Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (959)

Search Parameters:
Keywords = small regulatory RNAs

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 672 KB  
Review
Saying “Yes” to NONO: A Therapeutic Target for Neuroblastoma and Beyond
by Sofya S. Pogodaeva, Olga O. Miletina, Nadezhda V. Antipova, Alexander A. Shtil and Oleg A. Kuchur
Cancers 2025, 17(19), 3228; https://doi.org/10.3390/cancers17193228 - 3 Oct 2025
Abstract
Pediatric tumors such as neuroblastoma are characterized by a genome-wide ‘transcriptional burden’, surmising the involvement of multiple alterations of gene expression. Search for master regulators of transcription whose inactivation is lethal for tumor cells identified the non-POU domain-containing octamer-binding protein (NONO), a member [...] Read more.
Pediatric tumors such as neuroblastoma are characterized by a genome-wide ‘transcriptional burden’, surmising the involvement of multiple alterations of gene expression. Search for master regulators of transcription whose inactivation is lethal for tumor cells identified the non-POU domain-containing octamer-binding protein (NONO), a member of the Drosophila Behavior/Human Splicing family known for the ability to form complexes with macromolecules. NONO emerges as an essential mechanism in normal neurogenesis as well as in tumor biology. In particular, NONO interactions with RNAs, largely with long non-coding MYCN transcripts, have been attributed to the aggressiveness of neuroblastoma. Broadening its significance beyond MYCN regulation, NONO guards a subset of transcription factors that comprise a core regulatory circuit, a self-sustained loop that maintains transcription. As a component of protein–protein complexes, NONO has been implicated in the control of cell cycle progression, double-strand DNA repair, and, generally, in cell survival. Altogether, the pro-oncogenic roles of NONO justify the need for its inactivation as a therapeutic strategy. However, considering NONO as a therapeutic target, its druggability is a challenge. Recent advances in the inactivation of NONO and downstream signaling with small molecular weight compounds make promising the development of pharmacological antagonists of NONO pathway(s) for neuroblastoma treatment. Full article
(This article belongs to the Special Issue Precision Medicine and Targeted Therapies in Neuroblastoma)
27 pages, 1191 KB  
Review
Small RNA and Epigenetic Control of Plant Immunity
by Sopan Ganpatrao Wagh, Akshay Milind Patil, Ghanshyam Bhaurao Patil, Sumeet Prabhakar Mankar, Khushboo Rastogi and Masamichi Nishiguchi
DNA 2025, 5(4), 47; https://doi.org/10.3390/dna5040047 - 1 Oct 2025
Abstract
Plants have evolved a complex, multilayered immune system that integrates molecular recognition, signaling pathways, epigenetic regulation, and small RNA-mediated control. Recent studies have shown that DNA-level regulatory mechanisms, such as RNA-directed DNA methylation (RdDM), histone modifications, and chromatin remodeling, are critical for modulating [...] Read more.
Plants have evolved a complex, multilayered immune system that integrates molecular recognition, signaling pathways, epigenetic regulation, and small RNA-mediated control. Recent studies have shown that DNA-level regulatory mechanisms, such as RNA-directed DNA methylation (RdDM), histone modifications, and chromatin remodeling, are critical for modulating immune gene expression, allowing for rapid and accurate pathogen-defense responses. The epigenetic landscape not only maintains immunological homeostasis but also promotes stress-responsive transcription via stable chromatin modifications. These changes contribute to immunological priming, a process in which earlier exposure to pathogens or abiotic stress causes a heightened state of preparedness for future encounters. Small RNAs, including siRNAs, miRNAs, and phasiRNAs, are essential for gene silencing before and after transcription, fine-tuning immune responses, and inhibiting negative regulators. These RNA molecules interact closely with chromatin features, influencing histone acetylation/methylation (e.g., H3K4me3, H3K27me3) and guiding DNA methylation patterns. Epigenetically encoded immune memory can be stable across multiple generations, resulting in the transgenerational inheritance of stress resilience. Such memory effects have been observed in rice, tomato, maize, and Arabidopsis. This review summarizes new findings on short RNA biology, chromatin-level immunological control, and epigenetic memory in plant defense. Emerging technologies, such as ATAC-seq (Assay for Transposase-Accessible Chromatin using Sequencing), ChIP-seq (Chromatin Immunoprecipitation followed by Sequencing), bisulfite sequencing, and CRISPR/dCas9-based epigenome editing, are helping researchers comprehend these pathways. These developments hold an opportunity for establishing epigenetic breeding strategies that target the production of non-GMO, stress-resistant crops for sustainable agriculture. Full article
Show Figures

Figure 1

16 pages, 1149 KB  
Review
Beyond Genes: Non-Canonical Mechanisms Driving Antimicrobial Resistance in Bacteria
by Leonard Koolman, Chijioke Emenike, Debasis Mitra and Sourav Chattaraj
Bacteria 2025, 4(4), 50; https://doi.org/10.3390/bacteria4040050 - 1 Oct 2025
Abstract
Antimicrobial resistance (AMR) is traditionally discussed in the context of horizontally acquired resistance genes and point mutations at target loci. However, this gene-centred model fails to account for a large number of clinically important modalities of resistance. There is now substantial evidence implicating [...] Read more.
Antimicrobial resistance (AMR) is traditionally discussed in the context of horizontally acquired resistance genes and point mutations at target loci. However, this gene-centred model fails to account for a large number of clinically important modalities of resistance. There is now substantial evidence implicating bacteria in the ability to escape the effects of antibiotics in a variety of non-canonical ways, which are not considered in traditional diagnostic and surveillance pipelines. Among these factors, we can list those arising from global regulatory networks, phase variability, epigenetic tuning, small RNAs, genome structural variability, and phenotypic states like tolerance and persistence. This review will blend the current knowledge on these alternative pathways of resistance and underscore how they intersect with canonical genetic determinants. We will highlight cases where resistance emerges in the absence of known resistance genes, analyse the role of regulatory plasticity in efflux pump expression and membrane remodelling, and examine the contributions of bacterial stress responses and post-transcriptional control. Additionally, we will address methodological gaps in the detection of these mechanisms and their implications for clinical treatment failure, resistance surveillance, and drug development. By integrating insights from molecular microbiology, systems biology, and genomics, this review aims to offer a framework for understanding AMR as a multifaceted, context-dependent phenotype, not merely a genotype. We conclude by identifying knowledge gaps and suggesting priorities for research and diagnostic innovation in this evolving field. Full article
Show Figures

Figure 1

30 pages, 1900 KB  
Review
The Life of MicroRNAs: Biogenesis, Function and Decay in Cancer
by Shuang Ding and Pingping Wang
Biomolecules 2025, 15(10), 1393; https://doi.org/10.3390/biom15101393 - 30 Sep 2025
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that play pivotal roles in post-transcriptional gene regulation, influencing development, differentiation, and disease pathogenesis. Since their discovery in 1993, miRNAs have been recognized for their evolutionary conservation and pleiotropic effects, with the 2024 Nobel Prize underscoring their [...] Read more.
MicroRNAs (miRNAs) are small non-coding RNAs that play pivotal roles in post-transcriptional gene regulation, influencing development, differentiation, and disease pathogenesis. Since their discovery in 1993, miRNAs have been recognized for their evolutionary conservation and pleiotropic effects, with the 2024 Nobel Prize underscoring their significance in post-transcriptional regulation via the RNA interference (RNAi) pathway. This review synthesizes the complete life cycle of miRNAs—from transcription and processing to function and decay—emphasizing regulatory mechanisms and their implications in human diseases, particularly cancer. We discuss how epitranscriptomic modifications influence miRNA biogenesis and activity, explore their nuclear and mitochondrial functions, and address emerging challenges in miRNA-based therapeutics, including the expanding small RNA landscape such as tRNA-derived small RNAs (tsRNAs), and Argonaute (AGO)-independent activities. Despite hurdles such as modest multi-target effects, off-target interactions, and delivery challenges, miRNAs remain promising as both biomarkers and therapeutic agents, underscoring the need for sustained research to bridge preclinical insights with clinical applications. Full article
(This article belongs to the Special Issue Molecular Signalling Pathways in Tumorigenesis and Tumor Suppression)
Show Figures

Figure 1

23 pages, 748 KB  
Review
Polyamine Induction of Secondary Metabolite Biosynthetic Genes in Fungi Is Mediated by Global Regulator LaeA and α-NAC Transcriptional Coactivator: Connection to Epigenetic Modification of Histones
by Juan F. Martín
Molecules 2025, 30(19), 3903; https://doi.org/10.3390/molecules30193903 - 27 Sep 2025
Abstract
Polyamines are polycationic compounds present in all living cells that exert functions at different levels in the metabolism. They bind to DNA and RNA and modulate DNA replication and gene expression. Some of these regulatory effects are exerted by promoting condensation of nucleosomes, [...] Read more.
Polyamines are polycationic compounds present in all living cells that exert functions at different levels in the metabolism. They bind to DNA and RNA and modulate DNA replication and gene expression. Some of these regulatory effects are exerted by promoting condensation of nucleosomes, a mechanism closely connected with epigenetic modification by histone methylation and acetylation. The polyamines 1,3-diaminopropane and spermidine induce expression of the global regulator LaeA and increase by several folds the formation of the α-NAC transcriptional co-activator, a subunit of the nascent polypeptide-associated complex. The global regulator LaeA controls the switch from primary growth to secondary metabolite production and differentiation when an essential nutrient in the growth medium becomes limiting. α-NAC exerts significant control over the biosynthesis of secondary metabolites and fungal pathogenicity on plants. When purified α-NAC protein is added to a tomato host plant, it induces plant resistance to fungal infections and triggers the development of system-acquired resistance in other plants. Spermidine extends the life of yeast cells and prolongs the half-life of penicillin gene transcripts in Penicillium chrysogenum. This article discusses advances in the basis of understanding the mechanism of plant–fungi interaction and the effect of small fungal metabolites and epigenetic modifiers in this interaction. Full article
(This article belongs to the Special Issue Natural Products Biosynthesis: Present and Perspectives)
Show Figures

Figure 1

30 pages, 680 KB  
Review
The Hidden Regulators: MicroRNAs in Pediatric Heart Development and Disease
by Adam Kozik, Michał Piotrowski, Julia Izabela Karpierz, Mariusz Kowalewski and Jakub Batko
J. Clin. Med. 2025, 14(19), 6833; https://doi.org/10.3390/jcm14196833 - 26 Sep 2025
Abstract
The development and function of the heart are governed by a highly coordinated network of regulatory mechanisms, among which miRNAs play a central role. These small, non-coding molecules modulate gene expression predominantly through mRNA degradation. This narrative review aims to summarize current knowledge [...] Read more.
The development and function of the heart are governed by a highly coordinated network of regulatory mechanisms, among which miRNAs play a central role. These small, non-coding molecules modulate gene expression predominantly through mRNA degradation. This narrative review aims to summarize current knowledge about biogenesis, its impact on heart development and function, and its clinical implications in pediatric cardiology. We discuss how specific miRNAs contribute to shaping the normal heart and influencing the pathogenesis of congenital malformations. Furthermore, we review disease-specific miRNA signatures identified in the most common congenital heart defects and some acquired diseases, including hypoplastic left heart syndrome (HLHS), tetralogy of Fallot (TOF), bicuspid aortic valve (BAV), septation defects, cardiomyopathies, arrhythmias, and myocarditis. Many studies indicate that circulating and tissue miRNAs can become non-invasive biomarkers for early diagnosis and disease monitoring. Experimental data suggest their potential use in treatment despite many delivery and safety challenges. However, further research is necessary to fully exploit the potential of miRNAs and effectively translate these findings into clinical practice in pediatric cardiology. Full article
(This article belongs to the Section Cardiology)
Show Figures

Figure 1

17 pages, 4323 KB  
Article
Dysregulation of miRNAs in Sicilian Patients with Huntington’s Disease
by Michele Salemi, Francesca Antonia Schillaci, Maria Grazia Salluzzo, Giovanna Marchese, Giovanna Maria Ventola, Concetta Simona Perrotta, Vincenzo Di Stefano, Giuseppe Lanza and Raffaele Ferri
Diagnostics 2025, 15(19), 2454; https://doi.org/10.3390/diagnostics15192454 - 25 Sep 2025
Abstract
Background/Objectives: Huntington’s disease (HD) is an autosomal dominant neurodegenerative disorder caused by a CAG nucleotide repeat expansion in the Huntingtin (HTT) gene. Dysregulation of microRNAs (miRNAs), key post-transcriptional regulators of gene expression, has been implicated in HD pathogenesis, although their [...] Read more.
Background/Objectives: Huntington’s disease (HD) is an autosomal dominant neurodegenerative disorder caused by a CAG nucleotide repeat expansion in the Huntingtin (HTT) gene. Dysregulation of microRNAs (miRNAs), key post-transcriptional regulators of gene expression, has been implicated in HD pathogenesis, although their specific roles remain incompletely understood. Methods: Peripheral blood mononuclear cells from Sicilian HD patients and matched healthy controls were subjected to small RNA sequencing. Differential expression analysis was conducted using DESeq2 (version 1.44.0), with significance defined as |fold change| ≥ 1.5 and adjusted p ≤ 0.05. Ingenuity Pathway Analysis (IPA) was applied to assess functional enrichment, focusing on neurological diseases, inflammatory processes, and miRNA–RNA messenger (mRNA) interaction networks. Results: A total of 790 differentially expressed miRNAs were identified in HD patients (270 upregulated and 520 downregulated). IPA revealed enrichment in pathways related to organismal injury, neurological disease, and inflammatory responses. Four major regulatory networks linked differentially expressed miRNAs to neurodegenerative processes, with target genes involved in neuroinflammation, cellular stress responses, and metabolic dysfunction. Cross-referencing with previous RNA-seq data identified 5721 high-confidence miRNA–mRNA interactions, implicating 721 target genes across 54 key canonical pathways. Conclusions: HD patients exhibit a distinct and reproducible peripheral blood miRNA expression signature. These dysregulated miRNAs may represent accessible biomarkers and provide mechanistic insights into HD pathogenesis, with potential applications for diagnosis, prognosis, and therapeutic development. Full article
(This article belongs to the Special Issue Neurological Diseases: Biomarkers, Diagnosis and Prognosis)
Show Figures

Figure 1

31 pages, 4404 KB  
Article
Integrative Transcriptomic and Network-Based Analysis of Neuromuscular Diseases
by Federico García-Criado, Lucia Hurtado-García, Elena Rojano, Álvaro Esteban-Martos, Jesús Pérez-García, Pedro Seoane and Juan A. G. Ranea
Int. J. Mol. Sci. 2025, 26(19), 9376; https://doi.org/10.3390/ijms26199376 - 25 Sep 2025
Abstract
Neuromuscular diseases (NMDs) like Duchenne muscular dystrophy (DMD), limb–girdle muscular dystrophy (LGMD), and amyotrophic lateral sclerosis (ALS) are rare, progressive disorders with complex molecular mechanisms. Traditional transcriptomic analyses often struggle to capture systems-level dysregulation, especially given the small sample sizes typical of rare [...] Read more.
Neuromuscular diseases (NMDs) like Duchenne muscular dystrophy (DMD), limb–girdle muscular dystrophy (LGMD), and amyotrophic lateral sclerosis (ALS) are rare, progressive disorders with complex molecular mechanisms. Traditional transcriptomic analyses often struggle to capture systems-level dysregulation, especially given the small sample sizes typical of rare disease studies. Our differential expression analysis of eight public RNA-seq datasets from various cell types in DMD, LGMD, and ALS revealed not only disease-relevant pathways but also unexpected enrichments, such as renal development, suggesting systemic impacts beyond muscle tissue. To address limitations in capturing broader molecular mechanisms, we applied an integrative systems biology approach combining differential expression data, protein–protein interaction (PPI) networks, and network embedding techniques. Comparative functional enrichment revealed shared pathways, including glycosaminoglycan binding in both DMD and FUS-related ALS, implicating extracellular matrix–protein interactions in FUS mutation effects. Mapping DEGs onto the human PPI network and assessing their proximity to causal genes uncovered dysregulated non-coding RNAs, such as PAX8-AS1, SBF2-AS1, and NEAT1, potentially indicating common regulatory roles. We also found candidate genes within disease-proximal clusters, like HS3ST3A1, which may contribute to pathogenesis. Overall, this integrative approach reveals shared transcriptional programs and novel targets, advancing our understanding and potential treatment strategies for NMDs. Full article
Show Figures

Figure 1

20 pages, 929 KB  
Review
Genome Editing by Grafting
by Samuel Simoni, Marco Fambrini, Claudio Pugliesi and Ugo Rogo
Int. J. Mol. Sci. 2025, 26(19), 9294; https://doi.org/10.3390/ijms26199294 - 23 Sep 2025
Viewed by 121
Abstract
Grafting is the process of joining parts of two plants, allowing the exchange of molecules such as small RNAs (including microRNAs and small interfering RNAs), messenger RNAs, and proteins between the rootstock and the scion. Genome editing by grafting exploits RNAs, such as [...] Read more.
Grafting is the process of joining parts of two plants, allowing the exchange of molecules such as small RNAs (including microRNAs and small interfering RNAs), messenger RNAs, and proteins between the rootstock and the scion. Genome editing by grafting exploits RNAs, such as tRNA-like sequences (TLS motifs), to deliver the components (RNA) of the clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9) system from transgenic rootstock to wild-type scion. The complex Cas9 protein and sgRNA-TLS produced in the scion perform the desired modification without the integration of foreign DNA in the plant genome, resulting in heritable transgene-free genome editing. In this review, we examine the current state of the art of this innovation and how it helps address regulatory problems, improves crop recovery and selection, exceeds the usage of viral vectors, and may reduce potential off-target effects. We also discuss the promise of genome editing by grafting for plants recalcitrant to in vitro culture and for agamic-propagated species that must maintain heterozygosity for plant productivity, fruit quality, and adaptation. Furthermore, we explore the limitations of this technique, including variable efficiency, graft incompatibility among genotypes, and challenges in large-scale application, while highlighting its considerable potential for further improvement and future broader applications for crop breeding. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

17 pages, 5123 KB  
Article
Bioinformatics-Based Analysis of the Screening and Evaluation of Potential Targets of FTY720 for the Treatment of Non-Small Cell Lung Cancer
by Mengyuan Han, Sendaer Hailati, Dilihuma Dilimulati, Alhar Baishan, Alifeiye Aikebaier and Wenting Zhou
Biology 2025, 14(10), 1311; https://doi.org/10.3390/biology14101311 - 23 Sep 2025
Viewed by 173
Abstract
Background: A range of cancer cells are significantly inhibited by FTY720. It is unknown, nevertheless, how FTY720 influences the onset of non-small cell lung cancer (NSCLC). Using bioinformatics techniques, we analyzed and the possible molecular mechanisms and targets of FTY720 for the treatment [...] Read more.
Background: A range of cancer cells are significantly inhibited by FTY720. It is unknown, nevertheless, how FTY720 influences the onset of non-small cell lung cancer (NSCLC). Using bioinformatics techniques, we analyzed and the possible molecular mechanisms and targets of FTY720 for the treatment of NSCLC. Methods: DEGs (Differentially expressed genes) were acquired by differential analysis of the dataset GSE10072. Obtained FTY720 target genes and NSCLC disease genes from databases such as Swiss-TargetPrediction and GeneCard. Subsequently, target and disease genes, as well as DEGs, were merged for Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, gene ontology (GO), and protein interaction analysis. The overlapping genes of DEGs and target genes, and disease genes were also obtained separately and subjected to survival as well as expression analyses. We constructed the regulatory network of miRNAs and transcription factors (TFs) on hub genes. Finally, the immune cell association of hub genes was evaluated using the ssGSEA method, molecular docking of FTY720 to hub genes was carried out utilizing Autodock, and molecular dynamics simulations were conducted. Results: In this study, 444 DEGs, 232 target genes of FTY720, and 466 disease genes were obtained. Moreover, a total of 1062 genes were obtained by removing duplicate values after merging, among which PIK3R1, Akt1, and S1PR1 had the highest DEGREE values in the protein interactions network, and these genes were primarily enriched in MAPK, PI3K-Akt signaling pathways, with the PI3K-Akt signaling pathway being the most prominent. Among the overlapping genes, three potential targets of FTY720 for NSCLC treatment were found: S1PR1, ZEB2, and HBEGF. ZEB2 and S1PR1 were determined to be hub genes and to significantly affect NSCLC prognosis by survival analysis. Furthermore, hsa-miR-132-3p, hsa-miR-192-5p, and hsa-miR-6845-3p were strongly associated with FTY720 for the treatment of NSCLC; CTBP1 (carboxy-terminal binding protein 1), EZH2 (protein lysine N-methyltransferase), and ZNF610 (zinc-finger protein 610) may all influence the expression of ZEB2 and S1PR1. Hub genes had a substantial negative link with memory B cells and a significant positive correlation with memory CD8 T cells and Th17 helper T cells. The molecular docking and kinetic simulation results of FTY720 with the two hub genes indicate that the protein-ligand complex has good stability. Conclusion: Our research indicates that FTY720 may inhibit NSCLC via possible targets ZEB2 and S1PR1, further laying the theoretical foundation for the utilization of FTY720 in NSCLC treatment. Full article
(This article belongs to the Topic Advances in Anti-Cancer Drugs: 2nd Edition)
Show Figures

Figure 1

26 pages, 2624 KB  
Review
The Dual Role of RASSF4 in Tumorigenesis: Mechanisms and Epigenetic Targeting Strategies
by Rui Tian, Yixin Wu, Wenbin Yuan, Lingli Tian, Rui Zhang, Hao Lyu, Shuai Xiao, Dong Guo, Qi Zhang, Declan William Ali, Marek Michalak, Cefan Zhou, Jingfeng Tang and Xing-Zhen Chen
Biology 2025, 14(9), 1289; https://doi.org/10.3390/biology14091289 - 18 Sep 2025
Viewed by 348
Abstract
RASSF4 is a key member of the Ras-associated domain family (RASSF) that exhibits dual functionality in tumorigenesis, playing critical yet context-dependent roles in various malignancies. Its expression is epigenetically regulated through promoter hypermethylation, histone modifications, and microRNAs including miR-155 and miR-196a-5p, which directly [...] Read more.
RASSF4 is a key member of the Ras-associated domain family (RASSF) that exhibits dual functionality in tumorigenesis, playing critical yet context-dependent roles in various malignancies. Its expression is epigenetically regulated through promoter hypermethylation, histone modifications, and microRNAs including miR-155 and miR-196a-5p, which directly target its 3′ untranslated region. In most cancers, such as non-small cell lung cancer (NSCLC) and gastric adenocarcinoma (GAC), RASSF4 acts as a tumor suppressor by inhibiting the RAS/MAPK pathway while activating the Hippo signaling cascade, ultimately inducing cell cycle arrest and apoptosis. Conversely, in aRMS, RASSF4 is upregulated by the PAX3-FOXO1 fusion oncoprotein and promotes tumor growth through MST1 inhibition and subsequent YAP activation. This review systematically analyzes current evidence regarding RASSF4’s complex regulatory mechanisms and clinical significance. We propose targeted therapeutic strategies including epigenetic reactivation, gene intervention, and combination therapies. Furthermore, we identify RASSF4 as a promising diagnostic biomarker and therapeutic target based on integrated mechanistic and clinical evidence. Future research should focus on elucidating context-dependent regulatory switches, developing targeted delivery systems, and validating clinical utility through prospective trials. Full article
(This article belongs to the Special Issue Signalling Pathways in Cancer and Disease)
Show Figures

Figure 1

19 pages, 2849 KB  
Article
miR-194-3 Regulates Proliferation and Apoptosis of Follicular Granulosa Cells by Targeting CHD4 in Zhedong White Geese
by Peng Zheng, Zhengyu Zang, Size Wang, Chuicheng Zeng, Yue Pan, Yuanliang Zhang, Shan Yue, Shengjun Liu, He Huang and Xiuhua Zhao
Animals 2025, 15(18), 2676; https://doi.org/10.3390/ani15182676 - 12 Sep 2025
Viewed by 278
Abstract
The dynamic balance between proliferation and apoptosis of follicular granulosa cells (GCs) is crucial for follicular development in poultry. microRNAs play important roles in ovarian development and follicular function. Previous transcriptome analyses showed that miR-194-3 was significantly differentially expressed in the ovaries of [...] Read more.
The dynamic balance between proliferation and apoptosis of follicular granulosa cells (GCs) is crucial for follicular development in poultry. microRNAs play important roles in ovarian development and follicular function. Previous transcriptome analyses showed that miR-194-3 was significantly differentially expressed in the ovaries of Zhedong white geese during the laying and brooding stages. Therefore, the aim of this study was to investigate the regulatory role and molecular mechanism of miR-194-3 on the proliferation and apoptosis of follicular GCs in Zhedong white geese. We first screened the target gene CHD4 of miR-194-3 and constructed the miR-194-3 mimic and inhibitor, a small interfering RNA of target gene CHD4. The experimental results showed that the overexpression of miR-194-3 significantly down-regulated the mRNA and protein expression of GC proliferation genes (PCNA, CDK-2, and CCND-1), reduced the proportion of EdU-labeled positive cells, blocked cell cycle progression, simultaneously up-regulated the mRNA and protein expression of Caspase-3 and Caspase-9, and significantly increased the rate of apoptosis. In contrast, the inhibition of miR-194-3 expression promoted the proliferation of goose follicular GCs, accelerated cell cycle progression, and decreased the apoptosis rate. Bioinformatics prediction combined with the results of the dual luciferase reporter assay confirmed that CHD4 was a direct target gene of miR-194-3. The knockdown of CHD4 expression resulted in the down-regulation of PCNA, CDK-2 and CCND-1 expression; blockage of cell cycle progression; attenuation of cell proliferation; an up-regulation of Caspase-3 and Caspase-9 expression and a significant increase in apoptotic cell death. In summary, both miR-194-3 overexpression and CHD4 knockdown produced similar effects on goose follicular GC proliferation and apoptosis, suggesting that CHD4 may partially mediate the regulatory effects of miR-194-3; however, additional targets or pathways cannot be excluded. Full article
(This article belongs to the Section Poultry)
Show Figures

Figure 1

18 pages, 4537 KB  
Article
Study of FOXL2 Regulation on Ovarian Function in Chlamys farreri Through Comparative ChIP-Seq and Transcriptome Analysis Using RNA Interference
by Xiaoling Liu, Han Yun, Yan Xing, Shuo Wang, Xueying Zhou and Jianbai Zhang
Biology 2025, 14(9), 1259; https://doi.org/10.3390/biology14091259 - 12 Sep 2025
Viewed by 351
Abstract
FOXL2 (forkhead box protein L2) is a transcription factor, its function and regulatory mechanism have been mainly studied in mammals; related research on marine invertebrates is still insufficient. It was found that oogenesis was affected, and even a small number of cells resembling [...] Read more.
FOXL2 (forkhead box protein L2) is a transcription factor, its function and regulatory mechanism have been mainly studied in mammals; related research on marine invertebrates is still insufficient. It was found that oogenesis was affected, and even a small number of cells resembling spermatogonial morphology appeared in C. farreri ovaries after the FOXL2 was knocked down through RNA interference (RNAi) technology in our laboratory previously. Based on previous research, this paper conducted transcriptome sequencing and differential expression analysis on the ovarian tissues between the experimental group (post-RNAi) and the control group (pre-RNAi) of C. farreri, and used recombinant C. farreri FOXL2 protein for antibody production in Chromatin Immunoprecipitation Sequencing (ChIP seq) experiments to comprehensively analyze the pathways and key genes regulated by FOXL2 during oogenesis. The results showed that in the RNAi experimental group, 389 genes were upregulated, and 1615 genes were downregulated. Among the differentially expressed genes (DEGs), the differential genes related to gender or gonadal development are relatively concentrated in physiological processes such as steroid hormone synthesis, spermatogenesis, gonadal development, and ovarian function maintenance, as well as the FoxO and estrogen signaling pathways. Combining transcriptome and ChIP-seq data, it was found that there were some genes related to sex gonadal development among genes which were directly regulated by FOXL2, such as Wnt4, SIRT1, HSD17B8, GABABR1, KRAS, NOTCH1, HSD11B1, cPLA2, ADCY9, IP3R1, PLCB4, and Wnt1. This study lays the foundation for a deeper understanding of the FOXL2′s specific regulatory mechanism during oogenesis in scallops as a transcription factor. Full article
Show Figures

Figure 1

15 pages, 1677 KB  
Article
Synthetic Mimics of Wound-Induced Carrot MicroRNAs Affect Lipid Metabolism in Cultured Adipocytes
by Edwin E. Reza-Zaldívar and Daniel A. Jacobo-Velázquez
Nutrients 2025, 17(18), 2919; https://doi.org/10.3390/nu17182919 - 10 Sep 2025
Viewed by 303
Abstract
Background/Objectives: Plant-derived microRNAs (miRNAs) have emerged as cross-kingdom regulatory molecules, but their capacity to influence mammalian metabolism is still poorly understood. This study aimed to investigate whether miRNAs induced in carrots (Daucus carota) by postharvest wounding stress can modulate adipocyte lipid [...] Read more.
Background/Objectives: Plant-derived microRNAs (miRNAs) have emerged as cross-kingdom regulatory molecules, but their capacity to influence mammalian metabolism is still poorly understood. This study aimed to investigate whether miRNAs induced in carrots (Daucus carota) by postharvest wounding stress can modulate adipocyte lipid accumulation. Methods: High-throughput small RNA sequencing was performed to identify stress-responsive miRNAs in wounded carrots. Bioinformatic analyses predicted potential mammalian targets, focusing on genes involved in adipogenesis and lipid regulation, including those in the insulin and FoxO signaling pathways. Selected miRNAs were functionally validated in 3T3-L1 adipocytes by assessing intracellular triglyceride levels and glycerol release. Results: Six stress-responsive carrot miRNAs were predicted to target mammalian lipid metabolism genes. Functional assays revealed that miR165a-3p, miR232a-5p, and miR1799 significantly decreased intracellular triglyceride accumulation and increased glycerol release, suggesting enhanced lipolysis. These effects support the potential regulation of adipocyte metabolism through plant miRNA mimics. Conclusions: Our findings provide experimental evidence for stress-induced carrot miRNAs mimics potentially modulate fat accumulation. This work expands current understanding of dietary plant miRNAs and highlights their potential role as functional food components for metabolic health improvement. Further research is needed to establish their gastrointestinal stability, uptake from dietary matrices, and in vivo effects. Full article
(This article belongs to the Section Phytochemicals and Human Health)
Show Figures

Figure 1

22 pages, 3880 KB  
Article
Comprehensive Characterization of Long Non-Coding RNAs in Porcine Tissues: Expression Patterns and Functional Insights During Oocyte Development
by Yao Jiang, Yipeng Li, Qingpeng Shen, Xiaolong Yuan, Fei Gao and Bin Ma
Cells 2025, 14(18), 1409; https://doi.org/10.3390/cells14181409 - 9 Sep 2025
Viewed by 334
Abstract
Long non-coding RNAs (lncRNAs) are essential regulatory molecules involved in various biological processes in mammals. However, their expression patterns across multiple porcine tissues have not been systematically characterized. We analyzed 607 RNA-seq datasets derived from 14 porcine tissues, including backfat, gallbladder, heart, ileum, [...] Read more.
Long non-coding RNAs (lncRNAs) are essential regulatory molecules involved in various biological processes in mammals. However, their expression patterns across multiple porcine tissues have not been systematically characterized. We analyzed 607 RNA-seq datasets derived from 14 porcine tissues, including backfat, gallbladder, heart, ileum, jejunum, kidney, longissimus dorsi, liver, lung, skeletal muscle, ovary, pituitary, skeletal muscle, and spleen. Additionally, we examined 63 single-cell RNA-seq datasets from porcine primary oocytes at five developmental stages. For comparative analysis, we included 20 human and 17 mouse oocyte RNA-seq datasets. We identified 52,798 porcine lncRNAs, with tissue-specific expression patterns most prominent in oocytes and least in skeletal muscle. Among them, 2169 were classified as housekeeping and 14,469 as tissue-specific lncRNAs. Cross-species analysis revealed that a small subset of oocyte-expressed lncRNAs is conserved in humans and mice, associated with catalytic activity and circadian regulation. Additionally, 44 lncRNAs were differentially expressed during oocyte development, implicating them in neurogenesis, vesicle transport, and protein modification. Our findings not only contribute to the growing body of knowledge regarding lncRNAs in porcine biology but also pave the way for future research aimed at elucidating their functional roles in reproductive biology and other physiological processes. Full article
(This article belongs to the Section Reproductive Cells and Development)
Show Figures

Figure 1

Back to TopTop