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
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (280)

Search Parameters:
Keywords = RNA stability and degradation

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
28 pages, 2988 KiB  
Review
Circular RNAs as Targets for Developing Anticancer Therapeutics
by Jaewhoon Jeoung, Wonho Kim, Hyein Jo and Dooil Jeoung
Cells 2025, 14(14), 1106; https://doi.org/10.3390/cells14141106 - 18 Jul 2025
Viewed by 476
Abstract
Circular RNA (CircRNA) is a single-stranded RNA arising from back splicing. CircRNAs interact with mRNA, miRNA, and proteins. These interactions regulate various life processes, including transcription, translation, cancer progression, anticancer drug resistance, and metabolism. Due to a lack of cap and poly(A) tails, [...] Read more.
Circular RNA (CircRNA) is a single-stranded RNA arising from back splicing. CircRNAs interact with mRNA, miRNA, and proteins. These interactions regulate various life processes, including transcription, translation, cancer progression, anticancer drug resistance, and metabolism. Due to a lack of cap and poly(A) tails, circRNAs show exceptional stability and resistance to RNase degradation. CircRNAs exhibit dysregulated expression patterns in various cancers and influence cancer progression. Stability and regulatory roles in cancer progression make circRNAs reliable biomarkers and targets for the development of anticancer therapeutics. The dysregulated expression of circRNAs is associated with resistance to anticancer drugs. Enhanced glycolysis by circRNAs leads to resistance to anticancer drugs. CircRNAs have been known to regulate the response to chemotherapy drugs and immune checkpoint inhibitors. Exogenous circRNAs can encode antigens that can induce both innate and adaptive immunity. CircRNA vaccines on lipid nanoparticles have been shown to enhance the sensitivity of cancer patients to immune checkpoint inhibitors. In this review, we summarize the roles and mechanisms of circRNAs in anticancer drug resistance and glycolysis. This review discusses clinical applications of circRNA vaccines to overcome anticancer drug resistance and enhance the efficacy of immune checkpoint inhibitors. The advantages and disadvantages of circRNA vaccines are also discussed. Overall, this review stresses the potential value of circRNAs as new therapeutic targets and diagnostic/prognostic biomarkers for cancer Full article
Show Figures

Figure 1

26 pages, 1458 KiB  
Review
Innovation in mRNA Vaccines and RNAi via Protein Nanocages
by Sohrab Ahmadivand
Vaccines 2025, 13(6), 653; https://doi.org/10.3390/vaccines13060653 - 18 Jun 2025
Viewed by 927
Abstract
Self-assembling protein nanocages (SAPNs) are distinct natural structures formed by the self-assembly of identical subunits, providing a highly efficient platform and a novel strategy for vaccine development and RNAi therapy. Their internal cavity allows for precise cargo encapsulation, while the externally modifiable surface [...] Read more.
Self-assembling protein nanocages (SAPNs) are distinct natural structures formed by the self-assembly of identical subunits, providing a highly efficient platform and a novel strategy for vaccine development and RNAi therapy. Their internal cavity allows for precise cargo encapsulation, while the externally modifiable surface supports multivalent antigen presentation, thereby enhancing stability, targeted delivery, and immune activation. In addition to serving as stable subunit vaccines with multivalent antigen display, SAPNs can be incorporated into mRNA vaccines (SAPN-RNA vaccines) by pre-fusing with the antigen. This strategy stabilizes secreted antigenic proteins with prolonged presentation to the immune system, and improves vaccine efficacy while reducing off-target effects and minimizing required doses. Additionally, SAPNs can overcome cellular uptake barriers, enhance DNA vaccine efficacy, and enable the co-delivery of antigens and adjuvants. Functionalization with adjuvants or targeting ligands further improves their immunostimulatory properties and specificity. The SAPN-RNAi strategy optimizes siRNA delivery by promoting lysosomal escape, enhancing targeted uptake, and protecting siRNA from degradation through SAPN encapsulation. This review examines the structural and functional properties of protein nanocages and their applications in vaccine design and RNAi delivery, emphasizing their synergistic effects, and exploring current progress, challenges, and future directions. In conclusion, SAPNs represent a versatile multifunctional platform with broad applicability across subunit, mRNA and DNA vaccines, adjuvant co-delivery, and RNAi therapeutics, with significant potential against viral infections. Full article
(This article belongs to the Collection Advance in Nanoparticles as Vaccine Adjuvants)
Show Figures

Figure 1

30 pages, 4591 KiB  
Article
Evolution of the Soil Bacterial Community as a Function of Crop Management: A Metagenomic Study in Orange Tree (Citrus sinensis) Plantations
by Carlos Giménez-Valero, Alejandro Andy Maciá-Vázquez, Dámaris Núñez-Gómez, Agustín Conesa, Vicente Lidón and Pablo Melgarejo
Plants 2025, 14(12), 1781; https://doi.org/10.3390/plants14121781 - 11 Jun 2025
Viewed by 431
Abstract
Soil management significantly influences the structure and diversity of soil bacterial communities, affecting biodiversity and ecosystem functions. In semi-arid regions, water efficiency strategies like anti-weed netting are implemented, but their impact on soil microbial communities remains underexplored. This study evaluates the temporal evolution [...] Read more.
Soil management significantly influences the structure and diversity of soil bacterial communities, affecting biodiversity and ecosystem functions. In semi-arid regions, water efficiency strategies like anti-weed netting are implemented, but their impact on soil microbial communities remains underexplored. This study evaluates the temporal evolution of soil bacterial communities in orange tree (Citrus sinensis (L.) Osbeck) plantations under two conditions: with and without anti-weed netting. Soil samples were collected at three time points over a period of 18 months since the establishment of the crop and analyzed using high-throughput 16S rRNA sequencing, assessing alpha and beta diversity, taxonomic composition, and functional pathways via KEGG analysis. The results indicate that weed control netting contributes to stabilizing bacterial diversity over time and increases the relative abundance of dominant phyla such as Planctomycetota, Proteobacteria, Bacteroidota, and Acidobacteriota. Functional predictions revealed significant differences in metabolic pathways, including those associated with nitrogen fixation and organic matter degradation. These findings suggest that anti-weed netting not only influences the taxonomic composition of soil bacterial communities but also modulates their functional potential, with implications for sustainable agriculture in semi-arid environments. This study provides new insights into the interaction between soil management and soil bacterial communities, offering valuable information for optimizing agricultural practices and soil conservation strategies. Full article
(This article belongs to the Section Plant–Soil Interactions)
Show Figures

Figure 1

17 pages, 1078 KiB  
Review
MicroRNAs’ Impact on Heart Diseases
by Marco Antonio Cordeiro, Ana Elisa T. S. de Carvalho and Regina Celia Spadari
Int. J. Mol. Sci. 2025, 26(12), 5566; https://doi.org/10.3390/ijms26125566 - 10 Jun 2025
Viewed by 553
Abstract
Cardiovascular diseases (CVDs) are the most prevalent cause of global mortality, highlighting the importance of understanding their molecular bases. Recently, small non-coding RNAs (miRNAS) were shown to affect messenger RNA (mRNA) stability, either by inhibiting translation or by causing degradation through base pairing [...] Read more.
Cardiovascular diseases (CVDs) are the most prevalent cause of global mortality, highlighting the importance of understanding their molecular bases. Recently, small non-coding RNAs (miRNAS) were shown to affect messenger RNA (mRNA) stability, either by inhibiting translation or by causing degradation through base pairing with mRNAs, being negative regulators of protein translation. Moreover, miRNAs modulate many signaling pathways and cellular processes, including cell-to-cell communication. In the cardiovascular system, miRNAs control functions in cardiomyocytes, endothelial cells, smooth muscle cells, and fibroblasts. Because miRNA expression was detected in the blood of patients with various cardiovascular diseases, they are considered attractive candidates for noninvasive biomarkers. This study reviews the literature on the role played by miRNAs in heart development and diseases. The findings suggest that miRNA regulation may offer new perspectives for therapeutic interventions in heart diseases. Full article
Show Figures

Figure 1

18 pages, 6423 KiB  
Article
Silver Nanoparticles as a Novel Tissue Preservative: A Comparative Study with 10% Neutral Buffered Formalin
by Safa Taha, Amina Ismaeel, Muna Aljishi, Samvel Selvam, Angeleena Esther and Khaled Greish
Int. J. Mol. Sci. 2025, 26(11), 5335; https://doi.org/10.3390/ijms26115335 - 1 Jun 2025
Viewed by 466
Abstract
Tissue preservation plays an essential role in biomedical research and histopathological applications. Traditional methods, despite their efficiency, are associated with compromised long-term tissue integrity and probable ecotoxicities. This study explores the application of silver nanoparticles (AgNPs), known for their antimicrobial properties, as a [...] Read more.
Tissue preservation plays an essential role in biomedical research and histopathological applications. Traditional methods, despite their efficiency, are associated with compromised long-term tissue integrity and probable ecotoxicities. This study explores the application of silver nanoparticles (AgNPs), known for their antimicrobial properties, as a potential tissue preservative. In this work, AgNPs were synthesized via a chemical reduction method. Heart, liver, and kidney tissues were obtained from BALB/c mice and preserved using 10% neutral buffered formalin (NBF) and AgNPs solution for 72 h. Preservation efficiency was assessed by quantifying and measuring DNA and RNA integrity, evaluating protein stability, and conducting histopathological examinations. This study aimed to compare the performance of AgNPs against 10% NBF across these parameters to determine their suitability as an alternative fixative. Our results showed that AgNPs solution maintained consistent DNA, RNA, and protein concentrations/quality across all tissues over 72 h, whereas formalin treatment led to degradation over time. Conversely, 10% NBF demonstrated better preservation of tissue morphology. These results highlighted the differential strengths of each fixative, with AgNPs excelling in molecular preservation and NBF in structural integrity. Overall, AgNPs exhibited superior qualitative and quantitative preservation of nucleic acids and intracellular proteins, indicating their potential as an alternative to formalin for molecular testing. Despite their demonstrated efficacy in biomolecular preservation, further studies are needed to optimize tissue morphology preservation. Full article
Show Figures

Figure 1

20 pages, 1757 KiB  
Article
A Complete Approach for circRNA Therapeutics from Purification to Lyophilized Delivery Using Novel Ionizable Lipids
by Esther Broset, Ana Larraga, Verónica Lampaya, Víctor Navarro, Alexandre López-Gavín, Diego de Miguel, Álvaro Peña, Juan Martínez-Oliván and Diego Casabona
Int. J. Mol. Sci. 2025, 26(11), 5138; https://doi.org/10.3390/ijms26115138 - 27 May 2025
Viewed by 1446
Abstract
Circular RNA (circRNA) has gained significant attention as a potential therapeutic tool due to its remarkable stability and resistance to degradation by exonucleases. However, scalable and efficient methods for purification and delivery remain critical challenges that must be addressed. In this study, we [...] Read more.
Circular RNA (circRNA) has gained significant attention as a potential therapeutic tool due to its remarkable stability and resistance to degradation by exonucleases. However, scalable and efficient methods for purification and delivery remain critical challenges that must be addressed. In this study, we developed and evaluated an optimized affinity chromatography method using Oligo (dT) columns for the purification of circRNA, achieving high yield and purity compared with high-performance liquid chromatography. Additionally, we investigated the in vivo efficacy of circRNA-Oligo (dT) encapsulated in lipid nanoparticles (LNPs) formulated with emerging ionizable lipids, including CP-LC-0867 and CP-LC-0729. Our results showed that LNPs formulated with CP-LC-0867 consistently produced higher protein expression compared to SM-102, with sustained luciferase activity observed over a 14-day period. Furthermore, we assessed the lyophilization potential of LNP-circRNA-Oligo (dT) using CP-LC-0729 to extend the shelf life and eliminate the need for ultra-low-temperature storage. Remarkably, the lyophilized LNPs exhibited no significant differences in protein expression compared to their non-lyophilized counterparts, demonstrating that lyophilization is a viable strategy for extending the storage and transport of circRNA therapies. These findings underscore the potential of optimized new ionizable lipids, improved purification strategies, and lyophilization techniques to enhance the scalability, stability, and practical application of circRNA therapies. Full article
Show Figures

Graphical abstract

29 pages, 1950 KiB  
Review
Mechanisms of Immune Evasion in HIV-1: The Role of Virus-Host Protein Interactions
by Antonios Mouzakis, Vasileios Petrakis, Eleni Tryfonopoulou, Maria Panopoulou, Periklis Panagopoulos and Katerina Chlichlia
Curr. Issues Mol. Biol. 2025, 47(5), 367; https://doi.org/10.3390/cimb47050367 - 16 May 2025
Viewed by 2271
Abstract
This review explores the mechanisms by which Human Immunodeficiency Virus type 1 (HIV-1) regulatory proteins manipulate host cellular pathways to promote viral replication and immune evasion. Key viral proteins, such as Nef, Vpu, Vif, Vpr, and Env, disrupt immune defenses by downregulating surface [...] Read more.
This review explores the mechanisms by which Human Immunodeficiency Virus type 1 (HIV-1) regulatory proteins manipulate host cellular pathways to promote viral replication and immune evasion. Key viral proteins, such as Nef, Vpu, Vif, Vpr, and Env, disrupt immune defenses by downregulating surface molecules such as CD4 (Cluster of Differentiation 4) and Major Histocompatibility Complex (MHC) class I, degrading antiviral enzymes like APOBEC3G (Apolipoprotein B mRNA editing catalytic polypeptide-3G) and SAMHD1 (Sterile Alpha Motif and Histidine Aspartate domain-containing protein 1), and counteracting restriction factors including BST-2 (Bone Marrow Stromal Antigen 2)/Tetherin and SERINC5 (Serin Incorporator 5). These interactions support viral persistence and contribute to the establishment of chronic infection. Emerging therapeutic strategies aim to disrupt these HIV-host interactions to restore innate antiviral responses and enhance immune clearance. Approaches such as stabilizing host restriction factors or blocking viral antagonists offer a promising alternative to conventional antiretroviral therapy. By targeting host-dependent pathways, these interventions may reduce drug resistance, tackle latent reservoirs, and provide a pathway toward sustained viral remission or functional cure. Full article
Show Figures

Graphical abstract

12 pages, 3464 KiB  
Commentary
The Central Role of Ribosomal Proteins in p53 Regulation
by Mikael S. Lindström
Cancers 2025, 17(10), 1597; https://doi.org/10.3390/cancers17101597 - 8 May 2025
Viewed by 973
Abstract
The tumor suppressor protein p53 prevents the malignant transformation of cells by responding to DNA damage, oncogene activation, and abnormal growth signals including ribosome assembly defects. Under normal conditions, p53 activity is controlled by the regulatory proteins MDM2 and MDM4, which suppress its [...] Read more.
The tumor suppressor protein p53 prevents the malignant transformation of cells by responding to DNA damage, oncogene activation, and abnormal growth signals including ribosome assembly defects. Under normal conditions, p53 activity is controlled by the regulatory proteins MDM2 and MDM4, which suppress its function through ubiquitin-mediated degradation and transcriptional inhibition. A subset of ribosomal proteins initiates the p53 response to impaired ribosome biogenesis. The ability of some ribosomal proteins to control MDM2 and MDM4 activities, and thereby p53, underscores an intriguing aspect of cell biology: proteins primarily known for their roles in ribosome function can exert extra-ribosomal functions. One notable example is the cellular RNA-protein complex involving RPL5, RPL11, and 5S rRNA (5S RNP) which inhibits MDM2 and stabilizes p53. Another RP, RPL22, is frequently mutated in cancers with microsatellite instability and its paralog RPL22L1 is often amplified. Recent studies have revealed that RPL22 directly modulates the alternative splicing of MDM4 to promote p53 activation, suggesting that the ribosomal protein-p53 relationship is more complex than previously thought. Cellular responses to ribosome biogenesis inhibition extend beyond general alterations in transcription and translation to actively determine cancer cell fate by selectively engaging tumor-suppressor pathways. RPL22’s effect on MDM4 and other mRNA splicing events is a striking example. A better understanding of the mechanisms involved could guide the development of improved cancer treatments. Full article
(This article belongs to the Special Issue Insights from the Editorial Board Member)
Show Figures

Figure 1

17 pages, 1350 KiB  
Review
Regulatory Roles of E3 Ubiquitin Ligases and Deubiquitinases in Bone
by Haotian He, Lifei Wang, Bao Xian and Yayi Xia
Biomolecules 2025, 15(5), 679; https://doi.org/10.3390/biom15050679 - 7 May 2025
Viewed by 767
Abstract
E3 ubiquitin ligases and deubiquitinating enzymes (DUBs) are pivotal regulators of bone homeostasis, orchestrating osteoblast differentiation, proliferation, and osteoclast activity by controlling protein degradation and stability. This review delineates the roles of key E3 ligases (e.g., Smurf1, Smurf2, TRIM family) and DUBs (e.g., [...] Read more.
E3 ubiquitin ligases and deubiquitinating enzymes (DUBs) are pivotal regulators of bone homeostasis, orchestrating osteoblast differentiation, proliferation, and osteoclast activity by controlling protein degradation and stability. This review delineates the roles of key E3 ligases (e.g., Smurf1, Smurf2, TRIM family) and DUBs (e.g., USP family) in bone formation and resorption. E3 ligases such as Smurf1/2 inhibit osteogenesis by degrading BMP/Smad signaling components, while TRIM proteins and HERC ligases promote osteoblast differentiation. Conversely, DUBs like USP2 and USP34 stabilize β-catenin and Smad1/RUNX2, enhancing osteogenic pathways, whereas USP10 and USP12 suppress differentiation. Dysregulation of these enzymes contributes to osteoporosis, fracture non-union, and other bone disorders. The interplay between ubiquitination and deubiquitination, alongside the regulatory role of miRNA and environmental factors, underscores their therapeutic potential. Future research should focus on developing therapies targeting E3 ubiquitin ligases, deubiquitinases, miRNA regulators, and small-molecule inhibitors to restore bone homeostasis in osteoporosis and fracture healing disorders. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

17 pages, 584 KiB  
Review
microRNAs as Biomarkers of Breast Cancer
by Wojciech Jelski, Sylwia Okrasinska and Barbara Mroczko
Int. J. Mol. Sci. 2025, 26(9), 4395; https://doi.org/10.3390/ijms26094395 - 6 May 2025
Viewed by 1056
Abstract
Breast cancer (BC) is the most common type of cancer found in women. Detection of this cancer at an early stage is essential for effective treatment and a favorable prognosis. Potential early breast cancer biomarkers useful for diagnosing these tumors are microRNAs. These [...] Read more.
Breast cancer (BC) is the most common type of cancer found in women. Detection of this cancer at an early stage is essential for effective treatment and a favorable prognosis. Potential early breast cancer biomarkers useful for diagnosing these tumors are microRNAs. These are small single-stranded RNA chains that can regulate the post-transcriptional expression of many different oncogenes. Cancer cells contain miRNAs that play a special role in the etiology of cancer development. The role of microRNAs in the initiation and development of breast cancer gives us great hope for the creation of molecular tools for early cancer detection. MicroRNAs are characterized by a high stability due to RNase, which protects them from degradation and enables their detection in various biological fluids. Researchers have described multiple serum microRNA signatures useful for detecting breast cancer. This review discusses the importance and potential usefulness of microRNAs in detecting breast cancer at an early stage, predicting the course of the disease, and assessing the effectiveness of treatment. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

18 pages, 1714 KiB  
Review
The Role of N6-Methyladenosine (m6A) RNA Modification in the Pathogenesis of Parkinson’s Disease
by Yulu Wang, Tianyuan Zhao, Chunsen Yuan and Xuechai Chen
Biomolecules 2025, 15(5), 617; https://doi.org/10.3390/biom15050617 - 23 Apr 2025
Cited by 1 | Viewed by 1038
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease with a high prevalence among the middle-aged and elderly population. The pathogenesis of PD is closely linked to the misfolding and aggregation of α-synuclein, which contributes to the formation of Lewy bodies. These processes are associated [...] Read more.
Parkinson’s disease (PD) is a neurodegenerative disease with a high prevalence among the middle-aged and elderly population. The pathogenesis of PD is closely linked to the misfolding and aggregation of α-synuclein, which contributes to the formation of Lewy bodies. These processes are associated with the degeneration of dopaminergic neurons, a key neuropathological change that underlies the motor symptoms of PD. In addition, genetic susceptibility, mitochondrial dysfunction, oxidative stress and neuroinflammation are involved in the progress of the disease. Previous studies indicated that the dysregulation of epigenetic modifications, including DNA methylation and histone acetylation, may be the key pathophysiological factors in PD. N6-methyladenosine (m6A) is a dynamically reversible modification in eukaryotes RNA, and could regulate mRNA degradation, stability, maturation, and translation. Recently, clinical research has shown that the global m6A level is significantly reduced in PD patients as well as the expression changes in m6A-associated proteins. Moreover, the dysregulation of m6A modification was shown to impact dopamine metabolism and damage dopaminergic neurons, indicating that m6A RNA modification may play a critical role in the pathogenesis of PD. In this review, we summarize recent clinical studies on m6A RNA modification in PD patients and discuss the regulatory role of m6A modification in dopamine metabolism and dopaminergic neurons death. Furthermore, based on the different m6A modification databases and prediction websites, we analyzed the potential m6A modification sites on the mRNA of key PD pathogenic genes (SNCA, PRKN, PINK1, and LRRK2) for the first time, aiming to offer new gene targets and perspectives understanding the pathogenesis of PD. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

50 pages, 6313 KiB  
Review
A Review on the Stability Challenges of Advanced Biologic Therapeutics
by Sruthi Sarvepalli, Shashank Reddy Pasika, Vartika Verma, Anusha Thumma, Sandeep Bolla, Pavan Kumar Nukala, Arun Butreddy and Pradeep Kumar Bolla
Pharmaceutics 2025, 17(5), 550; https://doi.org/10.3390/pharmaceutics17050550 - 23 Apr 2025
Cited by 2 | Viewed by 2409
Abstract
Advanced biotherapeutic systems such as gene therapy, mRNA lipid nanoparticles, antibody–drug conjugates, fusion proteins, and cell therapy have proven to be promising platforms for delivering targeted biologic therapeutics. Preserving the intrinsic stability of these advanced therapeutics is essential to maintain their innate structure, [...] Read more.
Advanced biotherapeutic systems such as gene therapy, mRNA lipid nanoparticles, antibody–drug conjugates, fusion proteins, and cell therapy have proven to be promising platforms for delivering targeted biologic therapeutics. Preserving the intrinsic stability of these advanced therapeutics is essential to maintain their innate structure, functionality, and shelf life. Nevertheless, various challenges and obstacles arise during formulation development and throughout the storage period due to their complex nature and sensitivity to various stress factors. Key stability concerns include physical degradation and chemical instability due to various factors such as fluctuations in pH and temperature, which results in conformational and colloidal instabilities of the biologics, adversely affecting their quality and therapeutic efficacy. This review emphasizes key stability issues associated with these advanced biotherapeutic systems and approaches to identify and overcome them. In gene therapy, the brittleness of viral vectors and gene encapsulation limits their stability, requiring the use of stabilizers, excipients, and lyophilization. Keeping cells viable throughout the whole cell therapy process, from culture to final formulation, is still a major difficulty. In mRNA therapeutics, stabilization strategies such as the optimization of mRNA nucleotides and lipid compositions are used to address the instability of both the mRNA and lipid nanoparticles. Monoclonal antibodies are colloidally and conformationally unstable. Hence, buffers and stabilizers are useful to maintain stability. Although fusion proteins and monoclonal antibodies share structural similarities, they show a similar pattern of instability. Antibody–drug conjugates possess issues with conjugation and linker stability. This review outlines the stability issues associated with advanced biotherapeutics and provides insights into the approaches to address these challenges. Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Figure 1

15 pages, 4531 KiB  
Article
RNA Binding to CCRRM of PABPN1 Induces Conformation Change
by Shengping Zhang, Ting Chen, Yunlong Zhang and Changrui Lu
Biology 2025, 14(4), 432; https://doi.org/10.3390/biology14040432 - 17 Apr 2025
Viewed by 529
Abstract
Poly(A) Binding Protein Nuclear 1 (PABPN1) is a nuclear poly(A)-binding protein that is highly conserved in eukaryotes. It plays multifaceted roles in RNA processing and metabolism, with its dysregulation closely linked to various diseases. PABPN1 contains an alanine-rich N-terminus, a central coiled-coil domain [...] Read more.
Poly(A) Binding Protein Nuclear 1 (PABPN1) is a nuclear poly(A)-binding protein that is highly conserved in eukaryotes. It plays multifaceted roles in RNA processing and metabolism, with its dysregulation closely linked to various diseases. PABPN1 contains an alanine-rich N-terminus, a central coiled-coil domain (CC), a conserved RNA recognition motif (RRM) and a C-terminal extension. PABPN1 influences mRNA splicing and stability through its RNA-binding capabilities, thereby modulating gene expression. While PABPN1 is known to interact with RNA, the molecular mechanism underlying this interaction with RNA awaits further investigation. Here, we designed and purified a PABPN1 fragment encompassing the RNA-binding domain (CCRRM fragment, amino acids 114–254). Using a combination of 3D modeling, small-angle X-ray scattering (SAXS) and selective 2′-hydroxyl acylation analyzed by primer extension (SHAPE) assay, our result indicated that CCRRM exhibits a high affinity for poly(A) RNA, a moderate affinity for GU-rich and CU-rich sequences, and negligible binding to AU-rich and CA-rich sequences. RNA binding induces conformation change in the CC. These results suggest that PABPN1 could potentially be involved in cytoplasmic polyadenylation and may influence the regulation of mRNA translation and degradation, although further investigation is required to confirm this role. Full article
(This article belongs to the Section Biochemistry and Molecular Biology)
Show Figures

Figure 1

26 pages, 712 KiB  
Review
Unveiling Pharmacogenomics Insights into Circular RNAs: Toward Precision Medicine in Cancer Therapy
by Saud Alqahtani, Taha Alqahtani, Krishnaraju Venkatesan, Durgaramani Sivadasan, Rehab Ahmed, Hassabelrasoul Elfadil, Premalatha Paulsamy and Kalaiselvi Periannan
Biomolecules 2025, 15(4), 535; https://doi.org/10.3390/biom15040535 - 5 Apr 2025
Cited by 2 | Viewed by 1007
Abstract
Pharmacogenomics is revolutionizing precision medicine by enabling tailored therapeutic strategies based on an individual genetic and molecular profile. Circular RNAs (circRNAs), a distinct subclass of endogenous non-coding RNAs, have recently emerged as key regulators of drug resistance, tumor progression, and therapeutic responses. Their [...] Read more.
Pharmacogenomics is revolutionizing precision medicine by enabling tailored therapeutic strategies based on an individual genetic and molecular profile. Circular RNAs (circRNAs), a distinct subclass of endogenous non-coding RNAs, have recently emerged as key regulators of drug resistance, tumor progression, and therapeutic responses. Their covalently closed circular structure provides exceptional stability and resistance to exonuclease degradation, positioning them as reliable biomarkers and novel therapeutic targets in cancer management. This review provides a comprehensive analysis of the interplay between circRNAs and pharmacogenomics, focusing on their role in modulating drug metabolism, therapeutic efficacy, and toxicity profiles. We examine how circRNA-mediated regulatory networks influence chemotherapy resistance, alter targeted therapy responses, and impact immunotherapy outcomes. Additionally, we discuss emerging experimental tools and bioinformatics techniques for studying circRNAs, including multi-omics integration, machine learning-driven biomarker discovery, and high-throughput sequencing technologies. Beyond their diagnostic potential, circRNAs are being actively explored as therapeutic agents and drug delivery vehicles. Recent advancements in circRNA-based vaccines, engineered CAR-T cells, and synthetic circRNA therapeutics highlight their transformative potential in oncology. Furthermore, we address the challenges of standardization, reproducibility, and clinical translation, emphasizing the need for rigorous biomarker validation and regulatory frameworks to facilitate their integration into clinical practice. By incorporating circRNA profiling into pharmacogenomic strategies, this review underscores a paradigm shift toward highly personalized cancer therapies. circRNAs hold immense potential to overcome drug resistance, enhance treatment efficacy, and optimize patient outcomes, marking a significant advancement in precision oncology. Full article
(This article belongs to the Special Issue The Role of Non-Coding RNAs in Health and Disease)
Show Figures

Figure 1

24 pages, 3656 KiB  
Review
Modified-Release Pulmonary Delivery Systems for Labile Bioactives: Design, Development, and Applications
by Shivani Nana, Mershen Govender and Yahya E. Choonara
Pharmaceutics 2025, 17(4), 470; https://doi.org/10.3390/pharmaceutics17040470 - 3 Apr 2025
Cited by 1 | Viewed by 713
Abstract
Pulmonary delivery of bioactives has shown to be a promising route for the treatment of respiratory conditions, however, numerous physiological barriers, such as mucociliary clearance and immune responses, pose significant hurdles to treatment efficacy. These barriers specifically affect labile bioactives such as mRNA, [...] Read more.
Pulmonary delivery of bioactives has shown to be a promising route for the treatment of respiratory conditions, however, numerous physiological barriers, such as mucociliary clearance and immune responses, pose significant hurdles to treatment efficacy. These barriers specifically affect labile bioactives such as mRNA, peptides, proteins, and probiotics, which are susceptible to degradation due to the prevailing conditions. Various drug delivery platforms have been developed to address these challenges, including, among others, polymeric nanoparticles, micelles, liposomes, and solid lipid nanoparticles that encapsulate and protect the labile bioactives during formulation and administration, enabling improved bioavailability, sustained release, and enhanced formulation stability, while further modification of these platforms allows for targeted drug delivery. This review explores the advanced drug delivery systems that have been designed to protect and release labile active agents in a controlled and targeted manner to the lung, with a specific focus provided on the physiological barriers to effective pulmonary delivery and the formulation considerations to overcome these challenges. The outlook of this pertinent field of study has additionally been provided, highlighting the significant potential of the pulmonary delivery of labile bioactive agents for the prevention and treatment of a variety of respiratory ailments. Full article
(This article belongs to the Special Issue Application of Nanomaterials in Pulmonary Drug Delivery)
Show Figures

Figure 1

Back to TopTop