively ameliorates the adverse conditions. The available data revealed that lncR-MIAT acts through multiple mechanisms such as competitive endogenous RNA, natural antisense RNA and RNA/protein interactions. Moreover, the functional domains of lncR-MIAT accounting for certain specific cellular functions of the full-length transcript have been identified and characterized. These insights will not only tremendously advance our understanding of lncRNA biology and pathophysiology, but also offer good opportunities for more innovative and precise design of agents that have the potential to be developed into new drugs for better therapy of cardiovascular diseases (CVDs) in the future. Herein, we provide an overview of lncR-MIAT, focusing on its roles in cardiovascular diseases, underline the unique cellular/molecular mechanisms for its actions, and speculate the perspectives about the translational studies on the potential diagnostic and therapeutic applications of lncR-MIAT.
Collapse
Affiliation(s)
- Chao Yang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, People's Republic of China
- Department of Biochemistry, Qiqihar Medical University, Qiqihar, 161000, Heilongjiang, People's Republic of China
| | - Yong Zhang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, People's Republic of China
- Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin, 150086, People's Republic of China
| | - Baofeng Yang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, People's Republic of China.
| |
Collapse
|
44
|
McBride CD, Del Vecchio D. Predicting Composition of Genetic Circuits with Resource Competition: Demand and Sensitivity. ACS Synth Biol 2021; 10:3330-3342. [PMID: 34780149 DOI: 10.1021/acssynbio.1c00281] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The design of genetic circuits typically relies on characterization of constituent modules in isolation to predict the behavior of modules' composition. However, it has been shown that the behavior of a genetic module changes when other modules are in the cell due to competition for shared resources. In order to engineer multimodule circuits that behave as intended, it is thus necessary to predict changes in the behavior of a genetic module when other modules load cellular resources. Here, we introduce two characteristics of circuit modules: the demand for cellular resources and the sensitivity to resource loading. When both are known for every genetic module in a circuit library, they can be used to predict any module's behavior upon addition of any other module to the cell. We develop an experimental approach to measure both characteristics for any circuit module using a resource sensor module. Using the measured resource demand and sensitivity for each module in a library, the outputs of the modules can be accurately predicted when they are inserted in the cell in arbitrary combinations. These resource competition characteristics may be used to inform the design of genetic circuits that perform as predicted despite resource competition.
Collapse
Affiliation(s)
- Cameron D. McBride
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02142, United States
| | - Domitilla Del Vecchio
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
45
|
Walgrave H, Zhou L, De Strooper B, Salta E. The promise of microRNA-based therapies in Alzheimer's disease: challenges and perspectives. Mol Neurodegener 2021; 16:76. [PMID: 34742333 PMCID: PMC8572071 DOI: 10.1186/s13024-021-00496-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 10/17/2021] [Indexed: 02/06/2023] Open
Abstract
Multi-pathway approaches for the treatment of complex polygenic disorders are emerging as alternatives to classical monotarget therapies and microRNAs are of particular interest in that regard. MicroRNA research has come a long way from their initial discovery to the cumulative appreciation of their regulatory potential in healthy and diseased brain. However, systematic interrogation of putative therapeutic or toxic effects of microRNAs in (models of) Alzheimer's disease is currently missing and fundamental research findings are yet to be translated into clinical applications. Here, we review the literature to summarize the knowledge on microRNA regulation in Alzheimer's pathophysiology and to critically discuss whether and to what extent these increasing insights can be exploited for the development of microRNA-based therapeutics in the clinic.
Collapse
Affiliation(s)
- Hannah Walgrave
- VIB Center for Brain & Disease Research, Leuven, KU, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Lujia Zhou
- Division of Janssen Pharmaceutica NV, Discovery Neuroscience, Janssen Research and Development, Beerse, Belgium
| | - Bart De Strooper
- VIB Center for Brain & Disease Research, Leuven, KU, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
- UK Dementia Research Institute at University College London, London, UK
| | - Evgenia Salta
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
46
|
Dragomir MP, Knutsen E, Calin GA. Classical and noncanonical functions of miRNAs in cancers. Trends Genet 2021; 38:379-394. [PMID: 34728089 DOI: 10.1016/j.tig.2021.10.002] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 12/11/2022]
Abstract
Alterations in microRNAs (miRNAs) expression are causative in the initiation and progression of human cancers. The molecular events responsible for the widespread differential expression of miRNAs in malignancy are exemplified by their location in cancer-associated genomic regions, epigenetic mechanisms, transcriptional dysregulation, chemical modifications and editing, and alterations in miRNA biogenesis proteins. The classical miRNA function is synonymous with post-transcriptional repression of target protein genes. However, several studies have reported miRNAs functioning outside this paradigm and some of these novel modes of regulation of gene expression have been implicated in cancers. Here, we summarize key aspects of miRNA involvement in cancer, with a special focus on these lesser-studied mechanisms of action.
Collapse
Affiliation(s)
- Mihnea P Dragomir
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.
| | - Erik Knutsen
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway.
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
47
|
Xie X, Xiao Y, Xu K. Mechanism underlying circularRNA_014301-mediated regulation of neuronal cell inflammation and apoptosis. Exp Ther Med 2021; 22:1432. [PMID: 34707713 PMCID: PMC8543437 DOI: 10.3892/etm.2021.10867] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 08/25/2021] [Indexed: 11/05/2022] Open
Abstract
Spinal cord injury (SCI) causes damage to the spinal cord owing to trauma or disease and myelinated fiber tracts that transmit sensation and motor signals to and from the brain. Circular RNAs (circRNAs) are a recently discovered class of regulatory molecules, and their roles in SCI are still unknown. circRNA_014301 was indicated to be differentially expressed in the spinal cord at the site of SCI in a rat model. To analyze the role of circRNA_014301 in SCI, we exposed rat adrenal pheochromocytoma PC12 cells were exposed to increasing concentrations of lipopolysaccharide (LPS) and to construct a PC12 cell inflammatory model. Cell Counting Kit-8 assay was used to analyze cell viability. Reverse transcription-quantitative PCR and ELISA were used to detect the expression of inflammatory factors (IL-1β, IL-6 and TNF-α). Annexin V-FITC/PI double staining was employed to detect cell apoptosis, and western blotting was performed to detect the expression of apoptotic proteins (Bax/Bcl-2/cleaved caspase-3) and NF-κB. The results demonstrated that LPS induced inflammation in PC12 cells as evidenced by the reduced cell proliferation and enhanced expression of inflammatory and apoptotic factors under increasing LPS concentrations. Western blotting analyses indicated that circRNA_014301 induced the expression of p-NF-κB/NF-κB, Bax and cleaved caspase-3, and decreased the expression of Bcl-2 following LPS-induced inflammation, and this apoptosis-promoting effect was relieved by small interfering-RNA-mediated knockdown of circRNA_014301. Thus, circRNA_014301 silencing alleviated apoptosis and inflammation in PC12 cells. SCI is invariably associated with spinal cord inflammation, and LPS was used to stimulate apoptosis and inflammatory injury in PC12 cells, and create a cell model of SCI. By promoting PC12 cell apoptosis under inflammatory conditions, it was indicated that circRNA_014301 may suppress SCI. Therefore, circRNA_014301 may represent a potential target for SCI diagnosis and therapy.
Collapse
Affiliation(s)
- Xiankuan Xie
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310053, P.R. China
| | - Yuxiang Xiao
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310053, P.R. China
| | - Kan Xu
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
48
|
Singh R, Zogg H, Ro S. Role of microRNAs in Disorders of Gut-Brain Interactions: Clinical Insights and Therapeutic Alternatives. J Pers Med 2021; 11:jpm11101021. [PMID: 34683162 PMCID: PMC8541612 DOI: 10.3390/jpm11101021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Disorders of gut–brain interactions (DGBIs) are heterogeneous in nature and intertwine with diverse pathophysiological mechanisms. Regular functioning of the gut requires complex coordinated interplay between a variety of gastrointestinal (GI) cell types and their functions are regulated by multiple mechanisms at the transcriptional, post-transcriptional, translational, and post-translational levels. MicroRNAs (miRNAs) are small non-coding RNA molecules that post-transcriptionally regulate gene expression by binding to specific mRNA targets to repress their translation and/or promote the target mRNA degradation. Dysregulation of miRNAs might impair gut physiological functions leading to DGBIs and gut motility disorders. Studies have shown miRNAs regulate gut functions such as visceral sensation, gut immune response, GI barrier function, enteric neuronal development, and GI motility. These biological processes are highly relevant to the gut where neuroimmune interactions are key contributors in controlling gut homeostasis and functional defects lead to DGBIs. Although extensive research has explored the pathophysiology of DGBIs, further research is warranted to bolster the molecular mechanisms behind these disorders. The therapeutic targeting of miRNAs represents an attractive approach for the treatment of DGBIs because they offer new insights into disease mechanisms and have great potential to be used in the clinic as diagnostic markers and therapeutic targets. Here, we review recent advances regarding the regulation of miRNAs in GI pacemaking cells, immune cells, and enteric neurons modulating pathophysiological mechanisms of DGBIs. This review aims to assess the impacts of miRNAs on the pathophysiological mechanisms of DGBIs, including GI dysmotility, impaired intestinal barrier function, gut immune dysfunction, and visceral hypersensitivity. We also summarize the therapeutic alternatives for gut microbial dysbiosis in DGBIs, highlighting the clinical insights and areas for further exploration. We further discuss the challenges in miRNA therapeutics and promising emerging approaches.
Collapse
Affiliation(s)
| | | | - Seungil Ro
- Correspondence: ; Tel.: +1-775-784-1462; Fax: +1-775-784-6903
| |
Collapse
|
49
|
Novel lncRNA Panel as for Prognosis in Esophageal Squamous Cell Carcinoma Based on ceRNA Network Mechanism. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:8020879. [PMID: 34603485 PMCID: PMC8486540 DOI: 10.1155/2021/8020879] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/20/2021] [Indexed: 12/14/2022]
Abstract
Background The competitive endogenous RNA (ceRNA) mechanism has been discovered recently and regulating cancer-related gene expressions. The ceRNA network participates in multiple processes, such as cell proliferation and metastasis, and potentially drives the progression of cancer. In this study, we focus on the ceRNA networks of esophageal squamous cell carcinoma and discovered a novel biomarker panel for cancer prognosis. Methods RNA expression data of esophageal carcinoma from the TCGA database were achieved and constructed ceRNA network in esophageal carcinoma using R packages. Results Four miRNAs were discovered as the core of the ceRNA model, including miR-93, miR-191, miR-99b, and miR-3615. Moreover, we constructed a ceRNA network in esophageal carcinoma, which included 4 miRNAs and 6 lncRNAs. After ceRNA network modeling, we investigated six lncRNAs which could be taken together as a panel for prognosis prediction of esophageal cancer, including LINC02575, LINC01087, LINC01816, AL136162.1, AC012073.1, and AC117402.1. Finally, we tested the predictive power of the panel in all TCGA samples. Conclusions Our study discovered a new biomarker panel which may have potential values in the prediction of prognosis of esophageal carcinoma.
Collapse
|
50
|
He L, Man C, Xiang S, Yao L, Wang X, Fan Y. Circular RNAs' cap-independent translation protein and its roles in carcinomas. Mol Cancer 2021; 20:119. [PMID: 34526007 PMCID: PMC8442428 DOI: 10.1186/s12943-021-01417-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/31/2021] [Indexed: 02/07/2023] Open
Abstract
Circular RNAs a kind of covalently closed RNA and widely expressed in eukaryotes. CircRNAs are involved in a variety of physiological and pathological processes, but their regulatory mechanisms are not fully understood. Given the development of the RNA deep-sequencing technology and the improvement of algorithms, some CircRNAs are discovered to encode proteins through the cap-independent mechanism and participate in the important process of tumorigenesis and development. Based on an overview of CircRNAs, this paper summarizes its translation mechanism and research methods, and reviews the research progress of CircRNAs translation in the field of oncology in recent years. Moreover, this paper aims to provide new ideas for tumor diagnosis and treatment through CircRNAs translation.
Collapse
Affiliation(s)
- Lian He
- Cancer Institue, Affiliated People's Hospital of Jiangsu University, No 8, Dianli Road, Zhenjiang, Jiangsu Province, 212002, People's Republic of China
| | - Changfeng Man
- Cancer Institue, Affiliated People's Hospital of Jiangsu University, No 8, Dianli Road, Zhenjiang, Jiangsu Province, 212002, People's Republic of China
| | - Shouyan Xiang
- Cancer Institue, Affiliated People's Hospital of Jiangsu University, No 8, Dianli Road, Zhenjiang, Jiangsu Province, 212002, People's Republic of China
| | - Lin Yao
- Cancer Institue, Affiliated People's Hospital of Jiangsu University, No 8, Dianli Road, Zhenjiang, Jiangsu Province, 212002, People's Republic of China
| | - Xiaoyan Wang
- Department of Gastroenterology, Affiliated Suqian First People's Hospital of Nanjing Medical University, No 120, Suzhi Road, Suqian, Jiangsu Province, 223812, People's Republic of China.
| | - Yu Fan
- Cancer Institue, Affiliated People's Hospital of Jiangsu University, No 8, Dianli Road, Zhenjiang, Jiangsu Province, 212002, People's Republic of China.
| |
Collapse
|