1
|
Lu J, Chen Y, Liu X, Wang J, He Y, Shi T, Chen W, Yan W. Artificial intelligence-driven microRNA signature for early detection of gastric cancer: discovery and clinical functional exploration. Br J Cancer 2025; 132:957-972. [PMID: 40234666 DOI: 10.1038/s41416-025-02984-9] [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: 09/24/2024] [Revised: 03/01/2025] [Accepted: 03/12/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a leading cause of cancer-related deaths worldwide, with late-stage diagnoses frequently leading to poor outcomes. This underscores the need for effective early-stage gastric cancer (ESGC) diagnostics. METHODS We introduce ESGCmiRD, an innovative artificial intelligence-driven strategy that identifies a miRNA signature for ESGC detection by integrating robust expression patterns, ESGC relevance, and regulatory capabilities of microRNA (miRNA) based on multiple networks. Expression and biological roles of miRNAs in GC were validated and explored via bioinformatics analysis and in vitro studies. miRNA-target interaction was confirmed by dual-luciferase reporter assay. Molecular docking predicted miRNA-drug binding affinities, assessing the miRNA signature's therapeutic potential. RESULTS ESGCmiRD identified a blood miRNA signature (miR-320b, miR-222-3p, miR-181a-5p, miR-103a-3p, miR-107) for ESGC detection, demonstrated high diagnostic accuracy with AUC values of 0.986, 0.977, 0.815, and 0.811 in the test and three validation sets (GSE211692, TCGA-STAD, and our cohort), respectively. The five miRNAs were overexpressed in ESGC plasma and directly target PTEN, promoting GC cell proliferation, migration, and invasion. Molecular docking suggested Paclitaxel had the strongest potential interaction with these miRNAs. CONCLUSION This method identifies a robust miRNA signature for ESGC detection and sheds light on gastric carcinogenesis mechanisms, opening doors for potential therapeutic strategies.
Collapse
Affiliation(s)
- Jiachun Lu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuqi Chen
- Department of Gastroenterology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin Liu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiayu Wang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuxin He
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.
| | - Wenying Yan
- School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China.
- Suzhou Key Lab of Multi-modal Data Fusion and Intelligent Healthcare, Suzhou, China.
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Suzhou, China.
| |
Collapse
|
2
|
Liu FF, Li K. The Abnormal ERα-miRNA Cross-Talk in AD-Affected Human Hippocampus: A Bioinformatics Perspective. Mol Neurobiol 2025; 62:7998-8012. [PMID: 39966328 PMCID: PMC12078360 DOI: 10.1007/s12035-025-04771-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
Estrogen's impact on Alzheimer's disease (AD) is multifaceted, with its receptors potentially influencing AD pathology in both beneficial and detrimental ways. This study aims to dissect the intricate cross-talk between estrogen receptor alpha (ERα) and microRNAs (miRNAs) in AD-affected human hippocampus. Through a comprehensive literature review in the PubMed database, coupled with a GeneCards database search, we obtained AD-related key miRNAs and genes in the hippocampus. Using bioinformatics tools, we predicted target genes and miRNAs of ERα, and the targets of the identified miRNAs. The integration of these elements resulted in the construction of an ERα-related FFL network, which includes 13 miRNAs and 56 core genes. Gene ontology (GO) and pathway enrichment analyses were conducted, revealing significant enrichment in biological processes such as neuron death and response to metal ions, and cellular components like membrane microdomains. Notably, the AKT-associated signaling pathway was prominently highlighted, with key genes including GSK3A, CDKN1A, AKT2, and MDM2, and key miRNAs including miR-485 and let-7f, suggesting a potential role of ERα in modulating this pathway in AD. The findings of this study provide a novel perspective on the regulatory network of ERα in the hippocampal region of AD and may pave the way for future research into the therapeutic potential of targeting the ERα pathway in neurodegenerative diseases.
Collapse
Affiliation(s)
- Fang-Fang Liu
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, No. 26 Shengli Street, Hankou District, Wuhan, 430014, People's Republic of China
| | - Ke Li
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Hankou District, Wuhan, 430030, Hubei, People's Republic of China.
| |
Collapse
|
3
|
Perez-Moreno E, Ortega-Hernández V, Zavala VA, Gamboa J, Fernández W, Carvallo P. Suppression of breast cancer metastatic behavior by microRNAs targeting EMT transcription factors. A relevant participation of miR-196a-5p and miR-22-3p in ZEB1 expression. Breast Cancer Res Treat 2025:10.1007/s10549-025-07723-5. [PMID: 40382762 DOI: 10.1007/s10549-025-07723-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 05/06/2025] [Indexed: 05/20/2025]
Abstract
PURPOSE Metastasis, the leading cause of cancer-associated deaths, is promoted by transcription factors SNAIL, SLUG, ZEB1 and TWIST through the activation of epithelial-mesenchymal transition (EMT). MicroRNAs can suppress EMT, emerging as candidate molecular biomarkers and novel therapeutic targets. Herein, we evaluated microRNAs downregulated in breast cancer (BC) tissues expressing EMT transcription factors, to find new potential regulators of EMT. METHODS Candidate microRNAs were selected from microarray data by their inversely correlated expression with SNAIL, SLUG, ZEB1 and TWIST, evaluated in BC tissues through immunohistochemistry. We selected eight microRNAs predicted in silico as probable modulators of SNAIL, SLUG, ZEB1 and TWIST, and validate their interaction through the 3'UTR region in luciferase reporter gene assays. MDA-MB-231 cells were transfected with selected microRNAs to perform migration, invasion and cell proliferation assays, and western blot was used to evaluate protein levels. RESULTS MiR-30a-5p, miR-1271-5p, miR-196a-5p, miR-202-3p, miR-210-3p, miR-22-3p and miR-331-3p decreased luciferase activity through SNAIL, SLUG, ZEB1 and/or TWIST 3'UTR. These microRNAs, including miR-34b-3p, decreased migration, invasion and cell proliferation in MDA-MB-231 cells. MiR-30a-5p, miR-202-3p and miR-22-3p decreased vimentin expression, whereas miR-196a-5p and miR-22-3p decreased endogenous ZEB1 levels. MiR-196a-5p, miR-202-3p and miR-30a-5p also decreased CCR7 expression, a chemokine receptor involved in lymph node metastasis. CONCLUSION microRNAs selected in this work can regulate gene expression trough 3'UTR region of EMT-transcription factors. In BC cells, miR-196a-5p and miR-22-3p decrease ZEB1 levels, being novel modulators of EMT. Also, the eight evaluated microRNAs, reduced the metastatic hallmarks in BC cells.
Collapse
Affiliation(s)
- Elisa Perez-Moreno
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Victoria Ortega-Hernández
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Valentina A Zavala
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge Gamboa
- Unidad de Patología Mamaria, Hospital Clínico San Borja Arriarán, Santiago, Chile
| | - Wanda Fernández
- Unidad de Anatomía Patológica, Hospital Clínico San Borja Arriarán, Santiago, Chile
| | - Pilar Carvallo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
4
|
Fan T, Su Z, Wang X, Wei T, Zhao L, Liu S. TarP: A microRNA target gene prediction tool utilizing a polymorphic structured alignment approach. Int J Biol Macromol 2025; 314:144320. [PMID: 40383335 DOI: 10.1016/j.ijbiomac.2025.144320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 05/08/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
MicroRNAs (miRNAs) represent a vital class of small non-coding RNAs that play key regulatory roles in gene expression. Accurate identification of miRNA-mRNA interactions is essential for understanding their biological functions. However, current computational prediction tools suffer from several limitations, including species-specific biases, suboptimal accuracy, high false discovery rates, and incomplete target gene coverage. To address these challenges, we present TarP, a novel miRNA target prediction algorithm employing a Polymorphic structured alignment (PMS) approach. Our method mimics the natural binding process between miRNAs and their target mRNAs by integrating key biological interaction features. The algorithm utilizes five distinct nucleotide-binding motifs to perform a structured decomposition and alignment of potential mRNA targets. Predictions are then rigorously evaluated through a dual scoring system: a Structure (St) coefficient assessing binding conformation and an Energy (En) coefficient evaluating thermodynamic stability, ensuring high-confidence target selection. Using experimentally validated human miRNA-mRNA interaction datasets, we benchmarked TarP against four widely used prediction tools (miRanda, RNAhybrid, PITA, and TargetScan). Comparative analyses demonstrate that TarP achieves superior performance in both sensitivity and specificity, exhibiting enhanced accuracy in positive target identification and improved discrimination between true and false interactions. The TarP algorithm is freely available at: https://github.com/Whimonk/TarP.
Collapse
Affiliation(s)
- Ting Fan
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, PR China
| | - Zhuanzhuan Su
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, PR China
| | - Xin Wang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, PR China
| | - Tianqi Wei
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, PR China
| | - Lu Zhao
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, PR China
| | - Shiping Liu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, PR China.
| |
Collapse
|
5
|
Chen D, Zhang QT, Li WJ, Han HL, Smagghe G, Yan Y, Jiang HB, Wang JJ, Wei D. The competing endogenous RNA lnc94641-miR957-3p mediates male fertility in Zeugodacus cucurbitae Coquillett. PEST MANAGEMENT SCIENCE 2025. [PMID: 40371678 DOI: 10.1002/ps.8874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/03/2025] [Accepted: 04/20/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Insect spermatogenesis is a complex process. Numerous genes are involved in sperm motility, which is crucial for male fertility. Few long non-coding RNAs (lncRNAs) in the testis regulate insect spermatogenesis. We previously identified 364 testis-enriched lncRNAs in the globally invasive pest Zeugodacus cucurbitae Coquillett. One of these lncRNAs, lnc94641, is abundantly expressed in the testis; however, its role in spermatogenesis remains unknown. RESULTS Suppression of lnc94641 expression led to a 60% decrease in spermatozoa count and a 29% decrease in offspring hatchability. A microRNA (miRNA), miR-957-3p, was experimentally demonstrated to bind to lnc94641 competitively. miR-957-3p overexpression recapitulated reproductive defect phenotypes similar to those caused by lnc94641 knockdown. Furthermore, target gene predictions combined with quantitative reverse transcription PCR, RNA pull-down, and dual luciferase reporter assays confirmed that miR-957-3p targets voltage-gated potassium channel 5 (VGKC5) and odorant receptor 85c (OR85c), elucidating a functional lncRNA-miRNA-mRNA competing endogenous RNA (ceRNA) regulatory axis. Fluorescence in situ hybridization (FISH) assays demonstrated the co-localization of lnc94641, miR-957-3p, and VGKC5/OR85c in the mature and transformed regions of the testes. Suppression of VGKC5/OR85c expression resulted in a 68% and 50% decrease in spermatozoa number and an 18% and 21% decrease in offspring hatchability, mirroring the phenotype observed with lnc94641-silencing, thereby reinforcing the mechanistic coherence of this regulatory network. CONCLUSION These results revealed a ceRNA axis mediated by 'lnc94641-miR957-3p-VGKC5/OR85c' involved in spermatogenesis that impairs male fertility in the melon fly. Molecular perturbations (lncRNA knockdown or miRNA overexpression) consistently impair sperm production and offspring viability by dysregulating ion channels and chemosensory genes. This mechanistically resolved pathway, centered on the core components VGKC5 and OR85c, revealed conserved reproductive vulnerabilities that could enable the targeted genetic control of this agricultural pest. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Dong Chen
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| | - Qi-Tong Zhang
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| | - Wei-Jun Li
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- School of Agricultural Sciences, Jiangxi Agricultural University, Nanchang, China
| | - Hong-Liang Han
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| | - Guy Smagghe
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Ying Yan
- Department of Insect Biotechnology in Plant Protection, Institute for Insect Biotechnology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Hong-Bo Jiang
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| | - Jin-Jun Wang
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| | - Dong Wei
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| |
Collapse
|
6
|
Casuso A, Valenzuela-Muñoz V, Sáez-Vera C, Gallardo-Escárate C. Environmental Changes Drives the Transcriptome and Gene Regulation Plasticity During Sea Lice Infestation. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2025; 27:80. [PMID: 40314793 DOI: 10.1007/s10126-025-10459-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 04/15/2025] [Indexed: 05/03/2025]
Abstract
The sea louse, Caligus rogercresseyi, is one of the main concerns in the Chilean salmon industry. The free-living copepodid stage can recognize the host and initiate the parasitic phase, where environmental factors can shape the host recognition process. This study aimed to explore the ecological influence on the transcriptome of copepodids infesting Atlantic salmon experimentally exposed to different salinity and temperature (S/T) conditions. Herein, 200 salmon were infested with 35 copepodids per fish previously acclimatized to four S/T treatments: 32 and 26 PSU; 8 and 16°C. After 48 h of infestation, the attached copepodids from each experimental group were counted and digitalized for geometric morphometric analysis. Copepodids were then collected for RNA sequencing to analyze transcriptome modulation and gene regulation. Morphological changes in copepodids were mainly associated with temperature rather than salinity conditions. The transcriptome survey revealed molecular signatures related to salinity and temperature changes, where salinity drives the gene expression of copepodids. Notably, specific genes, such as those encoding cuticle proteins and trypsin-like kinases, were regulated by all three post-transcriptional mechanisms assessed: alternative splicing, miRNA, and gene fusion. The transcriptome analysis revealed that trypsin-like kinase genes exhibited upregulation and downregulation across the various S/T conditions. In contrast, cuticle protein genes were consistently downregulated in the 32 PSU/8°C, 26 PSU/8°C, and 26 PSU/16°C groups compared to the 32 PSU/16°C control. This suggests that the three post-transcriptional mechanisms may exert a combined influence on the expression of specific genes, potentially driven by salinity and temperature environmental conditions in sea lice biology.
Collapse
Affiliation(s)
- Antonio Casuso
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, 4030000, Concepción, Chile
| | - Valentina Valenzuela-Muñoz
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, 4030000, Concepción, Chile
| | - Constanza Sáez-Vera
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, 4030000, Concepción, Chile
| | - Cristian Gallardo-Escárate
- Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, 4030000, Concepción, Chile.
| |
Collapse
|
7
|
Verkuijl SAN, Del Corsano G, Capriotti P, Yen PS, Inghilterra MG, Selvaraj P, Hoermann A, Martinez-Sanchez A, Ukegbu CV, Kebede TM, Vlachou D, Christophides GK, Windbichler N. A suppression-modification gene drive for malaria control targeting the ultra-conserved RNA gene mir-184. Nat Commun 2025; 16:3923. [PMID: 40280899 PMCID: PMC12032250 DOI: 10.1038/s41467-025-58954-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
Gene drive technology presents a promising approach to controlling malaria vector populations. Suppression drives are intended to disrupt essential mosquito genes whereas modification drives aim to reduce the individual vectorial capacity of mosquitoes. Here we present a highly efficient homing gene drive in the African malaria vector Anopheles gambiae that targets the microRNA gene mir-184 and combines suppression with modification. Homozygous gene drive (miR-184D) individuals incur significant fitness costs, including high mortality following a blood meal, that curtail their propensity for malaria transmission. We attribute this to a role of miR-184 in regulating solute transport in the mosquito gut. However, females remain fully fertile, and pure-breeding miR-184D populations suitable for large-scale releases can be reared under laboratory conditions. Cage invasion experiments show that miR-184D can spread to fixation thereby reducing population fitness, while being able to propagate a separate antimalarial effector gene at the same time. Modelling indicates that the miR-184D drive integrates aspects of population suppression and population replacement strategies into a candidate strain that should be evaluated further as a tool for malaria eradication.
Collapse
Affiliation(s)
- Sebald A N Verkuijl
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, UK
| | - Giuseppe Del Corsano
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, UK
| | - Paolo Capriotti
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, UK
| | - Pei-Shi Yen
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, UK
| | | | - Prashanth Selvaraj
- Institute for Disease Modeling, Bill and Melinda Gates Foundation, Seattle, WA, USA
| | - Astrid Hoermann
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, UK
| | - Aida Martinez-Sanchez
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Chiamaka Valerie Ukegbu
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, UK
| | - Temesgen M Kebede
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, UK
| | - Dina Vlachou
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, UK
| | - George K Christophides
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, UK
| | - Nikolai Windbichler
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London, UK.
| |
Collapse
|
8
|
Kosek DM, Petzold K, Andersson ER. Mapping effective microRNA pairing beyond the seed using abasic modifications. Nucleic Acids Res 2025; 53:gkaf364. [PMID: 40298108 PMCID: PMC12038393 DOI: 10.1093/nar/gkaf364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 03/31/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that regulate gene expression by base-pairing to complementary sites in messenger RNAs (mRNAs). The primary element for site recognition is the seed region (nucleotides 2-8 in the miRNA), but for a minority of sites pairing outside the seed increases efficiency, with the supplementary region (nucleotides 13-16) typically having the greatest impact. However, the structural determinants of effective pairing outside the seed are not fully understood. Here, we use abasic modified nucleotides to disrupt pairing to residues 13 and 14 of miR-34a and measure the effect of this modification compared to wild-type miR-34a on the cellular transcriptome and proteome using RNA-seq and mass spectrometry. We find that a subset of sites with predicted supplementary pairing are affected by miRNA transfection, with up to two-fold decreases in site repression at the mRNA level. We show that miR-34a 3'-pairing is sensitive to GU wobble pairs in a position-specific manner and favors bulges in the miRNA over the target. These results were validated with luciferase reporter assays. Overall, this study demonstrates a novel methodological approach for elucidating the role of specific miRNA residues in target site selection, advancing our understanding of miRNA-mediated gene regulation.
Collapse
Affiliation(s)
- David M Kosek
- Department of Cell and Molecular Biology, Karolinska Institute, Biomedicum 9B, Solnavägen 9, 171 77Stockholm, Sweden
- Department of Medical Biochemistry and Microbiology, Uppsala University, Biomedical Centre D9:3, Husargatan 3, 752 37 Uppsala, Sweden
| | - Katja Petzold
- Department of Medical Biochemistry and Microbiology, Uppsala University, Biomedical Centre D9:3, Husargatan 3, 752 37 Uppsala, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Biomedicum 9B, Solnavägen 9, 171 77 Stockholm, Sweden
- Science for Life Laboratory, Uppsala Biomedical Centre, Uppsala University, 75237 Uppsala, Sweden
- Center of Excellence for the Chemical Mechanisms of Life, Uppsala University, 75237 Uppsala, Sweden
| | - Emma R Andersson
- Department of Cell and Molecular Biology, Karolinska Institute, Biomedicum 9B, Solnavägen 9, 171 77Stockholm, Sweden
| |
Collapse
|
9
|
Roy A, Chakraborty S. Selective inhibition of rabies virus gene expression by human miRNAs: a therapeutic approach using the 7mer-m8 model. Virus Genes 2025:10.1007/s11262-025-02155-1. [PMID: 40234303 DOI: 10.1007/s11262-025-02155-1] [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: 12/08/2024] [Accepted: 04/03/2025] [Indexed: 04/17/2025]
Abstract
MicroRNAs, abbreviated as miRNAs, have a substantial impact on viral infections through their ability to control gene expression and influence the interactions between the host and the virus. This work investigates the capacity of miRNAs to selectively inhibit the expression of rabies virus genes, specifically Nucleoprotein N, Phosphoprotein M1 and M2, Transmembrane Glycoprotein G, and L protein. The 7mer-m8 model was utilized to identify human miRNAs that target these viral genes. The interactions between the miRNAs and the genes were then assessed based on binding energy, GC content, and stability. The findings indicated that miRNAs, including miR-1279, miR-4251, miR-4288, and miR-12117, successfully target the N gene. In addition, other miRNAs target the remaining viral genes, indicating their capacity to bind and potentially inhibit viral replication. In addition, docking experiments have verified the stability of miRNA-mRNA duplexes, as evidenced by the high free energy values that indicate strong and reliable contacts between miRNA and gene. These findings indicate that certain human miRNAs have the potential to be effective therapeutic agents against the rabies virus by suppressing gene expression. This offers a new and innovative strategy to fight against this deadly infection.
Collapse
Affiliation(s)
- Aparajita Roy
- Department of Biotechnology, Assam University, Silchar, Assam, 788011, India
| | - Supriyo Chakraborty
- Department of Biotechnology, Assam University, Silchar, Assam, 788011, India.
| |
Collapse
|
10
|
Webb J, Zhao M, Campbell AH, Paul NA, Cummins SF, Eamens AL. The microRNA Pathway of Macroalgae: Its Similarities and Differences to the Plant and Animal microRNA Pathways. Genes (Basel) 2025; 16:442. [PMID: 40282402 PMCID: PMC12026948 DOI: 10.3390/genes16040442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 03/31/2025] [Accepted: 04/05/2025] [Indexed: 04/29/2025] Open
Abstract
In plants and animals, the microRNA (miRNA) class of small regulatory RNA plays an essential role in controlling gene expression in all aspects of development, to respond to environmental stress, or to defend against pathogen attack. This well-established master regulatory role for miRNAs has led to each protein-mediated step of both the plant and animal miRNA pathways being thoroughly characterized. Furthermore, this degree of characterization has led to the development of a suite of miRNA-based technologies for gene expression manipulation for fundamental research or for use in industrial or medical applications. In direct contrast, molecular research on the miRNA pathway of macroalgae, specifically seaweeds (marine macroalgae), remains in its infancy. However, the molecular research conducted to date on the seaweed miRNA pathway has shown that it shares functional features specific to either the plant or animal miRNA pathway. In addition, of the small number of seaweed species where miRNA data is available, little sequence conservation of individual miRNAs exists. These preliminary findings show the pressing need for substantive research into the seaweed miRNA pathway to advance our current understanding of this essential gene expression regulatory process. Such research will also generate the knowledge required to develop novel miRNA-based technologies for use in seaweeds. In this review, we compare and contrast the seaweed miRNA pathway to those well-characterized pathways of plants and animals and outline the low degree of miRNA sequence conservation across the polyphyletic group known as the seaweeds.
Collapse
Affiliation(s)
- Jessica Webb
- Seaweed Research Group, University of the Sunshine Coast, Maroochydore, QLD 4558, Australia (M.Z.); (A.H.C.); (N.A.P.); (S.F.C.)
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore, QLD 4558, Australia
| | - Min Zhao
- Seaweed Research Group, University of the Sunshine Coast, Maroochydore, QLD 4558, Australia (M.Z.); (A.H.C.); (N.A.P.); (S.F.C.)
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore, QLD 4558, Australia
| | - Alexandra H. Campbell
- Seaweed Research Group, University of the Sunshine Coast, Maroochydore, QLD 4558, Australia (M.Z.); (A.H.C.); (N.A.P.); (S.F.C.)
- School of Health, University of the Sunshine Coast, Maroochydore, QLD 4558, Australia
| | - Nicholas A. Paul
- Seaweed Research Group, University of the Sunshine Coast, Maroochydore, QLD 4558, Australia (M.Z.); (A.H.C.); (N.A.P.); (S.F.C.)
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore, QLD 4558, Australia
| | - Scott F. Cummins
- Seaweed Research Group, University of the Sunshine Coast, Maroochydore, QLD 4558, Australia (M.Z.); (A.H.C.); (N.A.P.); (S.F.C.)
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore, QLD 4558, Australia
| | - Andrew L. Eamens
- Seaweed Research Group, University of the Sunshine Coast, Maroochydore, QLD 4558, Australia (M.Z.); (A.H.C.); (N.A.P.); (S.F.C.)
- School of Health, University of the Sunshine Coast, Maroochydore, QLD 4558, Australia
| |
Collapse
|
11
|
Sciaraffa N, Santoni D, Li Greci A, Genovese SI, Coronnello C, Arancio W. Transcripts derived from AmnSINE1 repetitive sequences are depleted in the cortex of autism spectrum disorder patients. FRONTIERS IN BIOINFORMATICS 2025; 5:1532981. [PMID: 40270680 PMCID: PMC12015672 DOI: 10.3389/fbinf.2025.1532981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/24/2025] [Indexed: 04/25/2025] Open
Abstract
Aims Autism spectrum disorder (ASD) is a brain developmental disability with a not-fully clarified etiogenesis. Current ASD research largely focuses on coding regions of the genome, but up to date much less is known about the contribution of non-coding elements to ASD risk. The non-coding genome is largely made of DNA repetitive sequences (RS). Although RS were considered slightly more than "junk DNA", today RS have a recognized role in almost every aspect of human biology, especially in developing human brain. Our aim was to test if RS transcription may play a role in ASD. Methods Global RS transcription was firstly investigated in postmortem dorsolateral prefrontal cortex of 13 ASD patients and 39 matched controls. Results were validated in independent datasets. Results AmnSINE1 was the only RS significantly downregulated in ASD specimens. The role of AmnSINE1 in ASD has been investigated at multiple levels, showing that the 1,416 genes containing AmnSINE1 are associated with nervous system development and autism susceptibility. This has been confirmed in a different experimental setting, such as in organoid models of the human cerebral cortex, harboring different ASD causative mutations. AmnSINE1 related genes are transcriptionally co-regulated and are involved not only in brain formation but can specifically be involved in ASD development. Looking for a possible direct role of AmnSINE1 non-coding transcripts in ASD, we report that AmnSINE1 transcripts may alter the miRNA regulatory landscape for genes involved in neurogenesis. Conclusion Our findings provide preliminary evidence supporting a role for AmnSINE1 in ASD development.
Collapse
Affiliation(s)
| | - Daniele Santoni
- Institute for System Analysis and Computer Science “Antonio Ruberti”, National Research Council of Italy (IASI-CNR), Rome, Italy
| | - Andrea Li Greci
- Advanced Data Analysis Group, Ri. MED Foundation, Palermo, Italy
| | | | | | - Walter Arancio
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| |
Collapse
|
12
|
Despard BA, Selwyn JD, Shupp AN, Vollmer SV. A Network Approach to White Band Disease Challenged Staghorn Coral Acropora cervicornismicroRNAs and Their Targets. Ecol Evol 2025; 15:e71351. [PMID: 40290387 PMCID: PMC12022774 DOI: 10.1002/ece3.71351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 04/10/2025] [Accepted: 04/13/2025] [Indexed: 04/30/2025] Open
Abstract
Coral reefs are increasingly threatened by disease outbreaks, yet little is known about the genetic mechanisms underlying disease resistance. Since the 1970s, White Band Disease (WBD) has decimated the Caribbean staghorn coral Acropora cervicornis. However, 15% or more of individuals are highly disease-resistant, and the genes controlling the production of Argonaut proteins, involved in microRNA (miRNA) post-transcriptional gene silencing, are up-regulated in WBD-resistant corals. This suggests that miRNAs may be key regulators of coral immunity. In this study, we conducted an in situ disease transmission experiment with five healthy-exposed control tanks and five WBD-exposed tanks, each containing 50 A. cervicornis genotypes, sampled over 7 days and then sequenced miRNAs from 12 replicate genotypes, including 12 WBD-exposed and 12 healthy-exposed control fragments from two time points. We identified 67 bona fide miRNAs in A. cervicornis, 3 of which are differentially expressed in disease-resistant corals. We performed a phylogenetic comparison of miRNAs across cnidarians and found greater conservation of miRNAs in more closely related taxa, including all three differentially expressed miRNAs being conserved in more than one Acropora coral. One of the three miRNAs has putative genomic targets involved in the cnidarian innate immunity. In addition, community detection coupled with over-representation analysis of our miRNA-messenger RNA (mRNA) target network found two key unique A. cervicornis miRNAs regulating multiple important immune-related pathways such as Toll-like receptor pathway, endocytosis, and apoptosis. These findings highlight how multiple miRNAs may help the coral host maintain immune homeostasis in the presence of environmental stress including disease.
Collapse
Affiliation(s)
- Brecia A. Despard
- Department of Marine and Environmental SciencesNortheastern UniversityNahantMassachusettsUSA
| | - Jason D. Selwyn
- Department of Marine and Environmental SciencesNortheastern UniversityNahantMassachusettsUSA
- Genomics CORE LaboratoryTexas A&M University—Corpus ChristiCorpus ChristiTexasUSA
| | - Allison N. Shupp
- Department of Marine and Environmental SciencesNortheastern UniversityNahantMassachusettsUSA
| | - Steven V. Vollmer
- Department of Marine and Environmental SciencesNortheastern UniversityNahantMassachusettsUSA
| |
Collapse
|
13
|
Peng HY, Huang YL, Wu PH, Li LJ, Peng BY, Wu CY, Lin YL, Hsiao M, Chang JY, Mu-Hsin Chang P, Lee HL, Chang WM. The miR-876-5p/SOCS4/STAT3 pathway induced the expression of PD-L1 and suppressed antitumor immune responses. Cancer Cell Int 2025; 25:114. [PMID: 40140827 PMCID: PMC11938556 DOI: 10.1186/s12935-025-03704-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) remains a formidable challenge due to its high recurrence rates and poor prognosis. This study focuses on miR-876, a microRNA significantly associated with OSCC recurrence and clinical outcomes. Analysis of miRNA expression profiles from recurrent OSCC patients revealed that miR-876-5p is markedly upregulated in recurrent tumor tissues and the high expression of miR-876-5p correlates with reduced disease-free and overall survival. Functional assays demonstrated that miR-876 enhances OSCC cell growth, migration, and stemness, contributing to chemoresistance. Mechanistically, miR-876-5p directly targets SOCS4, leading to increased STAT3 activation and subsequent upregulation of PD-L1, which facilitates immune evasion. Additionally, exposure to the tobacco-specific carcinogen NNK was found to induce miR-876 expression and STAT3 activation, implicating environmental factors in miR-876 regulation and promote cancer recurrent. These findings identify the miR-876-5p-SOCS4-STAT3 axis as a critical pathway in OSCC progression, highlighting miR-876-5p as a potential biomarker and therapeutic target to improve treatment outcomes in OSCC patients.
Collapse
Affiliation(s)
- Hsuan-Yu Peng
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- Research Center of Oral Translational Medicine, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Li Huang
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Taipei Medical University Hospital, Taipei, Taiwan
| | - Ping-Hsiu Wu
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan
- TMU Proton Center, Taipei Medical University, Taipei, Taiwan
| | - Li-Jie Li
- Program of School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Oral Pathology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Bou-Yue Peng
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chia-Yu Wu
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Taipei Medical University Hospital, Taipei, Taiwan
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Lung Lin
- Program for Translational Medicine, College of Medical Sciences and Technology, Taipei Medical University, Taipei, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Jang-Yang Chang
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Peter Mu-Hsin Chang
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, College of Medicine, National Yang-Ming University, Taipei, Taiwan
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsin-Lun Lee
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan
- TMU Proton Center, Taipei Medical University, Taipei, Taiwan
| | - Wei-Min Chang
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.
- Research Center of Oral Translational Medicine, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
14
|
Mohebbi M, Manzourolajdad A, Bennett E, Williams P. A Multi-Input Neural Network Model for Accurate MicroRNA Target Site Detection. Noncoding RNA 2025; 11:23. [PMID: 40126347 PMCID: PMC11932204 DOI: 10.3390/ncrna11020023] [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: 12/16/2024] [Revised: 02/07/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
(1) Background: MicroRNAs are non-coding RNA sequences that regulate cellular functions by targeting messenger RNAs and inhibiting protein synthesis. Identifying their target sites is vital to understanding their roles. However, it is challenging due to the high cost and time demands of experimental methods and the high false-positive rates of computational approaches. (2) Methods: We introduce a Multi-Input Neural Network (MINN) algorithm that integrates diverse biologically relevant features, including the microRNA duplex structure, substructures, minimum free energy, and base-pairing probabilities. For each feature derived from a microRNA target-site duplex, we create a corresponding image. These images are processed in parallel by the MINN algorithm, allowing it to learn a comprehensive and precise representation of the underlying biological mechanisms. (3) Results: Our method, on an experimentally validated test set, detects target sites with an AUPRC of 0.9373, Precision of 0.8725, and Recall of 0.8703 and outperforms several commonly used computational methods of microRNA target-site predictions. (4) Conclusions: Incorporating diverse biologically explainable features, such as duplex structure, substructures, their MFEs, and binding probabilities, enables our model to perform well on experimentally validated test data. These features, rather than nucleotide sequences, enhance our model to generalize beyond specific sequence contexts and perform well on sequentially distant samples.
Collapse
Affiliation(s)
- Mohammad Mohebbi
- Department of Computer Science and Information Science, University of North Georgia, Dahlonega, GA 30597, USA; (E.B.); (P.W.)
| | | | - Ethan Bennett
- Department of Computer Science and Information Science, University of North Georgia, Dahlonega, GA 30597, USA; (E.B.); (P.W.)
| | - Phillip Williams
- Department of Computer Science and Information Science, University of North Georgia, Dahlonega, GA 30597, USA; (E.B.); (P.W.)
| |
Collapse
|
15
|
Bogusławska J, Rakhmetullina A, Grzanka M, Białas A, Rybicka B, Życka-Krzesińska J, Molcan T, Zielenkiewicz P, Pączek L, Piekiełko-Witkowska A. miR395e from Manihot esculenta Decreases Expression of PD-L1 in Renal Cancer: A Preliminary Study. Genes (Basel) 2025; 16:293. [PMID: 40149445 PMCID: PMC11942022 DOI: 10.3390/genes16030293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: microRNAs are small non-coding RNAs that regulate gene expression by inducing mRNA degradation or inhibiting translation. A growing body of evidence suggests that miRNAs may be utilized as anti-cancer therapeutics by targeting expression of key genes involved in cancerous transformation and progression. Renal cell cancer (RCC) is the most common kidney malignancy. The most efficient RCC treatments involve blockers of immune checkpoints, including antibodies targeting PD-L1 (Programmed Death Ligand 1). Interestingly, recent studies revealed the cross-kingdom horizontal transfer of plant miRNAs into mammalian cells, contributing to the modulation of gene expression by food ingestion. Here, we hypothesized that PD-L1 expression may be modulated by miRNAs originating from edible plants. Methods: To verify this hypothesis, we performed bioinformatic analysis to identify mes-miR395e from Manihot esculenta (cassava) as a promising candidate miRNA that could target PD-L1. To verify PD-L1 regulation mediated by the predicted plant miRNA, synthetic mes-miR395 mimics were transfected into cell lines derived from RCC tumors, followed by evaluation of PD-L1 expression using qPCR and Western blot. Results: Transfection of mes-miR395e mimics into RCC-derived cell lines confirmed that this miRNA decreases expression of PD-L1 in RCC cells at both mRNA and protein levels. Conclusions: This preliminary study shows the promise of plant miRNA as potential adjuvants supporting RCC treatment.
Collapse
Affiliation(s)
- Joanna Bogusławska
- Department of Biochemistry and Molecular Biology, Centre of Translational Research, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813 Warsaw, Poland; (J.B.); (M.G.); (A.B.); (B.R.); (J.Ż.-K.)
| | - Aizhan Rakhmetullina
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, ul. Pawińskiego 5a, 02-106 Warsaw, Poland; (A.R.); (T.M.); (P.Z.); (L.P.)
| | - Małgorzata Grzanka
- Department of Biochemistry and Molecular Biology, Centre of Translational Research, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813 Warsaw, Poland; (J.B.); (M.G.); (A.B.); (B.R.); (J.Ż.-K.)
| | - Alex Białas
- Department of Biochemistry and Molecular Biology, Centre of Translational Research, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813 Warsaw, Poland; (J.B.); (M.G.); (A.B.); (B.R.); (J.Ż.-K.)
| | - Beata Rybicka
- Department of Biochemistry and Molecular Biology, Centre of Translational Research, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813 Warsaw, Poland; (J.B.); (M.G.); (A.B.); (B.R.); (J.Ż.-K.)
| | - Joanna Życka-Krzesińska
- Department of Biochemistry and Molecular Biology, Centre of Translational Research, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813 Warsaw, Poland; (J.B.); (M.G.); (A.B.); (B.R.); (J.Ż.-K.)
| | - Tomasz Molcan
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, ul. Pawińskiego 5a, 02-106 Warsaw, Poland; (A.R.); (T.M.); (P.Z.); (L.P.)
- Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima Street 10, 10-748 Olsztyn, Poland
| | - Piotr Zielenkiewicz
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, ul. Pawińskiego 5a, 02-106 Warsaw, Poland; (A.R.); (T.M.); (P.Z.); (L.P.)
- Department of Systems Biology, Institute of Experimental Plant Biology and Biotechnology, University of Warsaw, ul. Miecznikowa 1, 02-096 Warsaw, Poland
| | - Leszek Pączek
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, ul. Pawińskiego 5a, 02-106 Warsaw, Poland; (A.R.); (T.M.); (P.Z.); (L.P.)
- Department of Clinical Immunology, Medical University of Warsaw, 02-006 Warsaw, Poland
| | - Agnieszka Piekiełko-Witkowska
- Department of Biochemistry and Molecular Biology, Centre of Translational Research, Centre of Postgraduate Medical Education, ul. Marymoncka 99/103, 01-813 Warsaw, Poland; (J.B.); (M.G.); (A.B.); (B.R.); (J.Ż.-K.)
| |
Collapse
|
16
|
Gao Y, Cao J, Han B, Sun D. Preliminary exploration of mRNA, lncRNA, and miRNA expressions in the bovine jejunum unveils novel aspects of Mycobacterium avium subspecies paratuberculosis infections. BMC Genomics 2025; 26:108. [PMID: 39905315 PMCID: PMC11796175 DOI: 10.1186/s12864-025-11299-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/28/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Paratuberculosis (Johne's disease, JD) is a chronic and enteric disease in a range of ruminants, often caused by Mycobacterium avium subspecies paratuberculosis (MAP) infection, leading to substantial economic losses worldwide. Yet, the molecular underpinning of paratuberculosis remains elusive. Here we performed RNA sequencing (RNA-seq) and small RNA sequencing (sRNA-seq) of the jejunum tissues from the Holstein cows with three distinct statuses of paratuberculosis, i.e., healthy, subclinical, and clinical to screen potential genes, lncRNAs, and miRNAs associated with the resistance or susceptibility to MAP infection and build ceRNA regulatory networks via miRNAs. RESULTS We applied whole transcriptome sequencing analysis to examine the jejunum tissue in nine Holstein cows. Starting with 19,994 expressed genes, 13,529 lncRNAs, and 735 miRNAs, we screened out differentially expressed genes (DEGs), lncRNAs, and miRNAs of three comparison groups, i.e., clinical vs. healthy, subclinical vs. healthy, and clinical vs. subclinical, subsequently identifying ceRNA pairs. Ultimately, we detected 76, 74, and 24 DEGs, 19, 39, and 10 lncRNAs, as well as 28, 61, and 20 miRNAs across the three comparison groups, respectively. Through integrating these DEGs with functional annotation, previously reported QTLs, and GWAS results, we proposed eight genes (LYZ, LYZ1, BOLA-DQB, BOLA-DQA1, TAP, CATD, VNN1, and PPARG), six lncRNAs, 48 miRNAs, and 107 ceRNA pairs implying their potential associations with susceptibility to MAP infection. CONCLUSION The present study provided a global view of the dynamics in transcriptomes of the bovine jejunum tissues in terms of JD status. Our results demonstrated that not only mRNAs but also lncRNAs and miRNAs played important roles in regulating MAP infection in dairy cattle. This study provided a valuable resource for understanding the molecular basis of JD, potentially contributing to the genetic improvement of JD resistance in dairy cattle.
Collapse
Affiliation(s)
- Yahui Gao
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jie Cao
- College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Bo Han
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Dongxiao Sun
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
17
|
Zhan Y, Tian F, Fan W, Li X, Wang X, Zhang H, Hong X, Wang X, Cai L, Song Y, Xing Y. Targeting piRNA-137463 Inhibits Tumor Progression and Boosts Sensitivity to Immune Checkpoint Blockade via De Novo Cholesterol Biosynthesis in Lung Adenocarcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414100. [PMID: 39692168 PMCID: PMC11809383 DOI: 10.1002/advs.202414100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/03/2024] [Indexed: 12/19/2024]
Abstract
The important role of PIWI-interacting RNAs (piRNAs) in tumors has garnered increasing attention. However, research on their role in lung adenocarcinoma (LUAD) remains limited. Elevated levels of piRNA-137463 have been linked to poor prognosis in LUAD patients. Inhibition of piRNA-137463 curbed the proliferation, migration, and invasion of LUAD cells, enhanced T cell cytotoxicity through increased IFN-γ secretion, disrupted cholesterol metabolism, and reduced intracellular cholesterol, lipid raft content, and PD-L1 expression in LUAD cells. Bioinformatic prediction identified a potential interaction between piRNA-137463 and lncRNA LOC100128494. Inhibiting piRNA-137463 increased the stability and expression of LOC100128494, which further modulated insulin-induced gene 1 protein (INSIG1) levels via a competitive endogenous RNA network involving LOC100128494 and miR-24-3p. Notably, the effect of piRNA-137463 in LUAD cells is dependent on the expression of LOC100128494 and INSIG1. Inhibiting the expression of piRNA-137463 with AntagopiRNA-137463 suppressed tumor growth and metastasis via LOC100128494 in nude mice and enhanced the response of LUAD to anti-PD-1 therapy in immune-competent mice. In summary, this study elucidates the role of piRNA-137463 in the reprogramming of cholesterol metabolism, which drives the progression of LUAD, thereby identifying a new target for the comprehensive clinical management of LUAD.
Collapse
Affiliation(s)
- Yuning Zhan
- The Fourth Department of Medical OncologyHarbin Medical University Cancer Hospital150 Haping RoadHarbin150081China
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted TheranosticsHarbin Medical UniversityHarbin150001China
| | - Fanglin Tian
- The Fourth Department of Medical OncologyHarbin Medical University Cancer Hospital150 Haping RoadHarbin150081China
| | - Weina Fan
- The Fourth Department of Medical OncologyHarbin Medical University Cancer Hospital150 Haping RoadHarbin150081China
| | - Xin Li
- The Fourth Department of Medical OncologyHarbin Medical University Cancer Hospital150 Haping RoadHarbin150081China
| | - Xiangyu Wang
- The Fourth Department of Medical OncologyHarbin Medical University Cancer Hospital150 Haping RoadHarbin150081China
| | - Hongxia Zhang
- Imaging CenterHarbin Medical University Cancer HospitalHarbin150081China
| | - Xin Hong
- The Fourth Department of Medical OncologyHarbin Medical University Cancer Hospital150 Haping RoadHarbin150081China
| | - Xin Wang
- The Fourth Department of Medical OncologyHarbin Medical University Cancer Hospital150 Haping RoadHarbin150081China
| | - Li Cai
- The Fourth Department of Medical OncologyHarbin Medical University Cancer Hospital150 Haping RoadHarbin150081China
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted TheranosticsHarbin Medical UniversityHarbin150001China
| | - Yang Song
- The Department of OrthopedicsThe Second Affiliated Hospital of Harbin Medical UniversityHarbin150001China
| | - Ying Xing
- The Fourth Department of Medical OncologyHarbin Medical University Cancer Hospital150 Haping RoadHarbin150081China
| |
Collapse
|
18
|
Lin S, Qiu P. Predicting microRNA target genes using pan-cancer correlation patterns. BMC Genomics 2025; 26:77. [PMID: 39871129 PMCID: PMC11773953 DOI: 10.1186/s12864-025-11254-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 01/17/2025] [Indexed: 01/29/2025] Open
Abstract
The interaction relationship between miRNAs and genes is important as miRNAs play a crucial role in regulating gene expression. In the literature, several databases have been constructed to curate known miRNA target genes, which are valuable resources but likely only represent a small fraction of all miRNA-gene interactions. In this study, we constructed machine learning models to predict miRNA target genes that have not been previously reported. Using the miRNA and gene expression data from TCGA, we performed a correlation analysis between all miRNAs and all genes across multiple cancer types. The correlations served as features to describe each miRNA-gene pair. Using the existing databases of curated miRNA targets, we labeled the miRNA-gene pairs, and trained machine learning models to predict novel miRNA-gene interactions. For the miRNA-gene pairs that were consistently predicted across the models, we called them significant miRNA-gene pairs. Using held-out miRNA target databases and a literature survey, we validated 5.5% of the predicted significant miRNA-gene pairs. The remaining predicted miRNA-gene pairs could serve as hypotheses for experimental validation. Additionally, we explored several additional datasets that provided gene expression data before and after a specific miRNA perturbation and observed consistency between the correlation direction of predicted miRNA-gene pairs and their regulatory patterns. Together, this analysis revealed a novel framework for uncovering previously unidentified miRNA-gene relationships, enhancing the collective comprehension of miRNA-mediated gene regulation.
Collapse
Affiliation(s)
- Shuting Lin
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, 30332, Georgia, USA
| | - Peng Qiu
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, 30332, Georgia, USA.
| |
Collapse
|
19
|
Gao Y, Takenaka K, Xu SM, Cheng Y, Janitz M. Recent advances in investigation of circRNA/lncRNA-miRNA-mRNA networks through RNA sequencing data analysis. Brief Funct Genomics 2025; 24:elaf005. [PMID: 40251826 PMCID: PMC12008121 DOI: 10.1093/bfgp/elaf005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 04/21/2025] Open
Abstract
Non-coding RNAs (ncRNAs) are RNA molecules that are transcribed from DNA but are not translated into proteins. Studies over the past decades have revealed that ncRNAs can be classified into small RNAs, long non-coding RNAs and circular RNAs by genomic size and structure. Accumulated evidences have eludicated the critical roles of these non-coding transcripts in regulating gene expression through transcription and translation, thereby shaping cellular function and disease pathogenesis. Notably, recent studies have investigated the function of ncRNAs as competitive endogenous RNAs (ceRNAs) that sequester miRNAs and modulate mRNAs expression. The ceRNAs network emerges as a pivotal regulatory function, with significant implications in various diseases such as cancer and neurodegenerative disease. Therefore, we highlighted multiple bioinformatics tools and databases that aim to predict ceRNAs interaction. Furthermore, we discussed limitations of using current technologies and potential improvement for ceRNAs network detection. Understanding of the dynamic interplay within ceRNAs may advance the biological comprehension, as well as providing potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Yulan Gao
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Gate 11 via Botany St, Sydney, NSW 2052, Australia
| | - Konii Takenaka
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Gate 11 via Botany St, Sydney, NSW 2052, Australia
| | - Si-Mei Xu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Gate 11 via Botany St, Sydney, NSW 2052, Australia
| | - Yuning Cheng
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Gate 11 via Botany St, Sydney, NSW 2052, Australia
| | - Michael Janitz
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Gate 11 via Botany St, Sydney, NSW 2052, Australia
| |
Collapse
|
20
|
Pandey V, Srivastava A, Gupta R, Zaki HEM, Shafiq Shahid M, Gaur RK. In silico identification of chilli genome encoded MicroRNAs targeting the 16S rRNA and secA genes of " Candidatus phytoplasma trifolii ". FRONTIERS IN BIOINFORMATICS 2025; 4:1493712. [PMID: 39834655 PMCID: PMC11743513 DOI: 10.3389/fbinf.2024.1493712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
Phytoplasma, a potentially hazardous pathogen associated with witches' broom, is an economically harmful disease-producing bacteria that damages chilli cultivation. Phytoplasma-infected plants display various symptoms that indicate significant disruptions in normal plant physiology and behaviour. Diseases caused by phytoplasma are widespread and have a major economic impact on crop quality and yield. This work focuses on identifying and examining chilli microRNAs (miRNAs) as potential targets against the 16S rRNA and secA gene of "Candidatus Phytoplasma trifolii" ("Ca. P. trifolii") through plant miRNA prediction algorithms. Mature chilli miRNAs (CA-miRNAs) were collected and used to hybridise the 16S rRNA and secA genes. A total of four common CA-miRNAs were picked according to genetic consensus. Three algorithms applied in the present study suggested that the physiologically relevant, top-ranked miR169b_2 has a possibly specific site at nucleotide position 1,006 for targeting the 'Ca. P. trifolii' 16S rRNA gene. The circos algorithm was then utilised to create the miRNA-mRNA regulatory network. The free energy between the miRNA:mRNA duplex was also computed, and the best value of -17.46 kcal/mol was obtained for CA-miR166c_2. Currently, there are no suitable commercial 'Ca. P. trifolii'-resistant chilli crops. As a result, the expected biological data provide useful evidence for developing 'Ca. P. trifolii'-resistant chilli plants.
Collapse
Affiliation(s)
- Vineeta Pandey
- Department of Biotechnology, Deen Dayal Upadhyaya Gorakhpur University, Gorakhpur, Uttar Pradesh, India
| | - Aarshi Srivastava
- Department of Biotechnology, Deen Dayal Upadhyaya Gorakhpur University, Gorakhpur, Uttar Pradesh, India
| | - Ramwant Gupta
- Department of Botany, Deen Dayal Upadhyaya Gorakhpur University, Gorakhpur, Uttar Pradesh, India
| | - Haitham E. M. Zaki
- Horticulture Department, Faculty of Agriculture, Minia University, El-Minia, Egypt
- Applied Biotechnology Department, University of Technology and Applied Sciences-Sur, Sur, Oman
| | - Muhammad Shafiq Shahid
- Department of Plant Sciences, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al‐khod, Oman
| | - Rajarshi K. Gaur
- Department of Biotechnology, Deen Dayal Upadhyaya Gorakhpur University, Gorakhpur, Uttar Pradesh, India
| |
Collapse
|
21
|
Shi Y, Wu X, Meng G, Ma X, La Y, Bao P, Chu M, Yan P. Identification and Analysis of Circular RNAs in Mammary Gland from Yaks Between Lactation and Dry Period. Animals (Basel) 2025; 15:89. [PMID: 39795032 PMCID: PMC11718809 DOI: 10.3390/ani15010089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/25/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
Lactation is a complex physiological process regulated by numerous genes and factors. Circular RNA (circRNA), a non-coding RNA, acts as a molecular sponge that sequesters microRNAs (miRNAs) to regulate target gene expression. Although circRNA has been linked to mammary gland lactation, its specific role in yaks remains underexplored. This study employed circular RNA sequencing (circRNA-seq) to examine the differential expression of circRNAs in yak mammary tissues during lactation and the dry period. Additionally, an enrichment analysis of the differentially expressed circRNAs (DECs) was performed. A competing endogenous RNA (ceRNA) network was then constructed to explore the potential of their roles in lactation and mammary gland development. We detected 18,905 circRNAs in yak mammary tissue, among which 302 showed differential expression. The host genes of these DECs were enriched in functions and pathways associated with yak milk synthesis and composition. Through the construction of a ceRNA network and the enrichment analysis of associated mRNAs, this study identified ceRNAs potentially involved in regulating lactation and mammary gland development. In conclusion, circRNAs in yak mammary tissues were identified and analyzed across lactation and dry periods, establishing a ceRNA network related to lactation regulation. These findings provide novel insights into the regulatory mechanisms governing lactation in yaks (Bos grunniens).
Collapse
Affiliation(s)
- Yilin Shi
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (Y.S.); (X.W.); (G.M.); (X.M.); (Y.L.); (P.B.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Xiaoyun Wu
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (Y.S.); (X.W.); (G.M.); (X.M.); (Y.L.); (P.B.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Guangyao Meng
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (Y.S.); (X.W.); (G.M.); (X.M.); (Y.L.); (P.B.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Xiaoming Ma
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (Y.S.); (X.W.); (G.M.); (X.M.); (Y.L.); (P.B.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Yongfu La
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (Y.S.); (X.W.); (G.M.); (X.M.); (Y.L.); (P.B.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Pengjia Bao
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (Y.S.); (X.W.); (G.M.); (X.M.); (Y.L.); (P.B.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Min Chu
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (Y.S.); (X.W.); (G.M.); (X.M.); (Y.L.); (P.B.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
| | - Ping Yan
- Key Laboratory of Yak Breeding of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China; (Y.S.); (X.W.); (G.M.); (X.M.); (Y.L.); (P.B.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730050, China
- Institute of Western Agriculture, Chinese Academy of Agricultural Sciences, Changji 931100, China
| |
Collapse
|
22
|
Marceca GP, Romano G, Acunzo M, Nigita G. ncRNA Editing: Functional Characterization and Computational Resources. Methods Mol Biol 2025; 2883:455-495. [PMID: 39702721 DOI: 10.1007/978-1-0716-4290-0_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Non-coding RNAs (ncRNAs) play crucial roles in gene expression regulation, translation, and disease development, including cancer. They are classified by size in short and long non-coding RNAs. This chapter focuses on the functional implications of adenosine-to-inosine (A-to-I) RNA editing in both short (e.g., miRNAs) and long ncRNAs. RNA editing dynamically alters the sequence and structure of primary transcripts, impacting ncRNA biogenesis and function. Notable findings include the role of miRNA editing in promoting glioblastoma invasiveness, characterizing RNA editing hotspots across cancers, and its implications in thyroid cancer and ischemia. This chapter also highlights bioinformatics resources and next-generation sequencing (NGS) technologies that enable comprehensive ncRNAome studies and genome-wide RNA editing detection. Dysregulation of RNA editing machinery has been linked to various human diseases, emphasizing the potential of RNA editing as a biomarker and therapeutic target. This overview integrates current knowledge and computational tools for studying ncRNA editing, providing insights into its biological significance and clinical applications.
Collapse
Affiliation(s)
| | - Giulia Romano
- Division of Pulmonary Diseases and Critical Care Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Mario Acunzo
- Division of Pulmonary Diseases and Critical Care Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Giovanni Nigita
- Department of Cancer Biology and Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
- Center for RNA Biology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
23
|
Chan TCL, Yagound B, Brown GP, Eyck HJF, Shine R, Rollins LA. Infection by the Lungworm Rhabdias pseudosphaerocephala Affects the Expression of Immune-Related microRNAs by Its Co-Evolved Host, the Cane Toad Rhinella marina. Mol Ecol 2025; 34:e17587. [PMID: 39544005 DOI: 10.1111/mec.17587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 10/09/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024]
Abstract
Parasites may suppress the immune function of infected hosts using microRNAs (miRNAs) to prevent protein production. Nonetheless, little is known about the diversity of miRNAs and their mode(s) of action. In this study, we investigated the effects of infection by a parasitic lungworm (Rhabdias pseudosphaerocephala) on miRNA and mRNA expression of its host, the invasive cane toad (Rhinella marina). To investigate the cane toad's innate and adaptive immune response to this parasite, we compared miRNA and mRNA expression in naïve toads that had never been infected by lungworms to toads that were infected with lungworms for the first time in their lives, and toads that were infected the second time in their lives (i.e., had two consecutive infections). In total, we identified 101 known miRNAs and 86 potential novel miRNAs. Compared to uninfected and single-infection toads, multiple-infection animals drastically downregulated three miRNAs. These miRNAs were associated with gene pathways related to the immune response, potentially reflecting the immunosuppression of cane toads by their parasites. Infected hosts did not respond with substantially differential mRNA transcription; only one gene was differentially expressed between control and single-infection hosts. Our study suggests that miRNA may play an important role in mediating host-parasite interactions in a system in which an ongoing range expansion by the host has generated substantial divergence in host-parasite interactions.
Collapse
Affiliation(s)
- Tsering C L Chan
- Ecology & Evolution Research Centre, School of Biological, Earth and Environmental Sciences, University of New South Wales, UNSW, Sydney, New South Wales, Australia
| | - Boris Yagound
- Ecology & Evolution Research Centre, School of Biological, Earth and Environmental Sciences, University of New South Wales, UNSW, Sydney, New South Wales, Australia
| | - Gregory P Brown
- School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Harrison J F Eyck
- Ecology & Evolution Research Centre, School of Biological, Earth and Environmental Sciences, University of New South Wales, UNSW, Sydney, New South Wales, Australia
| | - Richard Shine
- School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Lee A Rollins
- Ecology & Evolution Research Centre, School of Biological, Earth and Environmental Sciences, University of New South Wales, UNSW, Sydney, New South Wales, Australia
| |
Collapse
|
24
|
Bereczki Z, Benczik B, Balogh OM, Marton S, Puhl E, Pétervári M, Váczy-Földi M, Papp ZT, Makkos A, Glass K, Locquet F, Euler G, Schulz R, Ferdinandy P, Ágg B. Mitigating off-target effects of small RNAs: conventional approaches, network theory and artificial intelligence. Br J Pharmacol 2025; 182:340-379. [PMID: 39293936 DOI: 10.1111/bph.17302] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 05/07/2024] [Accepted: 06/17/2024] [Indexed: 09/20/2024] Open
Abstract
Three types of highly promising small RNA therapeutics, namely, small interfering RNAs (siRNAs), microRNAs (miRNAs) and the RNA subtype of antisense oligonucleotides (ASOs), offer advantages over small-molecule drugs. These small RNAs can target any gene product, opening up new avenues of effective and safe therapeutic approaches for a wide range of diseases. In preclinical research, synthetic small RNAs play an essential role in the investigation of physiological and pathological pathways as silencers of specific genes, facilitating discovery and validation of drug targets in different conditions. Off-target effects of small RNAs, however, could make it difficult to interpret experimental results in the preclinical phase and may contribute to adverse events of small RNA therapeutics. Out of the two major types of off-target effects we focused on the hybridization-dependent, especially on the miRNA-like off-target effects. Our main aim was to discuss several approaches, including sequence design, chemical modifications and target prediction, to reduce hybridization-dependent off-target effects that should be considered even at the early development phase of small RNA therapy. Because there is no standard way of predicting hybridization-dependent off-target effects, this review provides an overview of all major state-of-the-art computational methods and proposes new approaches, such as the possible inclusion of network theory and artificial intelligence (AI) in the prediction workflows. Case studies and a concise survey of experimental methods for validating in silico predictions are also presented. These methods could contribute to interpret experimental results, to minimize off-target effects and hopefully to avoid off-target-related adverse events of small RNA therapeutics. LINKED ARTICLES: This article is part of a themed issue Non-coding RNA Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.2/issuetoc.
Collapse
Affiliation(s)
- Zoltán Bereczki
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bettina Benczik
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Olivér M Balogh
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Szandra Marton
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Eszter Puhl
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Mátyás Pétervári
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Sanovigado Kft, Budapest, Hungary
| | - Máté Váczy-Földi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zsolt Tamás Papp
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - András Makkos
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Kimberly Glass
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Fabian Locquet
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Giessen, Germany
| | - Gerhild Euler
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Giessen, Germany
| | - Rainer Schulz
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Giessen, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Bence Ágg
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
25
|
Sarı-Tunel F, Demirkan A, Vural B, Yıldız CE, Komurcu-Bayrak E. Omics Data Integration Uncovers mRNA-miRNA Interaction Regions in Genes Associated with Chronic Venous Insufficiency. Genes (Basel) 2024; 16:40. [PMID: 39858587 PMCID: PMC11765502 DOI: 10.3390/genes16010040] [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: 11/15/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Chronic venous insufficiency (CVI), a chronic vascular dysfunction, is a common health problem that causes serious complications such as painful varicose veins and even skin ulcers. Identifying the underlying genetic and epigenetic factors is important for improving the quality of life of individuals with CVI. In the literature, many genes, variants, and miRNAs associated with CVI have been identified through genomic and transcriptomic studies. Despite molecular pathogenesis studies, how the genes associated with CVI are regulated by miRNAs and the effect of variants in binding regions on expression levels are still not fully understood. In this study, previously identified genes, variants, and miRNAs associated with CVI, common variants in the mRNA-miRNA binding regions, were investigated using in silico analyses. Methods: For this purpose, miRNA research tools, MBS (miRNA binding site) database, genome browsers, and the eQTL Calculator in the GTEx portal were used. Results: We identified SNVs associated with CVI that may play a direct role in the miRNA-mediated regulation of the ZNF664, COL1A2, HFE, MDN, MTHFR, SRPX, TDRD5, TSPYL4, VEGFA, and APOE genes. In addition, when the common SNVs in the mRNA binding region of 75 unique CVI related-miRNAs in five candidate genes associated with CVI were examined, seven miRNAs associated with the expression profiles of ABCA1, PIEZO1, and CASZ1 genes were identified. Conclusions: In conclusion, the relationship between genetic markers identified in the literature that play a role in the pathogenesis of the CVI and the expression profiles was evaluated for the first time in the mRNA-miRNA interaction axis.
Collapse
Affiliation(s)
- Fatma Sarı-Tunel
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, 34093 Istanbul, Turkey; (F.S.-T.); (B.V.)
- Graduate School Institute of Health Sciences, Istanbul University, 34093 Istanbul, Turkey
| | - Ayse Demirkan
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, School of Biosciences and Medicine and People-Centred AI Institute, University of Surrey, Guildford GU2 7XH, UK
| | - Burcak Vural
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, 34093 Istanbul, Turkey; (F.S.-T.); (B.V.)
| | - Cenk Eray Yıldız
- Department of Cardiovascular Surgery, Institute of Cardiology, Istanbul University-Cerrahpasa, 34098 Istanbul, Turkey;
| | - Evrim Komurcu-Bayrak
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, 34093 Istanbul, Turkey;
| |
Collapse
|
26
|
Chen W, Xie Y, Xu Z, Shang Y, Yang W, Wang P, Wu Z, Cai G, Hong L. Identification and Functional Analysis of miRNAs in Extracellular Vesicles of Semen Plasma from High- and Low-Fertility Boars. Animals (Basel) 2024; 15:40. [PMID: 39794983 PMCID: PMC11718777 DOI: 10.3390/ani15010040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Artificial insemination (AI), as an efficient assisted reproduction technology, can help the livestock industry to improve livestock and poultry breeds, optimize production performance and improve reproductive efficiency. AI technology has been widely used in pig production in China, but boar fertility affects the effectiveness of AI, and more and more studies have shown that there are significant differences in the fertility of boars with similar semen quality indicators. Therefore, this study aimed to identify biomarker molecules that indicate the level of boar fertility, which is important for improving the efficiency of AI. In this study, we collected 40 mL of ejaculates per boar used for extracellular vesicle (EV) characterization in 20 boars and identified 53 differentially expressed miRNAs by small RNA sequencing, of which 44 miRNAs were up-regulated in the high-fertility seminal EVs compared with low-fertility seminal EVs, and nine miRNAs were down-regulated. miR-26a was most significantly down-regulated in the high-fertility group compared to the low-fertility group, and it was hypothesized that this miRNA could be used as a biomolecular marker of semen reproductive performance. To further determine the effect of miR-26a on sperm function, we successfully established a miR-26a overexpression model and found that miR-26a reduced sperm viability, motility, acrosome integrity, plasma membrane integrity and ATP levels. Bioinformatics analysis and dual luciferase reporter analysis revealed that miR-26a directly targets High mobility group A1 (HMGA1). In conclusion, miR-26a can be used as a biomarker to identify high and low fertility in boar semen.
Collapse
Affiliation(s)
- Weidong Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
| | - Yanshe Xie
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
| | - Zhiqian Xu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China;
| | - Yijun Shang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
| | - Wenzheng Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
| | - Pengyao Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
| | - Zhenfang Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
- Yunfu Subcenter of Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu 527300, China
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou 510520, China
| | - Gengyuan Cai
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
- Yunfu Subcenter of Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu 527300, China
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou 510520, China
- National Regional Gene Bank of Livestock and Poultry (Gene Bank of Guangdong Livestock and Poultry), Guangzhou 510642, China
| | - Linjun Hong
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou 510520, China
- National Regional Gene Bank of Livestock and Poultry (Gene Bank of Guangdong Livestock and Poultry), Guangzhou 510642, China
| |
Collapse
|
27
|
Zhang JN, Gong R, Lu BT, Wang YQ, Chong Y, Wang XT, Lai QQ, Cao YH, Zhao MY. Integrated Analysis of Gene Expression and Immune Cell Infiltration Reveals Dysregulated Genes and miRNAs in Acute Kidney Injury. Mol Biotechnol 2024:10.1007/s12033-024-01344-x. [PMID: 39661223 DOI: 10.1007/s12033-024-01344-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 11/26/2024] [Indexed: 12/12/2024]
Abstract
Acute Kidney Injury (AKI) is a multifaceted condition characterised by rapid deterioration of renal function, often precipitated by diverse etiologies. A comprehensive understanding of the molecular underpinnings of AKI is pivotal for identifying potential diagnostic markers and therapeutic targets. This study utilised bioinformatics to elucidate gene expression and immune infiltration in AKI. Publicly available mRNA and miRNA datasets were harnessed to discern differentially expressed genes (DEGs) and miRNAs in AKI. The CIBERSORT algorithm was employed to quantify immune cell infiltration in AKI samples. Functional enrichment analyses were conducted to unravel the implicated biological processes. Furthermore, the expression of identified genes and miRNAs was validated by quantitative real-time PCR in an AKI model. Our study revealed significant dysregulation of three genes (Aspn, Clec2h, Tmigd1) and two miRNAs (mmu-miR-21a-3p, mmu-miR-223-3p) in AKI, each with p < 0.0001. These molecular markers are implicated in immune responses, tissue remodelling, and inflammation. We observed notable disturbances in specific immune cells, including activated and immature dendritic cells, M1 macrophages, and subsets of T cells (Treg, Th1, Th17). These alterations correlated significantly with AKI pathology, with dendritic cells and M1 macrophages showing p < 0.01, and T cell subsets demonstrating p < 0.05. These results highlight the intricate involvement of the immune system in AKI and indicate significant enrichment of pathways related to immune response, inflammation, and tissue remodelling, pointing to their pivotal roles in AKI pathophysiology. Our study underscored the significance of immune cell infiltration and dysregulated gene and miRNA expression in AKI. The identified genes (Clec2h, Aspn, and Tmigd1) and miRNAs (mmu-miR-21a-3p and mmu-miR-223-3p) offer potential diagnostic markers and therapeutic avenues for AKI. Subsequent investigations targeting these genes and miRNAs, along with the elucidated pathways, may augment the clinical management and outcomes for AKI patients.
Collapse
Affiliation(s)
- Jian-Nan Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang Province, China
| | - Rui Gong
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, 430022, China
| | - Bai-Tao Lu
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang Province, China
| | - Yi-Qi Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang Province, China
| | - Yang Chong
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang Province, China
| | - Xin-Tong Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang Province, China
| | - Qi-Qi Lai
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang Province, China
| | - Yan-Hui Cao
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang Province, China.
| | - Ming-Yan Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
28
|
Mastriano S, Kanoria S, Rennie W, Liu C, Li D, Cheng J, Ding Y, Lu J. High-Throughput Quantification of miRNA-3'-Untranslated-Region Regulatory Effects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.626985. [PMID: 39677669 PMCID: PMC11643113 DOI: 10.1101/2024.12.05.626985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
MicroRNAs (miRNAs) regulate gene expression post-transcriptionally, primarily through binding sites in 3' untranslated regions (3' UTRs). While computational and biochemical approaches have been developed to predict miRNA binding sites on target messenger RNAs, reliable and high-throughput assessment of the regulatory effects of miRNAs on full-length 3' UTRs can still be challenging. Utilizing a miniaturized and high-throughput reporter assay, we present a 'pilot miRNA-targeting map', containing 4,994 successfully measured miRNA:3' UTR regulatory outputs by pairwise assays between 461 miRNAs and eleven 3' UTRs. This collection represents a large experimental miRNA:3' UTR dataset to date on a single platform. The methodology can be generally applied to studies of miRNA-mediated regulation of critical genes. We found that seedless sites can lead to substantial downregulation. We utilized this dataset in the development of a quantitative total score for modeling the total regulatory effects by both seed and seedless sites on a full-length 3' UTR. To assess the predictive value of the total score, we analyzed data from mRNA expression and proteomics studies. We found that the score can discriminate the potent miRNA inhibition from the weak inhibition and is thus useful for quantitative prediction of miRNA regulation. The score has been added to the STarMir program of the Sfold package now available via GitHub at https://github.com/Ding-RNA-Lab/Sfold.
Collapse
Affiliation(s)
- Stephen Mastriano
- Department of Genetics and Yale Stem Cell Center, Yale University, New Haven, CT 06520, USA
- Yale Cancer Center and Center for RNA Science and Medicine, Yale University, New Haven, CT 06520, USA
| | - Shaveta Kanoria
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY 12201, USA
| | - William Rennie
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY 12201, USA
| | - Chaochun Liu
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY 12201, USA
| | - Dan Li
- Department of Genetics and Yale Stem Cell Center, Yale University, New Haven, CT 06520, USA
- Yale Cancer Center and Center for RNA Science and Medicine, Yale University, New Haven, CT 06520, USA
- Current address: Shanghai Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Jijun Cheng
- Department of Genetics and Yale Stem Cell Center, Yale University, New Haven, CT 06520, USA
- Yale Cancer Center and Center for RNA Science and Medicine, Yale University, New Haven, CT 06520, USA
| | - Ye Ding
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY 12201, USA
| | - Jun Lu
- Department of Genetics and Yale Stem Cell Center, Yale University, New Haven, CT 06520, USA
- Yale Cancer Center and Center for RNA Science and Medicine, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
29
|
Song J, Li W, Gao L, Yan Q, Zhang X, Liu M, Zhou S. miR-276 and miR-182013-5p modulate insect metamorphosis and reproduction via dually regulating juvenile hormone acid methyltransferase. Commun Biol 2024; 7:1604. [PMID: 39623057 PMCID: PMC11612435 DOI: 10.1038/s42003-024-07285-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024] Open
Abstract
Juvenile hormone (JH) represses insect metamorphosis and stimulates reproduction. JH titers are generally low in juveniles, drop to a nadir during metamorphosis, increase after eclosion and peak in vitellogenic phase. We found that Jhamt, a rate-limiting enzyme in JH biosynthesis, mirrors JH titer patterns in the migratory locust. Knocking down Jhamt reduced JH titers, led to precocious nymphal ecdysis, metamorphosis and impaired vitellogenesis. Jhamt is negatively regulated by miR-276 and positively by miR-182013-5p. miR-276 is abundant in late nymphal but low in adults, while miR-182013-5p shows the opposite pattern. In nymphs, miR-276 binds more to Jhamt, while in adults, miR-182013-5p dominates. Functionally, miR-276 reduced Jhamt and JH levels, shortening nymphal development and inhibiting Vg expression. Conversely, miR-182013-5p increased Jhamt and JH levels, prolonging nymphal development and enhancing Vg expression. Our findings identify miR-276 and miR-182013-5p as dual regulators in JH biosynthesis, acting as "brake" and "accelerator," respectively. This study provides new insights into JH titer fluctuations and miRNA regulation in insect metamorphosis and reproduction.
Collapse
Affiliation(s)
- Jiasheng Song
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, Henan University, Kaifeng, China
| | - Wanwan Li
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, Henan University, Kaifeng, China
| | - Lulu Gao
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, Henan University, Kaifeng, China
| | - Qiang Yan
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, Henan University, Kaifeng, China
| | - Xinyan Zhang
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, Henan University, Kaifeng, China
| | - Mingzhi Liu
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, Henan University, Kaifeng, China
| | - Shutang Zhou
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, Henan University, Kaifeng, China.
| |
Collapse
|
30
|
Anarlouei S, Roohy F, Mohamadynejad P. Effect of rs1058240 polymorphism in 3'-UTR of GATA3 gene on potential binding of miRNAs and its association with RRMS risk: bioinformatics analysis and case-control study. Int J Neurosci 2024; 134:1541-1546. [PMID: 37842852 DOI: 10.1080/00207454.2023.2272043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/31/2023] [Accepted: 10/12/2023] [Indexed: 10/17/2023]
Abstract
AIM Multiple sclerosis is believed to be an autoimmune disease that is influenced by T helper (Th) cell differentiation. GATA3 plays an important role in reducing the development and severity of MS by shifting the differentiation of Th cells to Th2 and regulatory T cells while inhibiting the differentiation of Th1 and Th17 cells. Considering the functional role of rs1058240 SNP in the 3'-UTR of GATA3 mRNA, the association of target SNP with the risk of RRMS was examined. METHODS Genomic DNA was extracted from whole blood samples of 200 RRMS patients and 226 healthy individuals as a control group. Different genotypes of rs1058240 SNP were determined using the RFLP-PCR technique. Statistical analysis was performed using SPSS software and χ2 and logistic regression tests. The stability of GATA3 mRNA secondary structures and the binding patterns of GATA3-miRNAs with different alleles were evaluated using RNAfold and RNAhybrid programs, respectively. RESULTS The results indicated that the GATA3 rs1058240 G allele (p value = 0.010, OR = 1.45, CI = 1.09-1.93) and GG genotype (adjusted p value = 0.017, OR = 2.27, 95%CI = 1.16-4.44) increased the risk of RRMS, particularly in women (adjusted p value = 0.006, OR = 2.99, 95%CI = 1.37-6.52). Bioinformatics analysis revealed that although the allelic variation of this polymorphism had only a slight effect on mRNA stability (-177 to -177.20), the G allele significantly increased miRNA binding strength and miRNA-mRNA thermodynamic stability for hsa-miR-337-5p, hsa-miR-4445-3p, hsa-miR-4485-3p, hsa-miR-95-3p (ΔMFE > 0) compared to the A allele. CONCLUSION The G allele and GG genotype of rs1058240 in GATA3 mRNA 3'-UTR were found to be risk factors for increasing the susceptibility to RRMS.
Collapse
Affiliation(s)
- Shirin Anarlouei
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Fatemeh Roohy
- Department of Biology, Faculty of Basic Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Parisa Mohamadynejad
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
31
|
Uthayopas K, de Sá AG, Alavi A, Pires DE, Ascher DB. PRIMITI: A computational approach for accurate prediction of miRNA-target mRNA interaction. Comput Struct Biotechnol J 2024; 23:3030-3039. [PMID: 39175797 PMCID: PMC11340604 DOI: 10.1016/j.csbj.2024.06.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/20/2024] [Accepted: 06/23/2024] [Indexed: 08/24/2024] Open
Abstract
Current medical research has been demonstrating the roles of miRNAs in a variety of cellular mechanisms, lending credence to the association between miRNA dysregulation and multiple diseases. Understanding the mechanisms of miRNA is critical for developing effective diagnostic and therapeutic strategies. miRNA-mRNA interactions emerge as the most important mechanism to be understood despite their experimental validation constraints. Accordingly, several computational models have been developed to predict miRNA-mRNA interactions, albeit presenting limited predictive capabilities, poor characterisation of miRNA-mRNA interactions, and low usability. To address these drawbacks, we developed PRIMITI, a PRedictive model for the Identification of novel miRNA-Target mRNA Interactions. PRIMITI is a novel machine learning model that utilises CLIP-seq and expression data to characterise functional target sites in 3'-untranslated regions (3'-UTRs) and predict miRNA-target mRNA repression activity. The model was trained using a reliable negative sample selection approach and the robust extreme gradient boosting (XGBoost) model, which was coupled with newly introduced features, including sequence and genetic variation information. PRIMITI achieved an area under the receiver operating characteristic (ROC) curve (AUC) up to 0.96 for a prediction of functional miRNA-target site binding and 0.96 for a prediction of miRNA-target mRNA repression activity on cross-validation and an independent blind test. Additionally, the model outperformed state-of-the-art methods in recovering miRNA-target repressions in an unseen microarray dataset and in a collection of validated miRNA-mRNA interactions, highlighting its utility for preliminary screening. PRIMITI is available on a reliable, scalable, and user-friendly web server at https://biosig.lab.uq.edu.au/primiti.
Collapse
Affiliation(s)
- Korawich Uthayopas
- The Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Alex G.C. de Sá
- The Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Azadeh Alavi
- School of Computational Technology, RMIT University, Melbourne, VIC 3000, Australia
| | - Douglas E.V. Pires
- The Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- School of Computing and Information Systems, University of Melbourne, Parkville, VIC 3052, Australia
| | - David B. Ascher
- The Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
32
|
Bader S, Tuller T. Advanced computational predictive models of miRNA-mRNA interaction efficiency. Comput Struct Biotechnol J 2024; 23:1740-1754. [PMID: 38689718 PMCID: PMC11058727 DOI: 10.1016/j.csbj.2024.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/06/2024] [Accepted: 04/07/2024] [Indexed: 05/02/2024] Open
Abstract
The modeling of miRNA-mRNA interactions holds significant implications for synthetic biology and human health. However, this research area presents specific challenges due to the multifaceted nature of mRNA downregulation by miRNAs, influenced by numerous factors including competition or synergism among miRNAs and mRNAs. In this study, we present an improved computational model for predicting miRNA-mRNA interactions, addressing aspects not previously modeled. Firstly, we integrated a novel set of features that significantly enhanced the predictor's performance. Secondly, we demonstrated the cell-specific nature of certain aspects of miRNA-mRNA interactions, highlighting the importance of designing models tailored to specific cell types for improved accuracy. Moreover, we introduce a miRNA binding site interaction model (miBSIM) that, for the first time, accounts for both the distribution of miRNA binding sites along the mRNA and their respective strengths in regulating mRNA stability. Our analysis suggests that distant miRNA sites often compete with each other, revealing the intricate interplay of binding site interactions. Overall, our new predictive model shows a significant improvement of up to 6.43% over previous models in the field. The code of our model is available at https://www.cs.tau.ac.il/~tamirtul/miBSIM.
Collapse
Affiliation(s)
- Sharon Bader
- Department of Biomedical Engineering, Tel-Aviv University, Tel Aviv, Israel
| | - Tamir Tuller
- Department of Biomedical Engineering, Tel-Aviv University, Tel Aviv, Israel
- The Segol School of Neuroscience, Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
33
|
Baidya AK, Tiwary BK. A combination of conserved and stage-specific lncRNA biomarkers to detect lung adenocarcinoma progression. J Biomol Struct Dyn 2024:1-13. [PMID: 39601689 DOI: 10.1080/07391102.2024.2431190] [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: 01/07/2024] [Accepted: 04/19/2024] [Indexed: 11/29/2024]
Abstract
Lung adenocarcinoma is highly heterogeneous at the molecular level between different stages; therefore, understanding molecular mechanisms contributing to such heterogeneity is needed. In addition, multiple stages of progression are critical factors for lung adenocarcinoma treatment. However, previous studies showed that cancer progression is associated with altered lncRNA expression, highlighting the tissue-specific and developmental stage-specific nature of lncRNAs in various diseases. Therefore, a study using an integrated network approach to explore the role of lncRNA in carcinogenesis was done using expression profiles revealing stage-specific and conserved lncRNA biomarkers in lung adenocarcinoma. We constructed ceRNA networks for each stage of lung adenocarcinoma and analysed them using network topology, differential co-expression network, protein-protein interaction network, functional enrichment, survival analysis, genomic analysis and deep learning to identify potential lncRNA biomarkers. The co-expression networks of healthy and three successive stages of lung adenocarcinoma have shown different network properties. One conserved and four stage-specific lncRNAs are identified as genome regulatory biomarkers. These lncRNAs can successfully identify lung adenocarcinoma and different stages of progression using deep learning. In addition, we identified five mRNAs, four miRNAs and twelve novel carcinogenic interactions associated with the progression of lung adenocarcinoma. These lncRNA biomarkers will provide a novel perspective into the underlying mechanism of adenocarcinoma progression and may be further helpful in early diagnosis, treatment and prognosis of this deadly disease.
Collapse
Affiliation(s)
- Anil K Baidya
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Pondicherry, India
| | - Basant K Tiwary
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Pondicherry, India
| |
Collapse
|
34
|
Nazir A, Uwishema O, Shariff S, Franco WXG, El Masri N, Ayele ND, Munyangaju I, Alzain FE, Wojtara M. A Thorough Navigation of miRNA's Blueprint in Crafting Cardiovascular Fate. Health Sci Rep 2024; 7:e70136. [PMID: 39502130 PMCID: PMC11535861 DOI: 10.1002/hsr2.70136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Cardiovascular diseases contribute significantly to global morbidity and mortality. MicroRNAs are crucial in the development and progression of these diseases by regulating gene expression in various cells and tissues. Their roles in conditions like atherosclerosis, heart failure, myocardial infarction, and arrhythmias have been widely researched. Materials and Methods The present study provides an overview of existing evidence regarding miRNAs' role in cardiovascular disease pathogenesis. Furthermore, the study examines current state-of-the-art technologies used in the study of miRNAs in cardiovascular disease. As a final point, we examine how miRNAs may serve as disease biomarkers, therapeutic targets, and prognostic indicators. Results In cardiology, microRNAs, small noncoding RNA molecules, are crucial to the posttranscriptional regulation of genes. Their role in regulating cardiac cell differentiation and maturation is critical during the development of the heart. They maintain the cardiac function of an adult heart by contributing to its electrical and contractile activity. By binding to messenger RNA molecules, they inhibit protein translation or degrade mRNA. Several cardiovascular diseases are associated with dysregulation of miRNAs, including arrhythmias, hypertension, atherosclerosis, and heart failure. miRNAs can be used as biomarkers to diagnose and predict diseases as well as therapeutic targets. A variety of state-of-the-art technologies have aided researchers in discovering, profiling, and analyzing miRNAs, including microarray analysis, next-generation sequencing, and others. Conclusion Developing new diagnostics and therapeutic approaches is becoming more feasible as researchers refine their understanding of miRNA function. Ultimately, this will reduce the burden of cardiovascular disease around the world.
Collapse
Affiliation(s)
- Abubakar Nazir
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineKing Edward Medical UniversityPakistan
| | - Olivier Uwishema
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
| | - Sanobar Shariff
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineYerevan State Medical UniversityYerevanArmenia
| | - William Xochitun Gopar Franco
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineUniversity of GuadalajaraGuadalajaraMexico
| | - Noha El Masri
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Faculty of MedicineBeirut Arab UniversityLebanon
| | - Nitsuh Dejene Ayele
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of Internal Medicine, Faculty of MedicineWolkite UniversityWolkiteEthiopia
| | - Isabelle Munyangaju
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Barcelona Institute for Global Health—Hospital ClínicUniversitat de Barcelona
| | - Fatima Esam Alzain
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineCollege of Medicine and General Surgery—Sudan University of Science and Technology
| | - Magda Wojtara
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| |
Collapse
|
35
|
Bai Y, Zhong H, Wang T, Lu ZJ. OligoFormer: an accurate and robust prediction method for siRNA design. Bioinformatics 2024; 40:btae577. [PMID: 39321261 PMCID: PMC11494384 DOI: 10.1093/bioinformatics/btae577] [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: 03/01/2024] [Revised: 08/14/2024] [Accepted: 09/23/2024] [Indexed: 09/27/2024] Open
Abstract
MOTIVATION RNA interference (RNAi) has become a widely used experimental approach for post-transcriptional regulation and is increasingly showing its potential as future targeted drugs. However, the prediction of highly efficient siRNAs (small interfering RNAs) is still hindered by dataset biases, the inadequacy of prediction methods, and the presence of off-target effects. To overcome these limitations, we propose an accurate and robust prediction method, OligoFormer, for siRNA design. RESULTS OligoFormer comprises three different modules including thermodynamic calculation, RNA-FM module, and Oligo encoder. Oligo encoder is the core module based on the transformer encoder. Taking siRNA and mRNA sequences as input, OligoFormer can obtain thermodynamic parameters, RNA-FM embedding, and Oligo embedding through these three modules, respectively. We carefully benchmarked OligoFormer against six comparable methods on siRNA efficacy datasets. OligoFormer outperforms all the other methods, with an average improvement of 9% in AUC, 6.6% in PRC, 9.8% in F1 score, and 5.1% in PCC compared to the best method among them in our inter-dataset validation. We also provide a comprehensive pipeline with prediction of siRNA efficacy and off-target effects using PITA score and TargetScan score. The ablation study shows RNA-FM module and thermodynamic parameters improved the performance and accelerated convergence of OligoFormer. The saliency maps by gradient backpropagation and base preference maps show certain base preferences in initial and terminal region of siRNAs. AVAILABILITY AND IMPLEMENTATION The source code of OligoFormer is freely available on GitHub at: https://github.com/lulab/OligoFormer. Docker image of OligoFormer is freely available on the docker hub at https://hub.docker.com/r/yilanbai/oligoformer.
Collapse
Affiliation(s)
- Yilan Bai
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Institute for Precision Medicine, Tsinghua University, Beijing, 100084, China
| | - Haochen Zhong
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Institute for Precision Medicine, Tsinghua University, Beijing, 100084, China
| | - Taiwei Wang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Institute for Precision Medicine, Tsinghua University, Beijing, 100084, China
- Academy for Advanced Interdisciplinary Studies (AAIS), and Peking University–Tsinghua University–National Institute of Biological Sciences Joint Graduate Program (PTN), Peking University, Beijing, 100871, China
| | - Zhi John Lu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Institute for Precision Medicine, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
36
|
Piombo E, Vetukuri RR, Konakalla NC, Kalyandurg PB, Sundararajan P, Jensen DF, Karlsson M, Dubey M. RNA silencing is a key regulatory mechanism in the biocontrol fungus Clonostachys rosea-wheat interactions. BMC Biol 2024; 22:219. [PMID: 39343898 PMCID: PMC11441109 DOI: 10.1186/s12915-024-02014-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Small RNA (sRNAs)- mediated RNA silencing is emerging as a key player in host-microbe interactions. However, its role in fungus-plant interactions relevant to biocontrol of plant diseases is yet to be explored. This study aimed to investigate Dicer (DCL)-mediated endogenous and cross-kingdom gene expression regulation in the biocontrol fungus Clonostachys rosea and wheat roots during interactions. RESULTS C. rosea Δdcl2 strain exhibited significantly higher root colonization than the WT, whereas no significant differences were observed for Δdcl1 strains. Dual RNA-seq revealed the upregulation of CAZymes, membrane transporters, and effector coding genes in C. rosea, whereas wheat roots responded with the upregulation of stress-related genes and the downregulation of growth-related genes. The expression of many of these genes was downregulated in wheat during the interaction with DCL deletion strains, underscoring the influence of fungal DCL genes on wheat defense response. sRNA sequencing identified 18 wheat miRNAs responsive to C. rosea, and three were predicted to target the C. rosea polyketide synthase gene pks29. Two of these miRNAs (mir_17532_x1 and mir_12061_x13) were observed to enter C. rosea from wheat roots with fluorescence analyses and to downregulate the expression of pks29, showing plausible cross-kingdom RNA silencing of the C. rosea gene by wheat miRNAs. CONCLUSIONS We provide insights into the mechanisms underlying the interaction between biocontrol fungi and plant roots. Moreover, the study sheds light on the role of sRNA-mediated gene expression regulation in C. rosea-wheat interactions and provides preliminary evidence of cross-kingdom RNA silencing between plants and biocontrol fungi.
Collapse
Affiliation(s)
- Edoardo Piombo
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Ramesh Raju Vetukuri
- Department of Plant Breeding, Swedish University of Agricultural Sciences, Lomma, Sweden
| | - Naga Charan Konakalla
- Department of Plant Breeding, Swedish University of Agricultural Sciences, Lomma, Sweden
| | - Pruthvi B Kalyandurg
- Department of Plant Breeding, Swedish University of Agricultural Sciences, Lomma, Sweden
| | - Poorva Sundararajan
- Department of Plant Breeding, Swedish University of Agricultural Sciences, Lomma, Sweden
| | - Dan Funck Jensen
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Magnus Karlsson
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Mukesh Dubey
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden.
| |
Collapse
|
37
|
Zhang J, Liu L, Wei X, Zhao C, Luo Y, Li J, Le TD. Scanning sample-specific miRNA regulation from bulk and single-cell RNA-sequencing data. BMC Biol 2024; 22:218. [PMID: 39334271 PMCID: PMC11438147 DOI: 10.1186/s12915-024-02020-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 09/24/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND RNA-sequencing technology provides an effective tool for understanding miRNA regulation in complex human diseases, including cancers. A large number of computational methods have been developed to make use of bulk and single-cell RNA-sequencing data to identify miRNA regulations at the resolution of multiple samples (i.e. group of cells or tissues). However, due to the heterogeneity of individual samples, there is a strong need to infer miRNA regulation specific to individual samples to uncover miRNA regulation at the single-sample resolution level. RESULTS Here, we develop a framework, Scan, for scanning sample-specific miRNA regulation. Since a single network inference method or strategy cannot perform well for all types of new data, Scan incorporates 27 network inference methods and two strategies to infer tissue-specific or cell-specific miRNA regulation from bulk or single-cell RNA-sequencing data. Results on bulk and single-cell RNA-sequencing data demonstrate the effectiveness of Scan in inferring sample-specific miRNA regulation. Moreover, we have found that incorporating the prior information of miRNA targets can generally improve the accuracy of miRNA target prediction. In addition, Scan can contribute to construct cell/tissue correlation networks and recover aggregate miRNA regulatory networks. Finally, the comparison results have shown that the performance of network inference methods is likely to be data-specific, and selecting optimal network inference methods is required for more accurate prediction of miRNA targets. CONCLUSIONS Scan provides a useful method to help infer sample-specific miRNA regulation for new data, benchmark new network inference methods and deepen the understanding of miRNA regulation at the resolution of individual samples.
Collapse
Affiliation(s)
- Junpeng Zhang
- School of Engineering, Dali University, Dali, 671003, Yunnan, China.
| | - Lin Liu
- UniSA STEM, University of South Australia, Mawson Lakes, SA, 5095, Australia
| | - Xuemei Wei
- School of Engineering, Dali University, Dali, 671003, Yunnan, China
| | - Chunwen Zhao
- School of Engineering, Dali University, Dali, 671003, Yunnan, China
| | - Yanbi Luo
- School of Engineering, Dali University, Dali, 671003, Yunnan, China
| | - Jiuyong Li
- UniSA STEM, University of South Australia, Mawson Lakes, SA, 5095, Australia
| | - Thuc Duy Le
- UniSA STEM, University of South Australia, Mawson Lakes, SA, 5095, Australia.
| |
Collapse
|
38
|
Tang L, Qiu H, Xu B, Su Y, Nyarige V, Li P, Chen H, Killham B, Liao J, Adam H, Yang A, Yu A, Jang M, Rubart M, Xie J, Zhu W. Microparticle Mediated Delivery of Apelin Improves Heart Function in Post Myocardial Infarction Mice. Circ Res 2024; 135:777-798. [PMID: 39145385 PMCID: PMC11392624 DOI: 10.1161/circresaha.124.324608] [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: 03/15/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Apelin is an endogenous prepropeptide that regulates cardiac homeostasis and various physiological processes. Intravenous injection has been shown to improve cardiac contractility in patients with heart failure. However, its short half-life prevents studying its impact on left ventricular remodeling in the long term. Here, we aim to study whether microparticle-mediated slow release of apelin improves heart function and left ventricular remodeling in mice with myocardial infarction (MI). METHODS A cardiac patch was fabricated by embedding apelin-containing microparticles in a fibrin gel scaffold. MI was induced via permanent ligation of the left anterior descending coronary artery in adult C57BL/6J mice followed by epicardial patch placement immediately after (acute MI) or 28 days (chronic MI) post-MI. Four groups were included in this study, namely sham, MI, MI plus empty microparticle-embedded patch treatment, and MI plus apelin-containing microparticle-embedded patch treatment. Cardiac function was assessed by transthoracic echocardiography. Cardiomyocyte morphology, apoptosis, and cardiac fibrosis were evaluated by histology. Cardioprotective pathways were determined by RNA sequencing, quantitative polymerase chain reaction, and Western blot. RESULTS The level of endogenous apelin was largely reduced in the first 7 days after MI induction and it was normalized by day 28. Apelin-13 encapsulated in poly(lactic-co-glycolic acid) microparticles displayed a sustained release pattern for up to 28 days. Treatment with apelin-containing microparticle-embedded patch inhibited cardiac hypertrophy and reduced scar size in both acute and chronic MI models, which is associated with improved cardiac function. Data from cellular and molecular analyses showed that apelin inhibits the activation and proliferation of cardiac fibroblasts by preventing transforming growth factor-β-mediated activation of Smad2/3 (supporessor of mothers against decapentaplegic 2/3) and downstream profibrotic gene expression. CONCLUSIONS Poly(lactic-co-glycolic acid) microparticles prolonged the apelin release time in the mouse hearts. Epicardial delivery of the apelin-containing microparticle-embedded patch protects mice from both acute and chronic MI-induced cardiac dysfunction, inhibits cardiac fibrosis, and improves left ventricular remodeling.
Collapse
Affiliation(s)
- Ling Tang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Huiliang Qiu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Bing Xu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Yajuan Su
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha (Y.S., J.X.)
| | - Verah Nyarige
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Pengsheng Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Houjia Chen
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Brady Killham
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Henderson Adam
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Aaron Yang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Alexander Yu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Michelle Jang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Michael Rubart
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (M.R.)
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha (Y.S., J.X.)
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| |
Collapse
|
39
|
Desideri F, Grazzi A, Lisi M, Setti A, Santini T, Colantoni A, Proietti G, Carvelli A, Tartaglia GG, Ballarino M, Bozzoni I. CyCoNP lncRNA establishes cis and trans RNA-RNA interactions to supervise neuron physiology. Nucleic Acids Res 2024; 52:9936-9952. [PMID: 38989616 PMCID: PMC11381359 DOI: 10.1093/nar/gkae590] [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: 11/06/2023] [Revised: 05/30/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024] Open
Abstract
The combination of morphogenetic and transcription factors together with the synergic aid of noncoding RNAs and their cognate RNA binding proteins contribute to shape motor neurons (MN) identity. Here, we extend the noncoding perspective of human MN, by detailing the molecular and biological activity of CyCoNP (as Cytoplasmic Coordinator of Neural Progenitors) a highly expressed and MN-enriched human lncRNA. Through in silico prediction, in vivo RNA purification and loss of function experiments followed by RNA-sequencing, we found that CyCoNP sustains a specific neuron differentiation program, required for the physiology of both neuroblastoma cells and hiPSC-derived MN, which mainly involves miR-4492 and NCAM1 mRNA. We propose a novel lncRNA-mediated 'dual mode' of action, in which CyCoNP acts in trans as a classical RNA sponge by sequestering miR-4492 from its pro-neuronal targets, including NCAM1 mRNA, and at the same time it plays an additional role in cis by interacting with NCAM1 mRNA and regulating the availability and localization of the miR-4492 in its proximity. These data highlight novel insights into the noncoding RNA-mediated control of human neuron physiology and point out the importance of lncRNA-mediated interactions for the spatial distribution of regulatory molecules.
Collapse
Affiliation(s)
- Fabio Desideri
- Center for Life Nano- & Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Alessandro Grazzi
- Center for Life Nano- & Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Michela Lisi
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Adriano Setti
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Tiziana Santini
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Alessio Colantoni
- Center for Life Nano- & Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Gabriele Proietti
- Centre for Human Technologies (CHT), Istituto Italiano di Tecnologia (IIT), 16152 Genova, Italy
| | - Andrea Carvelli
- Department of Neuroscience, The Scripps Research institute, La Jolla, CA 92037, USA
| | - Gian Gaetano Tartaglia
- Centre for Human Technologies (CHT), Istituto Italiano di Tecnologia (IIT), 16152 Genova, Italy
| | - Monica Ballarino
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Irene Bozzoni
- Center for Life Nano- & Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
40
|
Yu CQ, Wang XF, Li LP, You ZH, Ren ZH, Chu P, Guo F, Wang ZY. RBNE-CMI: An Efficient Method for Predicting circRNA-miRNA Interactions via Multiattribute Incomplete Heterogeneous Network Embedding. J Chem Inf Model 2024. [PMID: 39231016 DOI: 10.1021/acs.jcim.4c01118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
Circular RNA (circRNA)-microRNA (miRNA) interaction (CMI) plays crucial roles in cellular regulation, offering promising perspectives for disease diagnosis and therapy. Therefore, it is necessary to employ computational methods for the rapid and cost-effective prediction of potential circRNA-miRNA interactions. However, the existing methods are limited by incomplete data; therefore, it is difficult to model molecules with different attributes on a large scale, which greatly hinders the efficiency and performance of prediction. In this study, we propose an effective method for predicting circRNA-miRNA interactions, called RBNE-CMI, and introduce a framework that can embed incomplete multiattribute CMI heterogeneous networks. By combining the proposed method, we integrate different data sets in the CMI prediction field into one incomplete network for modeling, achieving superior performance in 5-fold cross-validation. Moreover, in the prediction task based on complete data, the proposed method still achieves better performance than the known model. In addition, in the case study, we successfully predicted 18 of the 20 potential cancer biomarkers. The data and source code can be found at https://github.com/1axin/RBNE-CMI.
Collapse
Affiliation(s)
- Chang-Qing Yu
- School of Information Engineering, Xijing University, Xi'an 710123 China
| | - Xin-Fei Wang
- College of Computer Science and Technology, Jilin University, Changchun 130012 China
| | - Li-Ping Li
- Yizhi School of Agriculture and Forestry, Xiangyang Polytechnic Institute, Xianyang 712000, China
| | - Zhu-Hong You
- School of Computer Science, Northwestern Polytechnical University, Xi'an 710072, China
| | - Zhong-Hao Ren
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Peng Chu
- School of Information Engineering, Xijing University, Xi'an 710123 China
| | - Feng Guo
- School of Information Engineering, Xijing University, Xi'an 710123 China
| | - Zhen-Yu Wang
- School of Telecommunications, Lanzhou University of Technology, Lanzhou 730000, China
| |
Collapse
|
41
|
Marturano G, Carli D, Cucini C, Carapelli A, Plazzi F, Frati F, Passamonti M, Nardi F. SmithHunter: a workflow for the identification of candidate smithRNAs and their targets. BMC Bioinformatics 2024; 25:286. [PMID: 39223476 PMCID: PMC11370224 DOI: 10.1186/s12859-024-05909-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND SmithRNAs (Small MITochondrial Highly-transcribed RNAs) are a novel class of small RNA molecules that are encoded in the mitochondrial genome and regulate the expression of nuclear transcripts. Initial evidence for their existence came from the Manila clam Ruditapes philippinarum, where they have been described and whose activity has been biologically validated through RNA injection experiments. Current evidence on the existence of these RNAs in other species is based only on small RNA sequencing. As a preliminary step to characterize smithRNAs across different metazoan lineages, a dedicated, unified, analytical workflow is needed. RESULTS We propose a novel workflow specifically designed for smithRNAs. Sequence data (from small RNA sequencing) uniquely mapping to the mitochondrial genome are clustered into putative smithRNAs and prefiltered based on their abundance, presence in replicate libraries and 5' and 3' transcription boundary conservation. The surviving sequences are subsequently compared to the untranslated regions of nuclear transcripts based on seed pairing, overall match and thermodynamic stability to identify possible targets. Ample collateral information and graphics are produced to help characterize these molecules in the species of choice and guide the operator through the analysis. The workflow was tested on the original Manila clam data. Under basic settings, the results of the original study are largely replicated. The effect of additional parameter customization (clustering threshold, stringency, minimum number of replicates, seed matching) was further evaluated. CONCLUSIONS The study of smithRNAs is still in its infancy and no dedicated analytical workflow is currently available. At its core, the SmithHunter workflow builds over the bioinformatic procedure originally applied to identify candidate smithRNAs in the Manila clam. In fact, this is currently the only evidence for smithRNAs that has been biologically validated and, therefore, the elective starting point for characterizing smithRNAs in other species. The original analysis was readapted using current software implementations and some minor issues were solved. Moreover, the workflow was improved by allowing the customization of different analytical parameters, mostly focusing on stringency and the possibility of accounting for a minimal level of genetic differentiation among samples.
Collapse
Affiliation(s)
| | - Diego Carli
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Via Selmi 3, 40126, Bologna, Italy
| | - Claudio Cucini
- Department of Life Sciences, University of Siena, 53100, Siena, Italy
| | - Antonio Carapelli
- Department of Life Sciences, University of Siena, 53100, Siena, Italy
- National Biodiversity Future Center (NBFC), 90133, Palermo, Italy
| | - Federico Plazzi
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Via Selmi 3, 40126, Bologna, Italy
| | - Francesco Frati
- Department of Life Sciences, University of Siena, 53100, Siena, Italy
- National Biodiversity Future Center (NBFC), 90133, Palermo, Italy
| | - Marco Passamonti
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Via Selmi 3, 40126, Bologna, Italy.
| | - Francesco Nardi
- Department of Life Sciences, University of Siena, 53100, Siena, Italy
- National Biodiversity Future Center (NBFC), 90133, Palermo, Italy
| |
Collapse
|
42
|
Lin CC, Law BF, Hettick JM. MicroRNA-mediated Krüppel-like factor 4 upregulation induces alternatively activated macrophage-associated marker and chemokine transcription in 4,4'-methylene diphenyl diisocyanate exposed macrophages. Xenobiotica 2024; 54:730-748. [PMID: 38568505 PMCID: PMC11489325 DOI: 10.1080/00498254.2024.2334329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/20/2024]
Abstract
1. Occupational exposure to 4,4'-methylene diphenyl diisocyanate (MDI) is associated with occupational asthma (OA) development. Alveolar macrophage-induced recruitment of immune cells to the lung microenvironment plays an important role during asthma pathogenesis. Previous studies identified that MDI/MDI-glutathione (GSH)-exposure downregulates endogenous hsa-miR-206-3p/hsa-miR-381-3p. Our prior report shows that alternatively activated (M2) macrophage-associated markers/chemokines are induced by MDI/MDI-GSH-mediated Krüppel-Like Factor 4 (KLF4) upregulation in macrophages and stimulates immune cell chemotaxis. However, the underlying molecular mechanism(s) by which MDI/MDI-GSH upregulates KLF4 remain unclear. 2. Following MDI-GSH exposure, microRNA(miR)-inhibitors/mimics or plasmid transfection, endogenous hsa-miR-206-3p/hsa-miR-381-3p, KLF4, or M2 macrophage-associated markers (CD206, TGM2), and chemokines (CCL17, CCL22, CCL24) were measured by either RT-qPCR, western blot, or luciferase assay. 3. MDI-GSH exposure downregulates hsa-miR-206-3p/hsa-miR-381-3p by 1.46- to 9.75-fold whereas upregulates KLF4 by 1.68- to 1.99-fold, respectively. In silico analysis predicts binding between hsa-miR-206-3p/hsa-miR-381-3p and KLF4. Gain- and loss-of-function, luciferase reporter assays and RNA-induced silencing complex-immunoprecipitation (RISC-IP) studies confirm the posttranscriptional regulatory roles of hsa-miR-206-3p/hsa-miR-381-3p and KLF4 in macrophages. Furthermore, hsa-miR-206-3p/hsa-miR-381-3p regulate the expression of M2 macrophage-associated markers and chemokines via KLF4. 4. In conclusion, hsa-miR-206-3p/hsa-miR-381-3p play a major role in regulation of MDI/MDI-GSH-induced M2 macrophage-associated markers and chemokines by targeting the KLF4 transcript, and KLF4-mediated regulation in macrophages.
Collapse
Affiliation(s)
- Chen-Chung Lin
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505
| | - Brandon F. Law
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505
| | - Justin M. Hettick
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505
| |
Collapse
|
43
|
Zhou B, Zheng Y, Suo Z, Zhang M, Xu W, Wang L, Ge D, Qu Y, Wang Q, Zheng H, Ni C. The role of lncRNAs related ceRNA regulatory network in multiple hippocampal pathological processes during the development of perioperative neurocognitive disorders. PeerJ 2024; 12:e17775. [PMID: 39135955 PMCID: PMC11318589 DOI: 10.7717/peerj.17775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/28/2024] [Indexed: 08/15/2024] Open
Abstract
Background Perioperative neurocognitive disorders (PND) refer to neurocognitive abnormalities during perioperative period, which are a great challenge for elderly patients and associated with increased morbidity and mortality. Our studies showed that long non-coding RNAs (lncRNAs) regulate mitochondrial function and aging-related pathologies in the aged hippocampus after anesthesia, and lncRNAs are associated with multiple neurodegenerations. However, the regulatory role of lncRNAs in PND-related pathological processes remains unclear. Methods A total of 18-month mice were assigned to control and surgery (PND) groups, mice in PND group received sevoflurane anesthesia and laparotomy. Cognitive function was assessed with fear conditioning test. Hippocampal RNAs were isolated for sequencing, lncRNA and microRNA libraries were constructed, mRNAs were identified, Gene Ontology (GO) analysis were performed, and lncRNA-microRNA-mRNA networks were established. qPCR was performed for gene expression verification. Results A total of 312 differentially expressed (DE) lncRNAs, 340 DE-Transcripts of Uncertain Coding Potential (TUCPs), and 2,003 DEmRNAs were identified in the hippocampus between groups. The lncRNA-microRNA-mRNA competing endogenous RNA (ceRNA) network was constructed with 29 DElncRNAs, 90 microRNAs, 493 DEmRNAs, 148 lncRNA-microRNA interaction pairs, 794 microRNA-mRNA interaction pairs, and 110 lncRNA-mRNA co-expression pairs. 795 GO terms were obtained. Based on the frequencies of involved pathological processes, BP terms were divided into eight categories: neurological system alternation, neuronal development, metabolism alternation, immunity and neuroinflammation, apoptosis and autophagy, cellular communication, molecular modification, and behavior changes. LncRNA-microRNA-mRNA ceRNA networks in these pathological categories were constructed, and involved pathways and targeted genes were revealed. The top relevant lncRNAs in these ceRNA networks included RP23-65G6.4, RP24-396L14.1, RP23-251I16.2, XLOC_113622, RP24-496E14.1, etc., and the top relevant mRNAs in these ceRNA networks included Dlg4 (synaptic function), Avp (lipophagy), Islr2 (synaptic function), Hcrt (regulation of awake behavior), Tnc (neurotransmitter uptake). Conclusion In summary, we have constructed the lncRNA-associated ceRNA network during PND development in mice, explored the role of lncRNAs in multiple pathological processes in the mouse hippocampus, and provided insights into the potential mechanisms and therapeutic gene targets for PND.
Collapse
Affiliation(s)
- Bowen Zhou
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuxiang Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zizheng Suo
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mingzhu Zhang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenjie Xu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lijuan Wang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dazhuang Ge
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yinyin Qu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Qiang Wang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cheng Ni
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
44
|
Böge FL, Ruff S, Hemandhar Kumar S, Selle M, Becker S, Jung K. Combined Analysis of Multi-Study miRNA and mRNA Expression Data Shows Overlap of Selected miRNAs Involved in West Nile Virus Infections. Genes (Basel) 2024; 15:1030. [PMID: 39202390 PMCID: PMC11353516 DOI: 10.3390/genes15081030] [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: 05/31/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
The emerging zoonotic West Nile virus (WNV) has serious impact on public health. Thus, understanding the molecular basis of WNV infections in mammalian hosts is important to develop improved diagnostic and treatment strategies. In this context, the role of microRNAs (miRNAs) has been analyzed by several studies under different conditions and with different outcomes. A systematic comparison is therefore necessary. Furthermore, additional information from mRNA target expression data has rarely been taken into account to understand miRNA expression profiles under WNV infections. We conducted a meta-analysis of publicly available miRNA expression data from multiple independent studies, and analyzed them in a harmonized way to increase comparability. In addition, we used gene-set tests on mRNA target expression data to further gain evidence about differentially expressed miRNAs. For this purpose, we also studied the use of target information from different databases. We detected a substantial number of miRNA that emerged as differentially expressed from several miRNA datasets, and from the mRNA target data analysis as well. When using mRNA target data, we found that the targetscan databases provided the most useful information. We demonstrated improved miRNA detection through research synthesis of multiple independent miRNA datasets coupled with mRNA target set testing, leading to the discovery of multiple miRNAs which should be taken into account for further research on the molecular mechanism of WNV infections.
Collapse
Affiliation(s)
- Franz Leonard Böge
- Institute of Animal Genomics, University of Veterinary Medicine Hannover, Bünteweg 17p, 30559 Hannover, Germany; (F.L.B.); (S.R.); (S.H.K.); (M.S.)
| | - Sergej Ruff
- Institute of Animal Genomics, University of Veterinary Medicine Hannover, Bünteweg 17p, 30559 Hannover, Germany; (F.L.B.); (S.R.); (S.H.K.); (M.S.)
| | - Shamini Hemandhar Kumar
- Institute of Animal Genomics, University of Veterinary Medicine Hannover, Bünteweg 17p, 30559 Hannover, Germany; (F.L.B.); (S.R.); (S.H.K.); (M.S.)
| | - Michael Selle
- Institute of Animal Genomics, University of Veterinary Medicine Hannover, Bünteweg 17p, 30559 Hannover, Germany; (F.L.B.); (S.R.); (S.H.K.); (M.S.)
| | - Stefanie Becker
- Institute of Parasitology, University of Veterinary Medicine Hannover, Bünteweg 17, 30539 Hannover, Germany;
| | - Klaus Jung
- Institute of Animal Genomics, University of Veterinary Medicine Hannover, Bünteweg 17p, 30559 Hannover, Germany; (F.L.B.); (S.R.); (S.H.K.); (M.S.)
| |
Collapse
|
45
|
Chen J, Liu K, Vadas MA, Gamble JR, McCaughan GW. The Role of the MiR-181 Family in Hepatocellular Carcinoma. Cells 2024; 13:1289. [PMID: 39120319 PMCID: PMC11311592 DOI: 10.3390/cells13151289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the fourth-leading cause of cancer-related death worldwide. Due to the high mortality rate in HCC patients, discovering and developing novel systemic treatment options for HCC is a vital unmet medical need. Among the numerous molecular alterations in HCCs, microRNAs (miRNAs) have been increasingly recognised to play critical roles in hepatocarcinogenesis. We and others have recently revealed that members of the microRNA-181 (miR-181) family were up-regulated in some, though not all, human cirrhotic and HCC tissues-this up-regulation induced epithelial-mesenchymal transition (EMT) in hepatocytes and tumour cells, promoting HCC progression. MiR-181s play crucial roles in governing the fate and function of various cells, such as endothelial cells, immune cells, and tumour cells. Previous reviews have extensively covered these aspects in detail. This review aims to give some insights into miR-181s, their targets and roles in modulating signal transduction pathways, factors regulating miR-181 expression and function, and their roles in HCC.
Collapse
Affiliation(s)
- Jinbiao Chen
- Liver Injury and Cancer Program, Cancer Innovations Centre, Centenary Institute, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia;
| | - Ken Liu
- Liver Injury and Cancer Program, Cancer Innovations Centre, Centenary Institute, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia;
- Royal Prince Alfred Hospital, Missenden Road, Camperdown, NSW 2050, Australia
| | - Mathew A. Vadas
- Vascular Biology Program, Healthy Ageing Centre, Centenary Institute, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia; (M.A.V.); (J.R.G.)
| | - Jennifer R. Gamble
- Vascular Biology Program, Healthy Ageing Centre, Centenary Institute, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia; (M.A.V.); (J.R.G.)
| | - Geoffrey W. McCaughan
- Liver Injury and Cancer Program, Cancer Innovations Centre, Centenary Institute, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia;
- Royal Prince Alfred Hospital, Missenden Road, Camperdown, NSW 2050, Australia
| |
Collapse
|
46
|
Qiu Y, Wang C, Wang J, L. V. Q, Sun L, Yang Y, Liu M, Liu X, Li C, Tang B. Revealing the dynamic whole transcriptome landscape of Clonorchis sinensis: Insights into the regulatory roles of noncoding RNAs and microtubule-related genes in development. PLoS Negl Trop Dis 2024; 18:e0012311. [PMID: 38991028 PMCID: PMC11265684 DOI: 10.1371/journal.pntd.0012311] [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: 01/11/2024] [Revised: 07/23/2024] [Accepted: 06/23/2024] [Indexed: 07/13/2024] Open
Abstract
Clonorchis sinensis is a significant zoonotic food-borne parasite that causes a range of hepatobiliary diseases, which in severe cases can even lead to cholangiocarcinoma. To explore new diagnostic and treatment strategies, the dynamic RNA regulatory processes across different developmental stages of C. sinensis were analyzed by using whole-transcriptome sequencing. The chromosomal-level genome of C. sinensis was used for sequence alignment and annotation. In this study, we identified a total of 59,103 RNAs in the whole genome, including 2,384 miRNAs, 25,459 mRNAs, 27,564 lncRNAs and 3,696 circRNAs. Differential expression analysis identified 6,556 differentially expressed mRNAs, 2,231 lncRNAs, 877 miRNAs and 20 circRNAs at different developmental stages. Functional enrichment analysis highlighted the critical role of microtubule-related biological processes in the growth and development of C. sinensis. And coexpression analysis revealed 97 lncRNAs and 85 circRNAs that were coexpressed with 42 differentially expressed mRNAs that associated with microtubules at different developmental stages of C. sinensis. The expression of the microtubule-related genes dynein light chain 2 (DLC2) and dynein light chain 4 (DLC4) increased with C. sinensis development, and DLC2/4 could be inhibited by albendazole. Finally, by constructing competing endogenous RNA (ceRNA) networks, the lncRNA-miRNA-mRNA and circRNA-miRNA-mRNA regulatory relationships were constructed, and the ceRNA networks of MSTRG.14258.5-novel_miR_2287-newGene_28215 and MSTRG.14258.5-novel_miR_2216-CSKR_109340 were verified. This study suggests, through whole transcriptome sequencing, that the context of microtubule regulation may play an essential role in the development and growth of C. sinensis.
Collapse
Affiliation(s)
- Yangyuan Qiu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Cunzhou Wang
- Jiashi County Hospital of Uygur Medicine, Kashgar City, Xinjiang Uygur Autonomous Region, PR China
| | - Jing Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Qingbo L. V.
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Lulu Sun
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Yaming Yang
- Yunnan Institute of Parasitic Diseases, Pu’er, PR China
| | - Mingyuan Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, PR China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, PR China
| | - Xiaolei Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Chen Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, PR China
| | - Bin Tang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, PR China
| |
Collapse
|
47
|
Han HL, Li JM, Chen D, Zhai XD, Smagghe G, Jiang H, Wang JJ, Wei D. Overexpression of miR-927-5p suppresses stalky expression and negatively reduces the spermatid production in Zeugodacus cucurbitae. PEST MANAGEMENT SCIENCE 2024; 80:3412-3422. [PMID: 38407521 DOI: 10.1002/ps.8044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND The melon fly, Zeugodacus cucurbitae Coquillett, is one of the major pests attacking Cucurbitaceae crops. Identifying critical genes or proteins regulating fertility is essential for sustainable pest control and a research hotspot in insect physiology. MicroRNAs (miRNAs) are short RNAs that do not directly participate in protein translation, but instead function in post-transcriptional regulation of gene expression involved in male fertility. RESULTS We found that miR-927-5p is highly expressed in the testes and investigated its function in spermatogenesis in Z. cucurbitae. Fluorescence in situ hybridization (FISH) showed miR-927-5p in the transformation and maturation region of the testis, and overexpression of miR-927-5p reduced the number of sperms by 53%. In continuation, we predicted 12 target genes of miR-927-5p using bioinformatics combined with transcriptome sequencing data, and found that miR-927-5p targets the new gene Stalky in insects, which was validated by quantitative real-time PCR, RNA pull-down and dual luciferase reporter assays. FISH also confirmed the co-localization of miR-927-5p and the transcript Stalky_1 in the testis. Moreover, silencing of Stalky_1 by RNA interference reduced the number of sperms by 32% and reduced sperm viability by 39% in physiologically mature male adults. Meanwhile, the silencing of Stalky_1 also resulted in low hatchability. CONCLUSION Our work not only presents a new, so far unreported mechanism regulating spermatogenesis by miR-927-5p targeting a new unknown target, Stalky, which is providing new knowledge on the regulatory network of insect spermatogenesis, but also lays a foundation for the development of SIT against important tephritid fly pests. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Hong-Liang Han
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| | - Jing-Ming Li
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| | - Dong Chen
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| | - Xiao-Di Zhai
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| | - Guy Smagghe
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
- Institute of Entomology, Guizhou University, Guiyang, China
| | - Hongbo Jiang
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| | - Jin-Jun Wang
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| | - Dong Wei
- Chongqing Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- International Joint Laboratory of China-Belgium on Sustainable Crop Pest Control, Academy of Agricultural Sciences, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
| |
Collapse
|
48
|
Menzies JAC, Maia Chagas A, Baden T, Alonso CR. A microRNA that controls the emergence of embryonic movement. eLife 2024; 13:RP95209. [PMID: 38869942 PMCID: PMC11175612 DOI: 10.7554/elife.95209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
Movement is a key feature of animal systems, yet its embryonic origins are not fully understood. Here, we investigate the genetic basis underlying the embryonic onset of movement in Drosophila focusing on the role played by small non-coding RNAs (microRNAs, miRNAs). To this end, we first develop a quantitative behavioural pipeline capable of tracking embryonic movement in large populations of fly embryos, and using this system, discover that the Drosophila miRNA miR-2b-1 plays a role in the emergence of movement. Through the combination of spectral analysis of embryonic motor patterns, cell sorting and RNA in situs, genetic reconstitution tests, and neural optical imaging we define that miR-2b-1 influences the emergence of embryonic movement by exerting actions in the developing nervous system. Furthermore, through the combination of bioinformatics coupled to genetic manipulation of miRNA expression and phenocopy tests we identify a previously uncharacterised (but evolutionarily conserved) chloride channel encoding gene - which we term Movement Modulator (Motor) - as a genetic target that mechanistically links miR-2b-1 to the onset of movement. Cell-specific genetic reconstitution of miR-2b-1 expression in a null miRNA mutant background, followed by behavioural assays and target gene analyses, suggest that miR-2b-1 affects the emergence of movement through effects in sensory elements of the embryonic circuitry, rather than in the motor domain. Our work thus reports the first miRNA system capable of regulating embryonic movement, suggesting that other miRNAs are likely to play a role in this key developmental process in Drosophila as well as in other species.
Collapse
Affiliation(s)
- Jonathan AC Menzies
- Department of Neuroscience, Sussex Neuroscience, School of Life Sciences, University of SussexBrightonUnited Kingdom
| | - André Maia Chagas
- Department of Neuroscience, Sussex Neuroscience, School of Life Sciences, University of SussexBrightonUnited Kingdom
| | - Tom Baden
- Department of Neuroscience, Sussex Neuroscience, School of Life Sciences, University of SussexBrightonUnited Kingdom
| | - Claudio R Alonso
- Department of Neuroscience, Sussex Neuroscience, School of Life Sciences, University of SussexBrightonUnited Kingdom
| |
Collapse
|
49
|
Chang WF, Huang PW, Li CL, Huang HS, Chou TY, Liao EC, Yu SJ. Radiocontrast medium induces histamine release in association with upregulation of miR‑19a‑3p and miR‑362‑3p expression. Biomed Rep 2024; 20:93. [PMID: 38765857 PMCID: PMC11099600 DOI: 10.3892/br.2024.1780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 03/21/2024] [Indexed: 05/22/2024] Open
Abstract
In Taiwan, the use of radiocontrast medium for clinical image diagnosis recently surpassed one million times and the overall prevalence of radiocontrast hypersensitivity was ~7%. A microRNA (miRNA/miRs) is a small non-coding RNA molecule that mostly plays a suppressor role in cells. However, the roles of miRNA expression in radiocontrast-induced mast cells activation remains to be elucidated. The aim of the present study was to investigate the role of miRNA on radiocontrast-induced mast cell activation. Computed tomography radiocontrast, ultravist and mouse mast cell line, P815, were used in the present study. Cell viability was detected by CCK-8 experiment. Levels of histamine and β-hexosaminidase were measured by ELISA. miRNA expression was detected by miRNA sequencing and reverse transcription-quantitative PCR. The results showed that ultravist could increase histamine release and reduce intracellular β-hexosaminidase levels of mast cells. A total of 102 miRNAs could be significantly upregulated by ultravist stimulation. Selected candidate miRNAs for the validation included miR-19a-3p and miR-362-3p which were also increased expression following stimulation with ultravist. In conclusion, ultravist could induce mast cell activation through upregulation of miR-19a-3p and miR-362-3p. Thus, miR-19a-3p and miR-362-3p could be promising candidates for development as novel targets for preventing radiocontrast-induced allergy in the future.
Collapse
Affiliation(s)
- Wei-Fang Chang
- Department of Radiology and Nuclear Medicine, Zuoying Armed Forces General Hospital, Kaohsiung 813, Taiwan, R.O.C
- Department of Radiology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan, R.O.C
| | - Po-Wei Huang
- Division of Urology, Department of Surgery, Zuoying Armed Forces General Hospital, Kaohsiung 813, Taiwan, R.O.C
| | - Chia-Ling Li
- Children's Medical Center, Taichung Veterans General Hospital, Taichung 407, Taiwan, R.O.C
| | - Hung-Sen Huang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan, R.O.C
| | - Ting-Yu Chou
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan, R.O.C
| | - En-Chih Liao
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan, R.O.C
- Department of Institute of Biomedical Sciences, MacKay Medical College, New Taipei City 252, Taiwan, R.O.C
| | - Sheng-Jie Yu
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan, R.O.C
- Institute of Biomedical Sciences, College of Life Sciences, National Chung Hsing University, Taichung 407, Taiwan, R.O.C
| |
Collapse
|
50
|
Yadav P, Tamilselvan R, Mani H, Singh KK. MicroRNA-mediated regulation of nonsense-mediated mRNA decay factors: Insights into microRNA prediction tools and profiling techniques. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2024; 1867:195022. [PMID: 38437914 DOI: 10.1016/j.bbagrm.2024.195022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 03/06/2024]
Abstract
Nonsense-mediated mRNA decay (NMD) stands out as a prominent RNA surveillance mechanism within eukaryotes, meticulously overseeing both RNA abundance and integrity by eliminating aberrant transcripts. These defective transcripts are discerned through the concerted efforts of translating ribosomes, eukaryotic release factors (eRFs), and trans-acting NMD factors, with Up-Frameshift 3 (UPF3) serving as a noteworthy component. Remarkably, in humans, UPF3 exists in two paralogous forms, UPF3A (UPF3) and UPF3B (UPF3X). Beyond its role in quality control, UPF3 wields significant influence over critical cellular processes, including neural development, synaptic plasticity, and axon guidance. However, the precise regulatory mechanisms governing UPF3 remain elusive. MicroRNAs (miRNAs) emerge as pivotal post-transcriptional gene regulators, exerting substantial impact on diverse pathological and physiological pathways. This comprehensive review encapsulates our current understanding of the intricate regulatory nexus between NMD and miRNAs, with particular emphasis on the essential role played by UPF3B in neurodevelopment. Additionally, we bring out the significance of the 3'-untranslated region (3'-UTR) as the molecular bridge connecting NMD and miRNA-mediated gene regulation. Furthermore, we provide an in-depth exploration of diverse computational tools tailored for the prediction of potential miRNA targets. To complement these computational approaches, we delineate experimental techniques designed to validate predicted miRNA-mRNA interactions, empowering readers with the knowledge necessary to select the most appropriate methodology for their specific research objectives.
Collapse
Affiliation(s)
- Priyanka Yadav
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Raja Tamilselvan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Harita Mani
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Kusum Kumari Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|