1
|
Rahimi M, Kariminezhad Z, Rondon EP, Fahmi H, Fernandes JC, Benderdour M. Chitosan nanovectors for siRNA delivery: New horizons for nonviral gene therapy. Carbohydr Polym 2025; 360:123581. [PMID: 40399008 DOI: 10.1016/j.carbpol.2025.123581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/25/2025] [Accepted: 04/04/2025] [Indexed: 05/23/2025]
Abstract
The growing interest in RNA-based therapeutics has positioned small interfering RNA (siRNA) as a promising tool for gene silencing with high specificity and efficacy. However, the successful clinical application of siRNA therapies requires efficient delivery systems to overcome extracellular and intracellular barriers. Chitosan, a naturally derived polysaccharide, has gained significant attention as a non-viral vector due to its biodegradability, biocompatibility, mucoadhesive properties, and capacity to enhance cellular uptake. These attributes make chitosan an attractive alternative to lipid-based nanoparticles, which currently dominate siRNA delivery platforms. Recent advancements in chitosan-based nanoformulations, including chemical modifications and functionalization strategies, have improved siRNA stability, targeting efficiency, and transfection potential, addressing key limitations such as low bioavailability and immunogenicity. Despite these advances, challenges remain in achieving optimal release kinetics, scalability, and consistent therapeutic efficacy. Future research efforts will focus on engineering chitosan derivatives with enhanced physicochemical properties, integrating multifunctional nanocarriers, and refining formulation strategies to bridge the gap between preclinical research and clinical translation. The continued development of chitosan-based siRNA therapeutics holds significant potential for advancing precision medicine and expanding treatment options for a variety of diseases, including cancer, metabolic disorders, and inflammatory conditions.
Collapse
Affiliation(s)
- Mahdi Rahimi
- Orthopedics Research Laboratory, Research Center, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Montréal, Québec H4J 1C5, Canada
| | - Zahra Kariminezhad
- Orthopedics Research Laboratory, Research Center, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Montréal, Québec H4J 1C5, Canada; Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Elsa-Patricia Rondon
- Orthopedics Research Laboratory, Research Center, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Montréal, Québec H4J 1C5, Canada; Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Hassan Fahmi
- Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Julio C Fernandes
- Orthopedics Research Laboratory, Research Center, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Montréal, Québec H4J 1C5, Canada; Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Mohamed Benderdour
- Orthopedics Research Laboratory, Research Center, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Montréal, Québec H4J 1C5, Canada.
| |
Collapse
|
2
|
Mansour HM, El-Khatib AS. Oligonucleotide-based therapeutics for neurodegenerative disorders: Focus on antisense oligonucleotides. Eur J Pharmacol 2025; 998:177529. [PMID: 40118328 DOI: 10.1016/j.ejphar.2025.177529] [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: 01/01/2025] [Revised: 03/04/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
Antisense oligonucleotides (ASOs) specifically bind to target RNA sequences and regulate protein expression through various mechanisms. ASOs are a promising therapeutic approach for treating neurodegenerative diseases. The ASO field is a growing area of drug development that focuses on targeting the root cause of diseases at the RNA level, providing a promising alternative to therapies that target downstream processes. Addressing challenges related to off-target effects and inadequate biological activity is essential to successfully develop ASO-based therapies. Researchers have investigated various chemical modifications and delivery strategies to overcome these challenges. This review discusses oligonucleotide-based therapies, particularly ASOs. We discuss the chemical modifications and mechanisms of action of ASOs. Additionally, we recap the results of preclinical and clinical studies testing different ASOs in various neurodegenerative disorders, including spinal muscular atrophy, Huntington's disease, amyotrophic lateral sclerosis, Alzheimer's disease, and Parkinson's disease. In conclusion, ASO drugs show promise as a therapeutic option for treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Heba M Mansour
- Central Administration of Biologicals, Innovative Products, and Clinical Studies, Egyptian Drug Authority, EDA, Giza, Egypt.
| | - Aiman S El-Khatib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
3
|
Yao Z, Liu T, Wang J, Fu Y, Zhao J, Wang X, Li Y, Yang X, He Z. Targeted delivery systems of siRNA based on ionizable lipid nanoparticles and cationic polymer vectors. Biotechnol Adv 2025; 81:108546. [PMID: 40015385 DOI: 10.1016/j.biotechadv.2025.108546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 02/04/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
As an emerging therapeutic tool, small interfering RNA (siRNA) had the capability to down-regulate nearly all human mRNAs via sequence-specific gene silencing. Numerous studies have demonstrated the substantial potential of siRNA in the treatment of broad classes of diseases. With the discovery and development of various delivery systems and chemical modifications, six siRNA-based drugs have been approved by 2024. The utilization of siRNA-based therapeutics has significantly propelled efforts to combat a wide array of previously incurable diseases and advanced at a rapid pace, particularly with the help of potent targeted delivery systems. Despite encountering several extracellular and intracellular challenges, the efficiency of siRNA delivery has been gradually enhanced. Currently, targeted strategies aimed at improving potency and reducing toxicity played a crucial role in the druggability of siRNA. This review focused on recent advancements on ionizable lipid nanoparticles (LNPs) and cationic polymer (CP) vectors applied for targeted siRNA delivery. Based on various types of targeted modifications, we primarily described delivery systems modified with receptor ligands, peptides, antibodies, aptamers and amino acids. Finally, we discussed the challenges and opportunities associated with siRNA delivery systems based on ionizable LNPs and CPs vectors.
Collapse
Affiliation(s)
- Ziying Yao
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Taiqing Liu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingwen Wang
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yunhai Fu
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jinhua Zhao
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoyu Wang
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yinqi Li
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaodong Yang
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiyao He
- Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
4
|
Aram C, Firuzpour F, Barancheshmeh M, Kamali MJ. Unveiling the translational and therapeutic potential of small interfering RNA molecules in combating SARS-CoV-2: A review. Int J Biol Macromol 2025; 318:145203. [PMID: 40513718 DOI: 10.1016/j.ijbiomac.2025.145203] [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: 03/20/2025] [Revised: 05/24/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025]
Abstract
The global COVID-19 pandemic, caused by SARS-CoV-2, has led to significant mortality, with over 6.5 million deaths worldwide. While vaccines have played a crucial role in reducing disease severity, the emergence of viral variants continues to undermine vaccine efficacy, highlighting the need for alternative antiviral strategies. This review explores the potential of RNA interference (RNAi), particularly small interfering RNAs (siRNAs), as a targeted therapeutic approach against SARS-CoV-2. siRNAs can silence specific viral genes with high precision, effectively inhibiting viral replication. We discuss the design and mechanism of siRNAs, their primary molecular targets such as the spike (S), membrane (M), and RNA-dependent RNA polymerase (RdRp) genes and summarize past and current research findings. Special emphasis is placed on delivery systems, especially lung-targeted strategies essential for respiratory infections. We also evaluate how siRNA therapeutics can overcome challenges posed by viral mutations and treatment resistance. The novelty of this work lies in its focused comparison of siRNAs with other non-coding RNAs and its integration of computational tools for siRNA design. This review presents a strategic overview of siRNA development and highlights its translational potential for the current pandemic and future coronavirus outbreaks.
Collapse
Affiliation(s)
- Cena Aram
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Faezeh Firuzpour
- Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | | | - Mohammad Javad Kamali
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
5
|
Coppola G, Cennamo F, Ciccone G, Ibba ML, Ruscio AD, Vito AD, Esposito CL, Catuogno S. Aptamer-based applications in delivering cancer gene therapies and beyond: state of the art and the missing links to clinical translation. Adv Drug Deliv Rev 2025:115639. [PMID: 40513886 DOI: 10.1016/j.addr.2025.115639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Revised: 06/03/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025]
Abstract
The possibility of correcting genetic and epigenetic alterations through gene therapies has been considered a cornerstone in oncology. However, modest results have been achieved in clinics, mainly due to inefficient tumor targeting and side effects. Nucleic acid aptamers are three-dimensional folded single-stranded DNAs or RNAs that selectively bind receptors on cellular membranes, being subsequently internalized via receptor-mediated endocytosis. Thanks to this capability, internalizing aptamers have been investigated as targeting moieties to deliver gene therapies more efficiently and selectively in tumor cells. Promising preclinical results suggested that aptamers could represent the long-awaited step forward in cancer gene therapy. Nevertheless, no clinical trials of aptamer-based gene therapies have been carried out two decades after the first preclinical application, indicating the field could not be sufficiently mature for translatability. The review aims to update thestate of the art regarding aptamers' contribution to gene therapy delivery and to critically highlight the main shortcomings that could have hindered clinical evaluations. In addition, pioneering insights regarding the use of aptamers as co-factors in CRISPR/Cas9 technology or as direct epigenetic regulators are also summarized, revealing more extended applicability not limited to the delivery of cancer gene therapies.
Collapse
Affiliation(s)
- Gabriele Coppola
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy.
| | - Fabiola Cennamo
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy.
| | - Giuseppe Ciccone
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", 81100 Caserta, Italy.
| | - Maria Luigia Ibba
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy; Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Caserta, Italy.
| | - Annalisa Di Ruscio
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA; Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, Boston, MA 02215, USA.
| | - Aldo Di Vito
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy.
| | - Carla Lucia Esposito
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy.
| | - Silvia Catuogno
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy.
| |
Collapse
|
6
|
Chen S, Tu S, Huang Y, Lin H, Wang Y, Dai X. LncRNA MKLN1-AS promotes glioma tumorigenesis and growth via activating the Hippo pathway through miR-126-5p/TEAD1 axis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6787-6799. [PMID: 39680098 DOI: 10.1007/s00210-024-03646-y] [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: 04/30/2024] [Accepted: 11/17/2024] [Indexed: 12/17/2024]
Abstract
The involvement of long non-coding RNAs (lncRNAs) in glioma carcinogenesis has gradually been identified. Herein, we aimed to explore the function and mechanism of lncRNA muskelin 1 antisense RNA (MKLN1-AS) in glioma cell oncogenic properties. Quantitative real-time polymerase chain reaction was utilized to test the expression of MKLN1-AS, miR-126-5p, and TEAD1 (TEA Domain Transcription Factor 1) mRNA expression. Oncogenic properties of glioma cells were characterized using 5-ethynyl-2'-deoxyuridine, flow cytometry, wound healing, transwell, and tube formation assays, respectively. Levels of TEAD1 protein, mobility-related proteins, and Hippo pathway-related proteins were examined by Western blotting. The binding between miR-126-5p and MKLN1-AS or TEAD1 was confirmed by using dual-luciferase reporter and pull-down assays. The murine xenograft model was established for in vivo analysis. Levels of MKLN1-AS in glioma tissues and cell lines were higher, functionally, MKLN1-AS deficiency could suppress glioma cell proliferation, migration, invasion, and angiogenesis, and induce apoptosis in vitro, as well as impede tumor growth in vivo. Mechanistically, miR-126-5p was targeted by MKLN1-AS, miR-126-5p directly targeted TEAD1. The suppressing effects of MKLN1-AS deficiency on glioma cell oncogenic properties were abolished by TEAD1 overexpression or miR-126-5p inhibition. Besides, MKLN1-AS/miR-126-5p mediates the activation of Hippo pathway by TEAD1. MKLN1-AS knockdown weakened glioma cell oncogenic phenotypes and growth via TEAD1-Hippo pathway through miR-126-5p, indicating a new therapeutic target for glioma molecular therapy.
Collapse
Affiliation(s)
- Shouren Chen
- Department of Neurosurgery, Zhangzhou Affiliated Hospital of Fujian Medical University, 59 Shengli West Road, Zhangzhou, Fujian, 363000, P.R. China
| | - Songjie Tu
- Department of Neurosurgery, Zhangzhou Affiliated Hospital of Fujian Medical University, 59 Shengli West Road, Zhangzhou, Fujian, 363000, P.R. China
| | - Yan Huang
- Department of Neurosurgery, Zhangzhou Affiliated Hospital of Fujian Medical University, 59 Shengli West Road, Zhangzhou, Fujian, 363000, P.R. China
| | - Hong Lin
- Department of Neurosurgery, Zhangzhou Affiliated Hospital of Fujian Medical University, 59 Shengli West Road, Zhangzhou, Fujian, 363000, P.R. China
| | - Yuzhe Wang
- Department of Neurosurgery, Zhangzhou Affiliated Hospital of Fujian Medical University, 59 Shengli West Road, Zhangzhou, Fujian, 363000, P.R. China
| | - Xuejun Dai
- Department of Neurosurgery, Zhangzhou Affiliated Hospital of Fujian Medical University, 59 Shengli West Road, Zhangzhou, Fujian, 363000, P.R. China.
| |
Collapse
|
7
|
Feron BKL, Gomez T, Youens NC, Mahmoud NAM, Abdelrahman HKS, Bugert JJ, Richardson SCW. Antiviral siRNA delivered using attenuated, anthrax toxin protects cells from the cytopathic effects of Zika virus. Virus Genes 2025; 61:342-354. [PMID: 40159353 PMCID: PMC12053335 DOI: 10.1007/s11262-025-02152-4] [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: 11/07/2024] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Curative drugs are needed for the treatment of viral infections. Small interfering (si)RNA offer such a prospect but require the development of safe, effective and non-hepatotropic subcellular delivery systems. Here, 5 candidate siRNA molecules targeting defined sequences within the Zika Virus (ZIKV) genome were assayed for their ability to reduce ZIKV induced cytopathic effects in vitro. The protection of Huh-7 cells from ZIKV cytopathic effects was recorded after electroporation and the siRNA Feron-Zv2, resulting in 122.7 ± 5.3% cell viability (n = 3 ± standard error of the mean (SEM), 100 nM siRNA) after exposure to ZIKV relative to a virus treated control (35.2 ± 7.1% cell viability (n = 3 ± SEM)). Protection of BHK-21 cells was recorded after transfection with an attenuated anthrax toxin containing an RNA binding domain. Treatment with Feron-Zv4 resulted in 75.1 ± 2.9% cell viability (n = 3 ± SEM, 25 nM siRNA) after exposure to ZIKV. This protection was mirrored by a system containing octameric PA where a maximum of 86.2 ± 4.4% cell viability was reported (n = 3 ± SEM, 75 nM siRNA) after treatment with Feron-Zv2. Scrambled siRNA afforded no measurable protection. Here we report for the first time that siRNA delivered by either attenuated anthrax toxin or octamer forming ATx can protect mammalian cells from ZIKV cytopathic effects.
Collapse
Affiliation(s)
- Benedita K L Feron
- Exogenix Laboratory, School of Science, University of Greenwich, Central Avenue, Chatham, Kent, ME4 4TB, UK
| | - Timothy Gomez
- Exogenix Laboratory, School of Science, University of Greenwich, Central Avenue, Chatham, Kent, ME4 4TB, UK
| | - Natalie C Youens
- Exogenix Laboratory, School of Science, University of Greenwich, Central Avenue, Chatham, Kent, ME4 4TB, UK
| | - Nourhan A M Mahmoud
- Department of Biochemistry, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, 12451, Egypt
| | - Hadeer K S Abdelrahman
- Department of Biochemistry, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, 12451, Egypt
| | | | - Simon C W Richardson
- Exogenix Laboratory, School of Science, University of Greenwich, Central Avenue, Chatham, Kent, ME4 4TB, UK.
| |
Collapse
|
8
|
Akhilesh, Singh AK, Tiwari V. TRPA1 siRNA-Loaded Nanoformulation Ameliorates Chemotherapy-Induced Peripheral Neuropathy. ACS Chem Neurosci 2025. [PMID: 40421769 DOI: 10.1021/acschemneuro.5c00090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025] Open
Abstract
Small interfering RNA (siRNA) has emerged as a cutting-edge therapeutic strategy, with significant promise for addressing peripheral neuropathies. Despite its immense revolutionary therapeutic potential, the application and sustained release of siRNA for the treatment of chronic pain remain an arduous scientific challenge. This study introduces a novel cationic lipid-based siRNA formulation specifically targeting transient receptor potential ankyrin 1 (TRPA1) for the systemic treatment of chemotherapy-induced neuropathic pain (CINP), a condition with no US-FDA-approved therapeutic options. CINP involves the upregulation of the TRPA1 channel, a key player in nociceptive signaling. Our approach leverages the selective silencing of the TRPA1 gene via siRNA encapsulated in liposomes, offering a targeted and safer therapeutic intervention. The proof-of-principle was established through in vivo experiments, demonstrating significant downregulation of TRPA1 mRNA and protein expressions in the spinal cord following intrathecal administration. Liposomal encapsulation improved siRNA stability and delivery, validated through sophisticated morphometric and analytical techniques. Behavioral assays revealed that both intravenous and intrathecal administrations of this TRPA1 siRNA formulation significantly reduced mechanical and cold hypersensitivity in CINP models. The sustained release profile of siRNA from liposomes ensured prolonged efficacy, contrasting sharply with the transient effects of nonencapsulated siRNA. Mechanistically, silencing of the TRPA1 gene led to decreased microglial activation and reduced expression of inflammatory markers such as ICAM-1 and iba1, mitigating neuroinflammatory responses in the dorsal root ganglia and spinal cord. Intravenous delivery notably outperformed intrathecal administration in downregulating TRPA1 and IL-6 expressions. Overall findings highlight the potential of this nanoengineered TRPA1 siRNA formulation to effectively modulate critical inflammatory pathways and manage CINP. This innovative and exciting strategy not only overcomes the limitations of conventional therapies but also paves the way for new approaches in chronic pain management with significant implications for future clinical applications.
Collapse
Affiliation(s)
- Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, UP India, 221005
| | - Anurag Kumar Singh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, UP India, 221005
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, UP India, 221005
| |
Collapse
|
9
|
Pandey SN, Babu MA, Ali H, H M, Maharana L, Goyal K, Rana M, Imran M. MUC1 as a diagnostic biomarker and siRNA-based therapeutic target in breast cancer: A clinical chemistry perspective. Clin Chim Acta 2025; 576:120387. [PMID: 40425136 DOI: 10.1016/j.cca.2025.120387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2025] [Revised: 05/24/2025] [Accepted: 05/24/2025] [Indexed: 05/29/2025]
Abstract
Breast cancer remains the leading cause of cancer mortality in women, and early detection coupled with real-time monitoring of tumor burden are clinical imperatives; yet existing imaging-based screening (e.g., mammography, ultrasound) suffers from sensitivities as low as 60-80% and even lower in dense breasts plus substantial false-positive rates, underscoring the critical need for molecular assays with higher accuracy. Current clinical assays for circulating MUC1 (CA15-3) achieve high specificity but exhibit limited sensitivity in early-stage disease, underscoring a critical unmet need for more sensitive, multiplexed biomarkers to enable timely intervention. Mass spectrometry-based glycoproteomic workflows offer multiplexed quantification of tumour-associated MUC1 glycoforms, substantially improving analytical specificity and dynamic range. Complementary liquid-biopsy platforms that detect anti-MUC1 autoantibodies further extend lead time for recurrence detection. Concurrently, small interfering RNA (siRNA) therapies targeting MUC1 delivered via ionizable lipid nanoparticles demonstrate efficient tumor accumulation, robust mRNA knockdown, and favourable safety in phaseI solid tumor trials. In this review, we critically assess the analytical performance and standardization challenges of current MUC1 assays, evaluate emerging mass spectrometry and immunoarray techniques, and examine chemical and nanocarrier strategies that surmount biological barriers to siRNA delivery. We propose a co-development framework for harmonized companion diagnostics and MUC1-directed RNAi therapeutics under unified regulatory pathways, paving the way for precision, biomarker-driven interventions in breast cancer care.
Collapse
Affiliation(s)
- Surya Nath Pandey
- Department of Pharmacology, Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad 244001 Uttar Pradesh, India
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA UNIVERSITY, Mathura, UP 281406, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Malathi H
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Laxmidhar Maharana
- Department of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha 751030, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Mohd Imran
- Center For Health Research, Northern Border University, Arar 73213, Saudi Arabia.
| |
Collapse
|
10
|
Kumar G, Ardekani AM. Machine-Learning Framework to Predict the Performance of Lipid Nanoparticles for Nucleic Acid Delivery. ACS APPLIED BIO MATERIALS 2025; 8:3717-3727. [PMID: 40267508 DOI: 10.1021/acsabm.4c01716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Lipid nanoparticles (LNPs) are highly effective carriers for gene therapies, including mRNA and siRNA delivery, due to their ability to transport nucleic acids across biological membranes, low cytotoxicity, improved pharmacokinetics, and scalability. A typical approach to formulate LNPs is to establish a quantitative structure-activity relationship (QSAR) between their compositions and in vitro/in vivo activities, which allows for the prediction of activity based on molecular structure. However, developing QSAR for LNPs can be challenging due to the complexity of multicomponent formulations, interactions with biological membranes, stability in physiological environments, and diverse physicochemical properties. To address these challenges, we developed a machine-learning (ML) framework to predict the activity and cell viability of LNPs for nucleic acid delivery. We curated data from 6454 LNP formulations reported across 21 independent studies and implemented 11 different molecular featurization techniques, ranging from descriptors and fingerprints to graph-based representations, alongside six ML algorithms for binary and multiclass classification. Using scaffold-based 5-fold cross-validation, our models achieved classification accuracies exceeding 90% for both activity and cell viability prediction tasks. Among all model-feature combinations, descriptor-based features combined with ensemble models such as balanced random forest and extra trees yielded the highest performance. Through SHAP-based feature attribution and interaction analysis, we identified key physicochemical properties and compositional features driving the LNP performance, highlighting the importance of synergistic effects among multiple molecular features. Furthermore, we developed a transfer-learning strategy to bridge in vitro-to-in vivo prediction gaps by incorporating base model predictions along with additional biological attributes, such as the particle size, polydispersity index, and ζ potential. Despite the smaller size and inherent class imbalance of the in vivo data set, the transfer-learning models demonstrated a promising predictive performance, with accuracies exceeding 82%. Our findings underscore the potential of interpretable ML frameworks to guide rational LNP design and provide a scalable approach to QSAR modeling in complex nanomedicine systems.
Collapse
Affiliation(s)
- Gaurav Kumar
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Arezoo M Ardekani
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
11
|
Yin K, Villareal L, Wu X, Arcos M, Lee J, Martin DR, In JG, Leslie K, Zhang DD, Xue X. The STEAP4 target NQO1 mediates colon tumorigenesis. J Cell Sci 2025; 138:jcs263402. [PMID: 40205952 DOI: 10.1242/jcs.263402] [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: 06/22/2024] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
Colorectal cancer (CRC) remains a major global health concern, necessitating advancements in therapeutic strategies. Understanding the mechanisms driving CRC is crucial for developing effective treatments. Previous studies, including our own, highlight the role of six-transmembrane epithelial antigen of prostate 4 (STEAP4) in promoting colon tumorigenesis through reactive oxygen species (ROS) generation, making it a promising target. Our research provides compelling evidence that STEAP4 knockout significantly reduces colon tumorigenesis in a genetically engineered mouse model. Suppressing STEAP4 via knockdown techniques effectively attenuated the nuclear factor erythroid 2-related factor 2 (NRF2)-NAD(P)H:quinone oxidoreductase 1 (NQO1) signaling pathway, inducing apoptosis and autophagy, leading to substantial reductions in xenograft tumor growth. In contrast, STEAP4 overexpression amplified ROS production and activated the NRF2-NQO1 pathway in a ferric iron (Fe3+)-dependent manner. Notably, bioactivatable drugs targeting NQO1 were highly effective at eradicating STEAP4-overexpressing colon cancer cells. These findings highlight the potential of targeted therapeutic interventions for CRC, particularly through STEAP4 modulation. In conclusion, our study advances understanding of the role of STEAP4 in colon tumorigenesis, offering promising avenues for novel CRC treatments.
Collapse
Affiliation(s)
- Kunlun Yin
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Luke Villareal
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Xiangxiang Wu
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Mariella Arcos
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Jordan Lee
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - David R Martin
- Department of Pathology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Julie G In
- Division of Gastroenterology, Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Kimberly Leslie
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Donna D Zhang
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL 33458, USA
| | - Xiang Xue
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM 87131, USA
| |
Collapse
|
12
|
Xia B, Zhu Q. Aptamer-ODN Chimeras: Enabling Cell-Specific ODN Targeting Therapy. Cells 2025; 14:697. [PMID: 40422200 DOI: 10.3390/cells14100697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/23/2025] [Accepted: 05/10/2025] [Indexed: 05/28/2025] Open
Abstract
Oligonucleotides (ODNs) such as siRNA, saRNA, and miRNA regulate gene expression through a variety of molecular mechanisms and show unique potential in the treatment of genetic diseases and rare diseases, but their clinical application is still limited by the efficiency of the delivery system, especially the problem of the insufficient targeting of extrahepatic tissues. As homologous nucleic acid molecules, aptamers have become a key tool to improve the targeted delivery of ODNs. Aptamer-ODN chimeras can not only bind to multiple proteins on the cell surface with high specificity and selectivity, but they can also internalize into cells. Furthermore, they outperform traditional delivery systems in terms of cost-effectiveness and chemical modification flexibility. This review systematically summarizes the origin and progress of aptamer-ODN chimera therapy, discusses some innovative design strategies, and proposes views on the future direction of aptamer-ODN chimeras.
Collapse
Affiliation(s)
- Bei Xia
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Qubo Zhu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| |
Collapse
|
13
|
Sristi, Gupta G, Abourehab MAS, Sahebkar A, Kesharwani P. Recent advances in PD-L1 siRNA nanocarriers for cancer therapy. Int J Biol Macromol 2025; 311:143994. [PMID: 40339860 DOI: 10.1016/j.ijbiomac.2025.143994] [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: 03/20/2025] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Tumor immune evasion depends on the programmed death-ligand 1 (PD-L1) mechanism, making it a prominent target in cancer therapy. Small interfering RNA (siRNA) designed to inhibit PD-L1 expression presents an innovative approach for boosting immunity against tumors. However, the therapeutic use of siRNA faces challenges, primarily due to its instability and inefficient cellular delivery. Recent advancements in nanocarrier technologies have shown promise in overcoming these obstacles, improving the delivery and efficacy of PD-L1 siRNA. This review comprehensively explores various nanocarrier systems, including lipid nanoparticles, polymeric carriers, and inorganic nanoparticles, highlighting their design innovations and applications in targeting PD-L1 in diverse cancer models. We discuss the synergistic effects of PD-L1 siRNA delivered via nanocarriers in conjunction with chemotherapy and immunomodulators, showcasing their potential to boost immune responses and reduce tumor growth. Additionally, we address ongoing challenges such as optimizing biodistribution and minimizing off-target effects, which hinder clinical translation. By synthesizing recent research findings, this review aims to illuminate the transformative potential of PD-L1 siRNA nanocarriers in cancer immunotherapy, paving the way for future studies aimed at enhancing therapeutic strategies and improving patient outcomes.
Collapse
Affiliation(s)
- Sristi
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Sector 125, Noida 201301, UP, India
| | - Garima Gupta
- Graphic Era Hill University, Dehradun 248002, India; School of Allied Medical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Mohammed A S Abourehab
- Department of Pharmaceutical Sciences, College of Pharmacy, Umm Al Qura University, Makkah, Saudi Arabia
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| |
Collapse
|
14
|
Jeon JY, Ayyar VS, Ouchi S, Fabbrini E, Koshkina A, Prusakiewicz JJ, Dallas J, Yang T, Jian W, Kang L, Cofsky K, Rady B, Tamamura R, Saito Y, Yamashita A, Vaughan T, Wendel S, Makimura H, Csonka D, Goyal N. Preclinical and Clinical Pharmacokinetics of JNJ-75220795, an siRNA Therapeutic Targeting PNPLA3, for Metabolic Dysfunction-Associated Steatohepatitis. J Clin Pharmacol 2025; 65:644-653. [PMID: 39654352 DOI: 10.1002/jcph.6174] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/21/2024] [Indexed: 04/29/2025]
Abstract
JNJ-75220795 or ARO-PNPLA3 is an investigational small interfering ribonucleic acid agent conjugated with N-acetyl-d-galactosamine that targets the PNPLA3 gene, currently being developed for metabolic dysfunction-associated steatohepatitis (MASH). This study evaluated the pharmacokinetics (PK) profile of single subcutaneous doses of JNJ-75220795 in preclinical species as well as in human subjects with homozygous or heterozygous PNPLA3 I148M mutation in two phase 1 studies-a first-in-human study in the United States and a first-in-Japanese study in Japan. Preclinical PK in rats and non-human primates (NHP) showed a rapid systemic absorption and elimination following single subcutaneous doses. JNJ-75220795 was predominantly distributed to the liver with peak liver concentrations reached at 4 h and still detectable at 672 and 336 h in rats and NHPs, respectively, with an apparent liver-to-plasma area under the curve (AUC) ratio of 2800 in rats. Consistent with the preclinical findings, clinical PK showed rapid systemic absorption and clearance in humans with median peak concentrations at 3.0-9.0 h and mean short half-life of 3.4-6.2 h. Plasma PK exposure parameters including Cmax and AUC increased approximately dose proportionally. Kidney had the second highest tissue exposure following the liver in rats. Renal excretion was a significant but minor elimination pathway as approximately 15%-25% of the administered dose was recovered in urine. Based on the overall data, JNJ-75220795 was primarily localized to the liver and exhibited sustained hepatic exposures, which confer prolonged pharmacodynamic effects in the target organ. The favorable PK profiles of single subcutaneous doses observed in the phase 1 studies support continued clinical development of JNJ-75220795 for the treatment of MASH.
Collapse
Affiliation(s)
- Jae Yoon Jeon
- Janssen Research & Development, LLC, Raritan, NJ, USA
| | | | | | | | | | | | - Jed Dallas
- Arrowhead Pharmaceuticals, Inc., Madison, WI, USA
| | - Txheng Yang
- Arrowhead Pharmaceuticals, Inc., Madison, WI, USA
| | - Wenying Jian
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Lijuan Kang
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Korin Cofsky
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Brian Rady
- Janssen Research & Development, LLC, Spring House, PA, USA
| | | | - Yuki Saito
- Janssen Pharmaceutical K.K., Tokyo, Japan
| | | | | | - Susan Wendel
- Janssen Research & Development, LLC, Raritan, NJ, USA
| | | | - Dénes Csonka
- Actelion Pharmaceuticals Ltd., Allschwil, Switzerland
| | - Navin Goyal
- Janssen Research & Development, LLC, Raritan, NJ, USA
| |
Collapse
|
15
|
Jain N, Roy AA, Madhusoodanan G, Preman NK, Pokale R, Pisay M, Mukharya A, Pandey A, Mutalik S. Unlocking the future: Precision oligonucleotide therapy for targeted treatment of neurodegenerative disorders. Int J Biol Macromol 2025; 310:143515. [PMID: 40288719 DOI: 10.1016/j.ijbiomac.2025.143515] [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: 03/20/2025] [Revised: 04/09/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
Neurodegenerative disorders are complex and devastating conditions of the central nervous system that profoundly impact quality of life. Given the limited treatment options available, there is a pressing need to develop novel therapeutic strategies. Oligonucleotides have emerged as key players in precision medicine for these disorders, but their potential is hindered by poor translocation across the blood-brain barrier. This review focuses on neurodegenerative disorders other than Alzheimer's and Parkinson's, which are widely reported in the literature, and aims to address the significant hurdles in oligonucleotide delivery for neurodegenerative diseases. It highlights recent advancements in CNS-targeting approaches, such as chemical conjugation, antibody-oligonucleotide conjugates, focused ultrasound, and viral and nanocarrier-based delivery systems. Each strategy's strengths and limitations are discussed, with potential solutions proposed for more effective treatments. Additionally, the review offers valuable insights into regulatory requirements and prospects for clinical translation, which are crucial for shaping the future of neurodegenerative therapies. By exploring these innovative approaches, the goal is to surmount challenges posed by the blood-brain barrier and develop more effective treatments, thereby enhancing the quality of life of the patients suffering from these debilitating conditions.
Collapse
Affiliation(s)
- Naitik Jain
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Amrita Arup Roy
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Geethu Madhusoodanan
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Namitha K Preman
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Rahul Pokale
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Muralidhar Pisay
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Anoushka Mukharya
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Abhijeet Pandey
- Novartis Healthcare Private Ltd, Novartis Knowledge Center, Hyderabad 500081, Telangana, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
16
|
Khandal J, Dohare S, Dongsar TS, Gupta G, Alsayari A, Wahab S, Kesharwani P. Gelatin nanocarriers in oncology: A biocompatible strategy for targeted drug delivery. Int J Biol Macromol 2025; 310:143244. [PMID: 40250682 DOI: 10.1016/j.ijbiomac.2025.143244] [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: 03/18/2025] [Revised: 04/06/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Cancer persists as a formidable global health crisis, with conventional therapies often compromised by systemic toxicity, poor tumor specificity, and therapeutic resistance. Nanotechnology has emerged as a transformative approach, leveraging nanoscale materials to enhance drug bioavailability, enable targeted delivery, and mitigate off-target effects. Among these innovations, gelatin-based nanoparticles (GNPs), derived from collagen and endorsed by the FDA have garnered significant attention as biocompatible, biodegradable nanocarriers uniquely suited for oncology applications. GNPs address critical extracellular barriers such as inefficient tumor penetration, rapid clearance, and nonspecific biodistribution by capitalizing on gelatin's intrinsic advantages: low immunogenicity, tumor microenvironment responsiveness (pH, enzymes, redox gradients), and tunable surface functionalization. This review highlights the versatility of GNPs in overcoming these challenges through advanced strategies like ligand-mediated targeting, combinatorial therapies, and size-transformable systems that enhance tumor accumulation and therapeutic precision. Case studies across lung, breast, skin, liver, colorectal, brain, and head/neck cancers demonstrate GNPs' ability to reduce IC50 values by 2 to 4-fold, achieve >90 % apoptosis in malignant cells, and minimize damage to healthy tissues. Despite the challenges in translating gelatin-based nanocarriers from preclinical studies to clinical applications in cancer therapy, their promising preclinical performance highlights their potential as patient-centric platforms capable of advancing precision oncology. Further their adaptability, multifunctionality, and capacity for stimuli-responsive drug release underscore their potential to improve clinical outcomes, offering a targeted, low-toxicity paradigm for managing diverse malignancies.
Collapse
Affiliation(s)
- Jayesh Khandal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Shubham Dohare
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Tenzin Sonam Dongsar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Garima Gupta
- Graphic Era Hill University, Dehradun 248002, India; School of Allied Medical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| |
Collapse
|
17
|
Chen W, Wang H, Yao Y, Du G, Sun Q, Lin Y, Cai X. Multi-Route Administration Delivery System Based on Tetrahedral Framework Nucleic Acids for Alleviating Type I Hypersensitivity Reactions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408104. [PMID: 40269571 DOI: 10.1002/smll.202408104] [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: 09/08/2024] [Revised: 02/26/2025] [Indexed: 04/25/2025]
Abstract
Type I hypersensitivity reactions are the most common type of hypersensitivity reactions and can cause various symptoms in different body systems. Currently, there is no standard treatment for the different manifestations of type I hypersensitivity reactions at different sites. To address this challenge, a nanocomposite named tFNAs-siRNA-siRNA (TSS) is designed based on tetrahedral framework nucleic acids (tFNAs). It is loaded with two siRNAs and can enter the body through multi-route administration. In terms of the realization of siRNAs' functions, TSS successfully delivers them and downregulates the expression of Gab2 and Syk, thereby inhibiting the migration and release of granules to the extracellular space in RBL-2H3 cells. Interestingly, TSS also exhibits the effect of hindering Ca2+ efflux from mitochondria to stabilize energy metabolism. In the passive cutaneous anaphylaxis and allergic rhinitis model, TSS shows excellent anti-allergic ability, and demonstrates an advantage in intraperitoneal injection, transdermal, and transnasal routes of administration. In sum, TSS offers a new therapeutic strategy for type I hypersensitivity reactions with multiple symptoms, highlighting the application potential of nucleic acids-based multi-route administration delivery system.
Collapse
Affiliation(s)
- Wen Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Hao Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Yangxue Yao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Guannan Du
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Qiang Sun
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
18
|
Shiryaeva O, Tolochko C, Alekseeva T, Dyachuk V. Targets and Gene Therapy of ALS (Part 1). Int J Mol Sci 2025; 26:4063. [PMID: 40362304 PMCID: PMC12071412 DOI: 10.3390/ijms26094063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/16/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the selective death of motor neurons, which causes muscle atrophy. Genetic forms of ALS are recorded only in 10% of cases. However, over the past decade, studies in genetics have substantially contributed to our understanding of the molecular mechanisms underlying ALS. The identification of key mutations such as SOD1, C9orf72, FUS, and TARDBP has led to the development of targeted therapy that is gradually being introduced into clinical trials, opening up a broad range of opportunities for correcting these mutations. In this review, we aimed to present an extensive overview of the currently known mechanisms of motor neuron degeneration associated with mutations in these genes and also the gene therapy methods for inhibiting the expression of their mutant proteins. Among these, antisense oligonucleotides, RNA interference (siRNA and miRNA), and gene-editing (CRISPR/Cas9) methods are of particular interest. Each has shown its efficacy in animal models when targeting mutant genes, whereas some of them have proven to be efficient in human clinical trials.
Collapse
Affiliation(s)
| | | | | | - Vyacheslav Dyachuk
- Almazov Federal Medical Research Centre, 197341 Saint Petersburg, Russia; (O.S.); (C.T.); (T.A.)
| |
Collapse
|
19
|
Choi AS, Moon TJ, Bhalotia A, Rajan A, Ogunnaike L, Hutchinson DW, Hwang I, Gokhale A, Kim JN, Ma T, Karathanasis E. Lipid Nanoparticles and PEG: Time Frame of Immune Checkpoint Blockade Can Be Controlled by Adjusting the Rate of Cellular Uptake of Nanoparticles. Mol Pharm 2025; 22:1859-1868. [PMID: 40035231 PMCID: PMC11975481 DOI: 10.1021/acs.molpharmaceut.4c01039] [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: 03/05/2025]
Abstract
The engineerability of lipid nanoparticles (LNPs) and their ability to deliver nucleic acids make LNPs attractive tools for cancer immunotherapy. LNP-based gene delivery can be employed for various approaches in cancer immunotherapy, including encoding tumor-associated antigens and silencing of negative immune checkpoint proteins. For example, LNPs carrying small interfering RNAs can offer several advantages, including sustained and durable inhibition of an immune checkpoint protein. Due to their tunable design, modifying the lipid composition of LNPs can regulate the rate of their uptake by immune cells and the rate of gene silencing. Controlling the kinetics of LNP uptake provides additional flexibility and strategies to generate appropriate immunomodulation in the tumor microenvironment. Here, we evaluated the effects of polyethylene glycol (PEG) content ranging from 0.5 to 6 mol % on the cellular uptake of LNPs by immune cells and gene silencing of PD-L1 after intratumoral administration. We evaluated the cellular uptake and PD-L1 blockade in vitro in cell studies and in vivo using the YUMM1.7 melanoma tumor model. Cell studies showed that the rate of cell uptake was inversely correlated to an increasing mol % of PEG in a linear relationship. In the in vivo studies, 0.5% PEG LNP initiated an immediate effect in the tumor with a significant decrease in the PD-L1 expression of immune cells observed within 24 h. In comparison, the gene silencing effect of 6% PEG LNP was delayed, with a significant decrease of PD-L1 expression in immune cell subsets being observed 72 h after administration. Notably, performance of the 6% PEG LNP at 72 h was comparable to that of the 0.5% PEG LNP at 24 h. Overall, this study suggests that PEG modifications and intratumoral administration of LNPs can be a promising strategy for an effective antitumor immune response.
Collapse
Affiliation(s)
- Andrew S Choi
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Taylor J Moon
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Anubhuti Bhalotia
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Aarthi Rajan
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Laolu Ogunnaike
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Diarmuid W Hutchinson
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Inga Hwang
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Aaditya Gokhale
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Justin N Kim
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Timothy Ma
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Efstathios Karathanasis
- Department of Biomedical engineering, School of Medicine, Case Western Reserve University, Cleveland, OH
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
20
|
Guo J, Qu H, Cui P, Xue Y. USP33 PROMOTES CERULEIN-INDUCED APOPTOTIC, OXIDATIVE, AND INFLAMMATORY INJURIES IN ACUTE PANCREATITIS BY DEUBIQUITINATING TRAF3. Shock 2025; 63:559-565. [PMID: 39637362 DOI: 10.1097/shk.0000000000002514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
ABSTRACT Background: Tumor necrosis factor receptor associated factor 3 (TRAF3) and deubiquitinating enzyme ubiquitin-specific protease 33 (USP33) have been identified to play important roles in inflammatory diseases, including acute pancreatitis (AP). Here, we aimed to explore whether USP33 affected AP progression by affecting TRAF3 expression through deubiquitination. Methods: Cerulein-treated HPDE6-C7 cells were used to mimic AP conditions in vitro . Levels of mRNAs and proteins were examined by qRT-PCR and western blot. Cell proliferation and apoptosis were evaluated using CCK-8 assay, EdU assay, and flow cytometry. Cell oxidative stress was assessed by detecting the production of superoxide dismutase and malonaldehyde. ELISA analysis detected IL-6 and TNF-α levels. Macrophage M1 polarization was evaluated by flow cytometry. Cellular ubiquitination analyzed the ubiquitination effect on TRAF3. Protein interaction between USP33 and TRAF3 was identified by immunofluorescence staining. Results: Cerulein dose-dependently induced apoptosis, oxidative stress, and inflammatory response in HPDE6-C7 cells and promoted macrophage M1 polarization to enhance inflammation ( P < 0.05). TRAF3 was highly expressed in AP patients (3.5±1.10 vs. 1.0 ±0.74, P < 0.05) and cerulein-induced HPDE6-C7 cells (3.3 ±0.34 vs. 1.0 ±0.10, P < 0.05). Knockdown of TRAF3 protected HPDE6-C7 cells from cerulein-induced apoptotic, oxidative and inflammatory injuries. Mechanistically, USP33 interacted with TRAF3 and induced TRAF3 deubiquitination to upregulate its expression ( P < 0.05). Further analyses showed that USP33 knockdown reversed cerulein-induced apoptosis, oxidative stress and inflammation in HPDE6-C7 cells by TRAF3 ( P < 0.05). Moreover, USP33-TRAF3 activated the NF-κB pathway ( P < 0.05). Conclusion: USP33 promoted cerulein-induced apoptosis, oxidative stress and inflammation in pancreatic ductal cells by deubiquitinating TRAF3, indicating a novel insight into the pathogenesis of AP.
Collapse
Affiliation(s)
- Jian Guo
- Department of general Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Huiheng Qu
- Department of General Surgery, JiangNan University Medical Center, Wuxi, JiangSu, China
| | - Peng Cui
- Department of general Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Yu Xue
- Department of general Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
21
|
Clua A, Fàbrega C, Musumeci D, Doria F, Pirota V, Ghirga F, Platella C, Eritja R, Montesarchio D. On the interaction of anticancer G-quadruplex ligands with GalNAc-functionalized G-quadruplex-forming carriers for selective recognition and treatment of hepatocellular carcinoma. Bioorg Chem 2025; 157:108299. [PMID: 40015110 DOI: 10.1016/j.bioorg.2025.108299] [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: 10/04/2024] [Revised: 02/03/2025] [Accepted: 02/18/2025] [Indexed: 03/01/2025]
Abstract
Lack of specificity towards cancer cells is a major drawback of most chemotherapeutic agents. The use of selective drug delivery systems capable of targeting cancer cells is a valuable perspective to overcome the serious adverse effects often associated with conventional treatments. In this frame, N-acetylgalactosamine (GalNAc)-functionalized G-quadruplex-forming oligonucleotides represent promising delivery systems due to their ability to selectively recognize the asialoglycoprotein receptor (ASGPR) overexpressed on the surface of hepatocellular carcinoma cells. Here, we investigated the interaction of two small molecules, previously proved to induce cancer cell death by selective recognition of cancer-related G-quadruplexes, i.e., the trifunctionalized naphthalene diimide NDI-5 and the alkaloid Dicentrine, with a set of chemically different GalNAc-functionalized G-quadruplexes, some of them derivatized with floxuridine units, here chosen as suitable drug carriers. Several biophysical techniques, such as fluorescence spectroscopy, circular dichroism and gel electrophoresis, were exploited to characterize these systems and evaluate their stability in pseudo-physiological solutions. In addition, the cytotoxicity of the best ligand/GalNAc-functionalized G-quadruplex complexes was evaluated in hepatic and cervical cancer cell lines, using a normal cell line as control, to assess the selective anticancer effects of our delivered drugs and, more in detail, their selectivity of action against the hepatic cancer cells. Taken together, the obtained results demonstrated the high potency and synergistic effects of ligand/GalNAc-functionalized G-quadruplex complexes as effective and selective anticancer drugs delivery systems, especially for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Anna Clua
- Nucleic Acids Chemistry Group, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain; Nucleic Acids Chemistry Group, Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08034 Barcelona, Spain
| | - Carme Fàbrega
- Nucleic Acids Chemistry Group, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain; Nucleic Acids Chemistry Group, Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08034 Barcelona, Spain
| | - Domenica Musumeci
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Filippo Doria
- Department of Chemistry, University of Pavia, 27100 Pavia, Italy
| | - Valentina Pirota
- Department of Chemistry, University of Pavia, 27100 Pavia, Italy
| | - Francesca Ghirga
- Department of Chemistry and Technology of Drugs, Sapienza University of Rome, 00185 Rome, Italy
| | - Chiara Platella
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy.
| | - Ramon Eritja
- Nucleic Acids Chemistry Group, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain; Nucleic Acids Chemistry Group, Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08034 Barcelona, Spain.
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| |
Collapse
|
22
|
Wang M, Wang Y, Zhang P, Gu C, Zhao X, Gong X, Yang X, Pan J, Xi Y. Neutrophil-like cell membrane-coated metal-organic frameworks for siRNA delivery targeting NOX4 to alleviate oxidative stress in acute ischemic injury. Acta Biomater 2025; 196:487-505. [PMID: 40024424 DOI: 10.1016/j.actbio.2025.02.061] [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: 11/30/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Although reperfusion is the most effective treatment for acute ischemic stroke, it often results in serious secondary ischemia/reperfusion (I/R) injury due to oxidative stress. This oxidative stress primarily results from the overproduction of reactive oxygen species (ROS) during reperfusion which, in turn, is largely induced by high expression of NADPH oxidase 4 (NOX4). Inhibiting NOX4 gene expression has therefore been proposed as a direct approach to reduce ROS production and promote angiogenesis. Recognizing both the potential of siRNA-based therapies for selective gene silencing and the critical role of neutrophil-endothelial interactions during I/R injury, here we present a unique therapeutic approach where neutrophil-like cell membrane coated porous metal-organic framework nanoparticles are loaded with siNOX4 (M-MOF-siNOX4) and designed to target damaged brain microvascular tissue. These then mitigate oxidative stress by suppressing NOX4 expression. Using an in vitro oxygen-glucose deprivation/re-oxygenation model, we demonstrate that M-MOF-siNOX4 nanoparticles specifically bind to activated endothelial cells, effectively reducing NOX4 expression, decreasing both ROS production and cell apoptosis, and restoring cell viability. Use of an in vivo mouse model of middle cerebral artery occlusion further confirmed M-MOF-siNOX4 nanoparticles to substantially alleviate brain damage and protect neurological function following ischemic stroke. Taken together, our study presents an innovative and effective siRNA-based strategy for reducing oxidative stress in ischemic stroke therapy. STATEMENT OF SIGNIFICANCE: Ischemia/reperfusion (I/R) injury, a major complication of acute ischemic stroke, is primarily driven by oxidative stress due to the excessive production of reactive oxygen species (ROS). Current treatments targeting oxidative stress and cell death often lack specificity, leading to off-target effects. This study introduces an innovative nanoparticle-based therapy using neutrophil-like cell membrane-coated metal-organic frameworks (MOFs) to deliver siNOX4, an siRNA targeting NOX4, a key ROS-producing enzyme. This approach enhances targeted delivery, reduces ROS production and cell death, and significantly improves neurological recovery in stroke models. By overcoming the limitations of existing therapies, this strategy holds strong potential for revolutionizing ischemic stroke treatment and addressing other disorders related to oxidative stress.
Collapse
Affiliation(s)
- Min Wang
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China
| | - Yunbo Wang
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China
| | - Pengqi Zhang
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China
| | - Chenjie Gu
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Xianlei Zhao
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China
| | - Xinghan Gong
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China
| | - Xiaohang Yang
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China.
| | - Jianwei Pan
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| | - Yongmei Xi
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
23
|
Su Y, Jin Y. A narrative review of papillary thyroid carcinoma-related long non-coding RNAs and their relevance to malignant tumors. Transl Cancer Res 2025; 14:2125-2149. [PMID: 40224997 PMCID: PMC11985200 DOI: 10.21037/tcr-24-1038] [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: 06/22/2024] [Accepted: 01/14/2025] [Indexed: 04/15/2025]
Abstract
Background and Objective In recent years, research on the relationship between papillary thyroid carcinoma (PTC) and long non-coding RNAs (lncRNAs) has been burgeoning. However, there has not been an analysis of the regulatory mechanisms of these lncRNAs in all tumors, nor a comprehensive categorization and comparison of these mechanisms. This review aims to uncover whether PTC-related lncRNAs also play an important role in other tumors and to identify a common pattern of action. Methods We conducted a statistical analysis of lncRNAs related to PTC that have been reported during the period from Jan 2022 to May 2024 through searching in the Embase, Web of Science, and PubMed databases, focusing on those with greater research value. Using them as the focal points of our study, we compiled data on their different regulatory mechanisms across various malignant tumors, emphasizing key findings. Key Content and Findings This comprehensive analysis not only provides valuable insights into potential regulatory mechanisms of these lncRNAs in PTC but also serves as a reference for exploring their broader regulatory networks within cancer. The principal discovery is that lncRNAs associated with PTC can competitively interact with microRNAs (miRNAs). This interaction influences miRNA-targeted messenger RNA (mRNA) and the expression of cancer-related proteins, ultimately facilitating the progression of PTC as well as other malignant tumors. Conclusions The lncRNAs associated with PTC exert regulatory functions in other malignancies as well and possess similar regulatory mechanisms. This provides a molecular basis for the future development of relevant targeted therapies.
Collapse
Affiliation(s)
- Yuanhao Su
- Department of General Surgery, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yi Jin
- Cell and Gene Research Therapy Institute, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
24
|
Zhang Y, Zang C, Mao M, Zhang M, Tang Z, Chen W, Zhu W. Advances in RNA therapy for the treatment of autoimmune diseases. Autoimmun Rev 2025; 24:103753. [PMID: 39842534 DOI: 10.1016/j.autrev.2025.103753] [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] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
Autoimmune diseases (ADs) are a group of complex, chronic conditions characterized by disturbance of immune tolerance, with examples including systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, and psoriasis. These diseases have unclear pathogenesis, and traditional therapeutic approaches remain limited. However, advances in high-throughput histology technology and scientific discoveries have led to the identification of various pathogenic factors contributing to ADs. Coupled with improvements in RNA nucleic acid-based drug synthesis, design, and delivery, RNA-based therapies have been extensively investigated for their potential in treating ADs. This paper reviews the progress in the use of miRNAs, lncRNAs, circRNAs, siRNAs, antisense oligonucleotides (ASOs), aptamers, mRNAs, and other RNA-based therapies in ADs, focusing on their therapeutic potential and application prospects, providing insights for future research and clinical treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Ying Zhang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Chenyang Zang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Manyun Mao
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Mi Zhang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Zhenwei Tang
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wangqing Chen
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China.
| | - Wu Zhu
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China.
| |
Collapse
|
25
|
Osawa T, Nakanishi R, Uda K, Muramoto S, Obika S. Synthesis and structural analysis of dinucleotides containing 2',3'-trans-bridged nucleic acids with trans-5,6- or 5,7-fused ring skeleton. Commun Chem 2025; 8:87. [PMID: 40121355 PMCID: PMC11929919 DOI: 10.1038/s42004-025-01486-2] [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: 10/31/2024] [Accepted: 03/06/2025] [Indexed: 03/25/2025] Open
Abstract
Artificial nucleic acids in which the conformation of the sugar or phosphate backbone of the oligonucleotide is appropriately fixed can form stable duplexes. In this study, we designed dinucleotides containing 2',3'-trans-bridged nucleic acids (2',3'-trans-BNAs) based on the idea that the sugar conformation and torsions angles δ, ε, ζ, α, and β of the backbone can be controlled by a 5,6- or 5,7-membered trans-fused ring skeleton cyclized between the 2'- and 3'-positions of the sugar moiety. Given that the construction of trans-5,6-fused ring skeletons is synthetically challenging, the synthesis was optimized and a detailed structural analysis of these new bridged 2',3'-trans-BNA systems was conducted. The 2',3'-trans-BNAs could be synthesized from a commercially available D-glucose derivative with the key intramolecular gold-catalyzed cyclization reaction achieved using a cyclization precursor bearing an intramolecular hydroxy group and an internal alkyne. Structural analysis of the 2',3'-trans-BNAs showed an N-type sugar conformation for all the derivatives, which is similar to that in RNA-duplex, and the ζ and α torsion angles for the 2',3'-trans-BNAs were a characteristic feature of the compounds that differ from the corresponding angles of the natural duplexes.
Collapse
Affiliation(s)
- Takashi Osawa
- Graduate School of Pharmaceutical Science, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ryota Nakanishi
- Graduate School of Pharmaceutical Science, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Keito Uda
- Graduate School of Pharmaceutical Science, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - So Muramoto
- Graduate School of Pharmaceutical Science, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Satoshi Obika
- Graduate School of Pharmaceutical Science, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
26
|
Chen L, Xu L, Zhang Y, Xia H. Dioscin alleviates the dysfunction of fibroblast-like synoviocytes by circ_0008267/miR-942-5p/FKBP5 axis during rheumatoid arthritis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03872-y. [PMID: 40116870 DOI: 10.1007/s00210-025-03872-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/31/2025] [Indexed: 03/23/2025]
Abstract
Dioscin is a natural, bioactive steroid saponin that has the antiarthritic activity. Circular RNAs (circRNAs) are stable noncoding RNAs involving in the pathogenesis of rheumatoid arthritis (RA). Here, this study aimed to probe the role and mechanism of dioscin and circ_0008267 in RA progression. Cell proliferation, apoptosis, invasive, and migratory abilities, as well as inflammatory response were evaluated by CCK-8 assay, EdU assay, flow cytometery, transwell assay, wound healing assay, and ELISA analysis, respectively. Levels of genes and protein were tested by qRT-PCR and western blotting. The interaction between miR-942-5p and circ_0008267 or FK506-binding protein 5 (FKBP5) was confirmed using dual-luciferase reporter and RNA pull-down assays. Dioscin treatment was demonstrated to suppress RA-FLS proliferation, invasion, migration, and inflammatory response, but induced cell apoptosis. Circ_0008267 is a stable circRNA, and was increased in RA samples. Moreover, its expression was reduced by dioscin in RA-FLS, overexpression of circ_0008267 reversed the effects of dioscin on RA-FLS. Mechanistically, circ_0008267 acted as a sponge for miR-942-5p, which targeted FKBP5. Dioscin reduced FKBP5 expression, but elevated miR-942-5p level in RA-FLS. MiR-942-5p inhibition or FKBP5 upregulation abolished the inhibitory effects of dioscin on RA-FLS dysfunction. Moreover, circ_0008267 deficiency impaired RA-FLS proliferation, invasion, migration, and inflammation through regulating FKBP5. Dioscin suppressed the proliferation, invasion, migration, and inflammatory response in RA-FLS via circ_0008267/miR-942-5p/FKBP5 axis, providing new insights for RA prevention.
Collapse
Affiliation(s)
- Lifeng Chen
- Department of Rheumatology, General Hospital of Central Theater Command, No. 627 Wuyi Road, Wuchang District, Wuhan, 430070, Hubei, China.
| | - Li Xu
- Department of Cardiovascular Medicine, Guiqian International General Hospital, Guiyang City, 550024, Guizhou Province, China
| | - Yujing Zhang
- Department of Rheumatology, General Hospital of Central Theater Command, No. 627 Wuyi Road, Wuchang District, Wuhan, 430070, Hubei, China
| | - Hao Xia
- Medical College of Wuhan University of Science and Technology, Wuhan, 430070, Hubei, China
| |
Collapse
|
27
|
Yang G, Huang X. LncRNA MALAT1's role in the development of retinopathy: A review. Medicine (Baltimore) 2025; 104:e41954. [PMID: 40128064 PMCID: PMC11936551 DOI: 10.1097/md.0000000000041954] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 03/06/2025] [Indexed: 03/26/2025] Open
Abstract
Long noncoding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) and retinopathy are 2 distinct yet interconnected areas of research in the field of ocular studies. MALAT1, with its diverse biological functions, has been extensively studied and demonstrated to play a role in various diseases, including ocular pathologies. Its involvement in alternative splicing regulation, transcriptional control, and the competing endogenous RNA (ceRNA) network suggests its potential implication in retinopathy. Retinopathy refers to a group of disorders that affect the retina, leading to vision impairment and, in severe cases, even blindness. These conditions include diabetic retinopathy, retinoblastoma, proliferative vitreoretinopathy, retinopathy of prematurity, and retinal neurodegeneration. The understanding of the molecular mechanisms underlying the development and progression of retinopathy, along with the potential involvement of MALAT1, can provide valuable insights for the diagnosis and treatment of this condition. Retinopathy, characterized by various manifestations and underlying mechanisms, presents a significant challenge in the field of ophthalmology. As a complex disease, its pathogenesis involves multifactorial factors, including angiogenic dysregulation, inflammatory responses, oxidative stress, and cellular signaling abnormalities. The emerging role of long noncoding RNA MALAT1 in retinopathy has attracted considerable attention. MALAT1 has been found to participate in multiple cellular processes, including alternative splicing regulation and transcriptional control. Additionally, the competing endogenous RNA (ceRNA) network involving MALAT1 indicates its potential relevance as a regulator in retinopathy. Further investigations into the specific mechanisms underlying MALAT1's involvement in retinopathy pathogenesis may provide valuable insights into the development of diagnostic and therapeutic approaches for managing retinal disorders.
Collapse
Affiliation(s)
- Gukun Yang
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, PR China
- Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, PR China
| | - Xionggao Huang
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, PR China
- Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, PR China
| |
Collapse
|
28
|
Harshita, Harish V, Upendra SL, Mohd S, Singh SK, Agrawal P, Vishwas S, Dua K. Next-Gen Cancer Treatment: Nanotechnology-Driven siRNA Delivery Solutions. Assay Drug Dev Technol 2025. [PMID: 40080397 DOI: 10.1089/adt.2024.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025] Open
Abstract
RNA interference through small interfering RNA (siRNA) has shown great promise as a potential cancer treatment strategy in recent years. However, the delivery of siRNA to target cancer cells efficiently remains a significant challenge. This review aims to highlight the recent advances in nanotechnology-enabled siRNA delivery for cancer treatment, bridging the gap between bench research and clinical application. A comprehensive literature search was conducted to identify recent studies focused on the utilization of nanotechnology for siRNA delivery in cancer treatment. Key databases, including PubMed, Scopus, and Web of Science, were used, and relevant articles were screened. Several nanotechnology-based platforms for siRNA delivery have emerged in recent years, providing enhanced selectivity, improved stability, and controlled release profiles. The primary types of nanocarriers discussed include lipid-based nanoparticles, inorganic nanoparticles, polymeric nanoparticles, and exosomes. Nanotechnology-based siRNA delivery systems represent a promising avenue for cancer treatment. Although significant progress has been made in preclinical studies, translating these findings to clinical applications poses several challenges, including scale-up production, safety, and targeted delivery. Nevertheless, the recent developments in this field hold great promise in revolutionizing cancer therapy, providing hope for more effective and personalized treatment options in the future.
Collapse
Affiliation(s)
- Harshita
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Vancha Harish
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Sakshi Lad Upendra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Sharfuddin Mohd
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Pooja Agrawal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| |
Collapse
|
29
|
Ariyoshi J, Asanuma H, Kamiya Y. Protocol for Controlling the Strand Selectivity of siRNA Using Acyclic Artificial Nucleic Acids. Curr Protoc 2025; 5:e70103. [PMID: 40110740 DOI: 10.1002/cpz1.70103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Small interfering RNA (siRNA) has emerged as a promising therapeutic candidate against previously intractable diseases. An effective siRNA must have high on-target activity while off-target effects are minimized. This balance can be achieved by enhancing the selectivity of the antisense strand through sequence optimization and appropriate chemical modifications. Acyclic artificial nucleic acids such as serinol nucleic acids (SNA) have demonstrated on-target activity while suppressing off-target effects. This article provides guidelines for designing SNA-modified siRNA and outlines a method for the experimental evaluation of the on-target and off-target activities of siRNAs, ensuring accurate functional validation in cell systems. These protocols benefit researchers developing siRNA-based therapeutics to optimize siRNA selectivity and efficacy while minimizing off-target effects through innovative design strategies. © 2025 Wiley Periodicals LLC. Basic Protocol 1: Design of SNA-modified siRNA Basic Protocol 2: Design and preparation of vector plasmids using inverse PCR Alternate Protocol: Design and preparation of vector plasmid using restriction enzymes and ligase Basic Protocol 3: Evaluation of the on- and off-target effects of siRNAs using the dual-luciferase assay Support Protocol 1: Agarose gel electrophoresis and protocol for purifying DNA from gels Support Protocol 2: Transformation and amplification of plasmids.
Collapse
Affiliation(s)
- Jumpei Ariyoshi
- Laboratory of Bioanalytical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Hiroyuki Asanuma
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Yukiko Kamiya
- Laboratory of Bioanalytical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| |
Collapse
|
30
|
Anwar AA, Jalan-Sakrikar N, Huebert RC. LncRNAs, RNA Therapeutics, and Emerging Technologies in Liver Pathobiology. Semin Liver Dis 2025; 45:1-14. [PMID: 39603269 DOI: 10.1055/a-2490-1921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The field of ribonucleic acid (RNA) biology has revealed an array of noncoding RNA species, particularly long noncoding RNAs (lncRNAs), which play crucial roles in liver disease pathogenesis. This review explores the diverse functions of lncRNAs in liver pathology, including metabolic-associated steatotic liver disease, hepatocellular carcinoma, alcohol-related liver disease, and cholangiopathies such as primary sclerosing cholangitis and cholangiocarcinoma. We highlight key lncRNAs that regulate lipid metabolism, inflammation, fibrosis, and oncogenesis in the liver, demonstrating their diagnostic and therapeutic potential. Emerging RNA-based therapies, such as mRNA therapy, RNA interference, and antisense oligonucleotides, offer approaches to modulate lncRNA activity and address liver disease at a molecular level. Advances in sequencing technologies and bioinformatics pipelines are simultaneously enabling the identification and functional characterization of novel lncRNAs, driving innovation in personalized medicine. In conclusion, this review highlights the potential of lncRNAs as biomarkers and therapeutic targets in liver disease and emphasizes the need for further research into their regulatory mechanisms and clinical applications.
Collapse
Affiliation(s)
- Abid A Anwar
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Nidhi Jalan-Sakrikar
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota
- Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic and Foundation, Rochester, Minnesota
| | - Robert C Huebert
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota
- Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic and Foundation, Rochester, Minnesota
| |
Collapse
|
31
|
Liu Y, Zhang S, Zhang M, Liu X, Wu Y, Wu Q, Chaput J, Wang Y. Chemical evolution of ASO-like DNAzymes for effective and extended gene silencing in cells. Nucleic Acids Res 2025; 53:gkaf144. [PMID: 40037707 PMCID: PMC11879453 DOI: 10.1093/nar/gkaf144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/21/2025] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
Antisense oligonucleotides (ASOs) and small interfering RNA (siRNA) therapeutics highlight the power of oligonucleotides in silencing disease-causing messenger RNAs (mRNAs). Another promising class of gene-silencing oligonucleotides is RNA-cleaving nucleic acid enzymes, which offer the potential for allele-specific RNA inhibition with greater precision than ASOs and siRNAs. Herein, we chemically evolved the nucleolytic DNA enzyme (DNAzyme) 10-23, by incorporating the modifications that are essential to the success of ASO drugs, including 2'-fluoro, 2'-O-methyl, and 2'-O-methoxyethyl RNA analogues, and backbone phosphorothioate, to enhance catalytic efficiency by promoting RNA substrate binding and preventing dimerization of 10-23. These ASO-like DNAzymes cleaved structured RNA targets in long transcripts, showed prolonged intracellular stability, and downregulated mRNA and protein levels of both exogenously transfected eGFP and endogenously elevated oncogenic c-MYC. In colon cancer HCT116 cells, the downregulation of oncogenic c-MYC RNA resulted in cell cycle arrest, reduced proliferation, and increased apoptosis. RACE (rapid amplification of cDNA ends) polymerase chain reaction and Sanger sequencing confirmed precise, site-specific mRNA transcript cleavage with minimal RNase H activation in cells. By merging ASO structural and pharmacokinetic advantages with DNAzyme catalytic versatility, these ASO-like 10-23 variants offer a promising new class of potent gene-silencing agents, representing a significant step toward therapeutic DNAzyme development.
Collapse
Affiliation(s)
- Yingyu Liu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310000, China
| | - Sheyu Zhang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310000, China
| | - Meiqi Zhang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310000, China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Xin Liu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310000, China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yashu Wu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310000, China
| | - Qin Wu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310000, China
- Zhejiang Cancer Hospital, The University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - John C Chaput
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697-3958, United States
- Department of Chemistry, University of California, Irvine, CA 92697-3958, United States
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697-3958, United States
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697-3958, United States
| | - Yajun Wang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310000, China
- Zhejiang Cancer Hospital, The University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
32
|
Zhang W, Zhu Z, Liu Y. The impact of the ATP-binding cassette (ABC) transporter family on multidrug resistance in head and neck tumors. Mol Biol Rep 2025; 52:256. [PMID: 39982595 DOI: 10.1007/s11033-025-10321-9] [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: 11/14/2024] [Accepted: 01/30/2025] [Indexed: 02/22/2025]
Abstract
The ATP-binding cassette (ABC) transporter family is among the largest protein superfamilies, consisting of seven subfamilies, and plays an important role in various physiological processes and in the clinical manifestations of many diseases. The early clinical signs of head and neck cancer (HNC) are often subtle, resulting in most patients being diagnosed at more advanced stages. This late diagnosis adversely affects tumor treatment, and the resistance of certain tumors to chemotherapy further poses significant challenges for clinical management. Several previous studies have indicated a correlation between the ABC protein family and multidrug resistance (MDR) in tumors. This article offers a thorough review of the subfamilies, structures, functions, and roles of ABC transporters in MDR related to head and neck tumors, with the aim of providing insights and recommendations for overcoming MDR in this context.
Collapse
Affiliation(s)
- Wanqing Zhang
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhengxin Zhu
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuehui Liu
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
33
|
Xu J, Chen G, Mo C, Sha Y, Luo S, Ou M. Development and evaluation of siRNA-mediated gene silencing strategies for ADO2 therapy utilizing iPSCs model and DMPC-SPIONs delivery system. Stem Cell Res Ther 2025; 16:66. [PMID: 39934917 PMCID: PMC11816505 DOI: 10.1186/s13287-025-04151-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 01/15/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Autosomal dominant osteodystrophy type II (ADO2) is an inherited disease characterized by an abnormal increase in bone mineral density, and CLCN7 (R286W) is its most common causative mutation. The aim of this study was to explore the new idea of siRNA technology applied to the in vitro treatment of ADO2. METHODS Urinary-derived cells from ADO2 patients were collected to establish induced pluripotent stem cells (iPSCs) model. The siRNA targeting CLCN7 (R286W) mutant mRNA was designed. the cytotoxicity of the delivery vector DMPC-SPIONs was comprehensively evaluated by CCK-8 assay, flow cytometry and scratch assay. Finally, qPCR was utilized to verify the post-transcriptional silencing effect of siRNAs. RESULTS We found that DMPC-SPIONs had low cytotoxicity and were able to effectively deliver siRNAs into ADO2-iPSCs. qPCR confirmed that siRNA-DMPC-SPIONs were able to significantly reduce the expression level of mutant CLCN7 (66%), while there was no significant effect on the expression of wild-type CLCN7. CONCLUSIONS This study developed a gene silencing strategy based on siRNAs and DMPC-SPIONs, which provides a potential new approach for the treatment of ADO2 and demonstrates the potential application of siRNA technology in the treatment of autosomal dominant genetic diseases. INNOVATIVE STATEMENTS In this study, we used the established ADO2-iPSCs using patient's urine-derived cells to explore the safety and efficacy of siRNA technology based on the principle of RNA interference for ADO2 treatment for the first time. In addition, we chose DMPC-SPIONs as the delivery vehicle for siRNA, which cleverly exploits the advantages of nanoparticles such as superparamagnetism, low cytotoxicity, and good bio-histocompatibility.
Collapse
Affiliation(s)
- Jiajun Xu
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
| | - Gengshuo Chen
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
| | - Chune Mo
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
| | - Yu Sha
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
| | - Sha Luo
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China
| | - Minglin Ou
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China.
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, China.
| |
Collapse
|
34
|
Choudry MW, Riaz R, Raza MH, Nawaz P, Ahmad B, Jahan N, Rafique S, Afzal S, Amin I, Shahid M. Development of non-viral targeted RNA delivery vehicles - a key factor in success of therapeutic RNA. J Drug Target 2025; 33:171-184. [PMID: 39392510 DOI: 10.1080/1061186x.2024.2416241] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/23/2024] [Accepted: 10/08/2024] [Indexed: 10/12/2024]
Abstract
Decade-long efforts in medicinal biotechnology have enabled large-scale in-vitro production of optimised therapeutic RNA constructs for stable in-vivo delivery and modify the expression of disease-related genes. The success of lipid nanoparticle-formulated mRNA vaccines against Severe acute respiratory syndrome Coronavirus-2 (SARS-Cov2) has opened a new era of RNA therapeutics and non-viral drug delivery systems. The major limiting factor in the clinical translation of RNA-based drugs is the availability of suitable delivery vehicles that can protect RNA payloads from degradation, offer controlled release, and pose minimal inherent toxicity. Unwanted immune response, payload size constraints, genome integration, and non-specific tissue targeting limit the application of conventional viral drug-delivery vehicles. This review summarises current research on nano-sized drug carriers, including lipid nanoparticles, polymer-based formulations, cationic nanoemulsion, and cell-penetrating peptides, for targeted therapeutic RNA delivery. Further, this paper highlights the biomimetic approaches (i.e. mimicking naturally occurring bio-compositions, molecular designs, and systems), including virus-like particles (VLPs), exosomes, and selective endogenous eNcapsidation (SEND) technology being explored as safer and more efficient alternatives.
Collapse
Affiliation(s)
- Muhammad Waqas Choudry
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Rabia Riaz
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Muhammad Hassan Raza
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Pashma Nawaz
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Bilal Ahmad
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Neelam Jahan
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Shazia Rafique
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Samia Afzal
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Iram Amin
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Muhammad Shahid
- Division of Molecular Virology, National Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| |
Collapse
|
35
|
Nestor MS, Hetzel J, Awad N, Bhupalam V, Lu P, Molyneaux M. A Novel Injectable Polypeptide Nanoparticle Encapsulated siRNA Targeting TGF-β1 and COX-2 for Localized Fat Reduction II: Phase I Clinical Trial. J Cosmet Dermatol 2025; 24:e16722. [PMID: 39692702 PMCID: PMC11845920 DOI: 10.1111/jocd.16722] [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: 08/27/2024] [Revised: 11/18/2024] [Accepted: 11/29/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Rising demand for non-invasive body contouring is driven by aesthetics and the obesity epidemic. Deoxycholic acid (DCA) is the only FDA-approved injectable for fat reduction but can cause side effects and significant local skin reactions (LSR). RNA interference, using small interfering RNA (siRNA) molecules, offers targeted fat reduction by silencing genes involved in fat maintenance. STP705, a siRNA injectable targeting TGF-β1 and COX-2, has shown promising preclinical results both in vitro and in animal models. AIMS To evaluate the safety and tolerability of STP705 for localized fat reduction in subjects undergoing abdominoplasty. METHODS This phase I dose-ranging, randomized, vehicle-controlled trial involved eight females undergoing abdominoplasty who received subcutaneous STP705 injections at varying concentrations and volumes in designated abdominal zones. Safety assessments, including physical exams, lab tests, ECGs, and local skin reactions (LSRs), were conducted at baseline and follow-ups. Histopathologic evaluations of biopsies collected during abdominoplasty assessed adipocyte apoptosis and tissue remodeling. RESULTS STP705 demonstrated a favorable safety profile with no clinically significant changes in lab values, vital signs, or ECGs. Adverse events (AEs) were rare and transient. The incidence, intensity, and duration of LSRs were low throughout the study. Histological analysis revealed adipocyte destruction, fat remodeling, and necrosis. CONCLUSION STP705 was safe and very well-tolerated and showed preliminary efficacy in inducing adipocyte apoptosis and tissue remodeling, suggesting a safer alternative or adjunct to existing fat reduction therapies. These findings support further trials to establish the safety and efficacy of STP705 for targeted fat reduction and body contouring. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT05422378.
Collapse
Affiliation(s)
- Mark S. Nestor
- Center for Clinical and Cosmetic ResearchAventuraFloridaUSA
- Department of Dermatology and Cutaneous Surgery, University of MiamiMiller School of MedicineMiamiFloridaUSA
| | - John Hetzel
- Center for Clinical and Cosmetic ResearchAventuraFloridaUSA
| | - Nardin Awad
- Center for Clinical and Cosmetic ResearchAventuraFloridaUSA
| | | | | | | |
Collapse
|
36
|
Linthorst NA, van Vlijmen BJ, Eikenboom JC. The future of siRNA-mediated approaches to treat von Willebrand disease. Expert Rev Hematol 2025; 18:109-122. [PMID: 39865861 PMCID: PMC11854048 DOI: 10.1080/17474086.2025.2459259] [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: 11/27/2024] [Revised: 12/19/2024] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
INTRODUCTION The clinical management of the inherited bleeding disorder von Willebrand disease (VWD) focuses on normalizing circulating levels of von Willebrand factor (VWF) and factor VIII (FVIII) to prevent or control bleeding events. The heterogeneous nature of VWD, however, complicates effective disease management and development of universal treatment guidelines. AREAS COVERED The current treatment modalities of VWD and their limitations are described and why this prompts the development of new treatment approaches. In particular, RNA-based therapeutics have gained significant interest because of their ability to reversibly alter gene expression with long-term efficacy. In the field of VWD, small-interfering RNAs (siRNAs) have been explored through various strategies to improve disease phenotypes. These different approaches are discussed as well as their potential impact on reshaping the future therapeutic landscape. EXPERT OPINION Current treatments for VWD often require frequent intravenous administration of VWF concentrates or desmopressin, with only short-term benefits. Moreover, remaining circulating mutant VWF can cause detrimental effects. Allele-selective siRNA-based therapies could provide more reliable and long-term disease correction by specifically targeting mutant VWF. This approach could be applied to a large part of the population aligning with the growing emphasis on personalized treatment and patient-centered care in VWD management.
Collapse
Affiliation(s)
- Noa A. Linthorst
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Bart J.M van Vlijmen
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen C.J Eikenboom
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
37
|
Yan H, Jiang N, Li X, Lin C, Wang F, Zhang J, Chen L, Li D. Exosomal lncRNAs as diagnostic and therapeutic targets in multiple myeloma. Front Oncol 2025; 14:1522491. [PMID: 39886670 PMCID: PMC11779718 DOI: 10.3389/fonc.2024.1522491] [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: 11/04/2024] [Accepted: 12/16/2024] [Indexed: 02/01/2025] Open
Abstract
Multiple Myeloma (MM) is the second most common malignancy of the hematopoietic system, accounting for approximately 10% of all hematological malignancies, and currently, there is no complete cure. Existing research indicates that exosomal long non-coding RNAs (lncRNAs) play a crucial regulatory role in the initiation and progression of tumors, involving various interactions such as lncRNA-miRNA, lncRNA-mRNA, and lncRNA-RNA binding proteins (RBP). Despite the significant clinical application potential of exosomal lncRNAs, research in this area still faces challenges due to their low abundance and technical limitations. To our knowledge, this review is the first to comprehensively integrate and elucidate the three mechanisms of action of exosomal lncRNAs in MM, and to propose potential therapeutic targets and clinical cases based on these mechanisms. We highlight the latest advancements in the potential of exosomal lncRNAs as biomarkers and therapeutic targets, offering not only a comprehensive analysis of the role of exosomal lncRNAs in MM but also new perspectives and methods for future clinical diagnosis and treatment of multiple myeloma.
Collapse
Affiliation(s)
- Hong Yan
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Nan Jiang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiaoying Li
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Chenyang Lin
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Fang Wang
- School of Dental Medicine, Dalian University, Dalian, Liaoning, China
| | - Juan Zhang
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lijuan Chen
- Department of Hematopathology, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - Dan Li
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
38
|
Zhang J, Wang Z, Liao Y, Tong J, Gao R, Zeng Z, Bai Y, Wei Y, Guo X. Black phosphorus nanoplatform coated with platelet membrane improves inhibition of atherosclerosis progression through macrophage targeting and efferocytosis. Acta Biomater 2025; 192:377-393. [PMID: 39608658 DOI: 10.1016/j.actbio.2024.11.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/13/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
Plaque rupture in atherosclerosis (AS) is a major cause of acute cardiovascular events. Macrophage-induced inflammatory responses and accumulation of excess reactive oxygen species (ROS) primarily induce unstable plaques. Therefore, targeting ROS clearance and functional modulation of macrophages are clinically crucial for improving plaque stability and inhibiting AS progression. Here, we constructed a bionic nano-delivery platform, PBP@siR@PM, using platelet membranes (PM) coated with black phosphorus nanosheets (BPNSs) to target macrophages in atherosclerotic plaques. Meanwhile, PM-coated BPNSs (PBP@siR@PM) were used to deliver small interfering RNA silencing Ca2+/calmodulin-dependent protein kinase γ (CaMKIIγ) into macrophages. Furthermore, macrophage efferocytosis was restored by inhibiting CaMKIIγ and increasing the expression of MerTK, a cytosolic receptor, thus promoting the clearance of apoptotic cells from plaques. This study demonstrated that intraplaque macrophage-targeted therapy using the bionic nano-delivery platform PBP@siR@PM effectively removed excess ROS from macrophages, promoted efferocytosis, cleared apoptotic cells in plaques, improved plaque stability, and largely inhibited AS progression in ApoE-/- mice after high fat diet. In summary, this study proposes a therapeutic strategy for AS and highlights the outstanding therapeutic potential of biomimetic nanomaterials in this type of chronic inflammatory disease. STATEMENT OF SIGNIFICANCE: Rupture of atherosclerotic unstable plaques is a major cause of acute cardiovascular events. Macrophage-induced chronic inflammation and oxidative stress due to overloaded ROS are major contributors to plaque rupture. In this study, we focused on the improvement of macrophage efferocytosis within the plaque for the effective treatment of atherosclerosis. A bionic nano-delivery platform was constructed using platelet membranes (PM) coated black phosphorus nanosheets (BPNSs) to target macrophages in atherosclerotic plaques. In conclusion, intraplaque macrophage-targeted therapy based on the bionic nano-delivery platform PBP@siR@PM effectively scavenges overloaded ROS in macrophages, promotes efferocytosis, removes apoptotic cells from plaques, and improves plaque stability, which significantly inhibits the progression of atherosclerosis in ApoE-/- mice after a high-fat diet.
Collapse
Affiliation(s)
- Jiahui Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhiwen Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuhan Liao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Junran Tong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ran Gao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhuanglin Zeng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yu Bai
- Department of Pulmonary and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, 100029 Beijing, China.
| | - Yumiao Wei
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Xiaopeng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
39
|
Ren L, Danser AHJ. Small Interfering RNA Therapy for the Management and Prevention of Hypertension. Curr Hypertens Rep 2025; 27:5. [PMID: 39808369 PMCID: PMC11732957 DOI: 10.1007/s11906-025-01325-8] [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] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
PURPOSE OF REVIEW To review currently existing knowledge on a new type of antihypertensive treatment, small interfering RNA (siRNA) targeting hepatic angiotensinogen. RECENT FINDINGS Targeting angiotensinogen synthesis in the liver with siRNA allows reaching a suppression of renin-angiotensin system (RAS) activity for up to 6 months after 1 injection. This might revolutionize antihypertensive treatment, as it could overcome non-adherence, the major reason for inadequate blood pressure control. Animal data support that its effects on blood pressure and end-organ damage are fully comparable to those of classical RAS blockers, and phase I and II clinical trials confirm its antihypertensive effectiveness and long-term action. Although its side effect profile is placebo-like, its long-term effects also pose a threat in patients who require immediate restoration of RAS activity, like in shock. Here tools are being developed, called REVERSIR, that allow immediate annihilation of the siRNA effect in the liver. One subcutaneous injection of angiotensinogen siRNA lowers blood pressure for 6 months without severe side effects. The decrease in angiotensinogen and blood pressure can be reversed with a drug called REVERSIR if needed.
Collapse
Affiliation(s)
- Liwei Ren
- Department of Pharmacy, The Second Clinical Medical College, The First Affiliated Hospital, Shenzhen People's Hospital, Jinan University, Southern University of Science and Technology), Shenzhen, China
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, room EE1418b, Erasmus MC, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands
| | - A H Jan Danser
- Department of Pharmacy, The Second Clinical Medical College, The First Affiliated Hospital, Shenzhen People's Hospital, Jinan University, Southern University of Science and Technology), Shenzhen, China.
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, room EE1418b, Erasmus MC, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands.
| |
Collapse
|
40
|
Roussis SG, Nguyen K, Rentel C. SEC x IP two-dimensional LCMS for the analysis of non-denatured and denatured cyclic-peptide siRNAs in a single step. J Chromatogr A 2025; 1740:465552. [PMID: 39631127 DOI: 10.1016/j.chroma.2024.465552] [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: 10/29/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/07/2024]
Abstract
The increased effectiveness of small interfering RNAs (siRNAs) to induce gene silencing has brought a great therapeutic promise to many diseases. siRNAs are under highly active current research and development. Ligand conjugation and chemical modifications of the sense (SS) and antisense (AS) strands of the siRNA duplex improve stability and facilitate delivery, but significantly increase the complexity of the analytical requirements. Two chromatographic methods are needed to guide synthesis and formulation: (1) a non-denaturing method to analyze the duplex, residual sense and antisense strands, their impurities, and those of the duplex, and (2) a denaturing method for each strand and its impurities. In this work, ion-pair reversed phase (IP-RP) and strong anion exchange (SAX) methods were not successful in the analysis of a cyclic-peptide (CP) siRNA, in the non-denaturing mode. Selection of the most appropriate chromatographic method is greatly challenged by the chemical properties of the conjugated ligands. However, separation was possible by size exclusion chromatography (SEC). The non-denaturing SEC method was implemented, using a 2D-LC system, in the 1D dimension of the analysis, coupled with a denaturing IP-RP method in the 2D dimension. The 2D-LC system greatly simplified the siRNA analysis by combining, for the first time, the non-denaturing and denaturing methods into a single-instrument, one sample injection method. An additional benefit of the 2D-LC system is the interfacing of MS-incompatible methods (e.g., SAX, SEC) to a mass spectrometer, broadening thus the analytical options, by coupling with MS-compatible methods (IP-RP, HILIC) in the 2D dimension. Application of the approach was exemplified in a CP-siRNA duplex formulation study to determine the optimal mixing ratio of the individual strands. A duplex maximum was reached at a sample solution AS:SS ratio of 0.9. The method was found to be independent of the amount and concentration of sample injected. A duplex annealing study found no significant temperature or salt effects in the formulation of the CP-siRNA duplex.
Collapse
|
41
|
Salim EL, Kristensen K, Sjögren E. Whole-Body Physiologically Based Pharmacokinetic Modeling of GalNAc-Conjugated siRNAs. Pharmaceutics 2025; 17:69. [PMID: 39861717 PMCID: PMC11769416 DOI: 10.3390/pharmaceutics17010069] [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: 12/05/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: N-acetyl-galactosamine small interfering RNAs (GalNAc-siRNA) are an emerging class of drugs due to their durable knockdown of disease-related proteins. Direct conjugation of GalNAc onto the siRNA enables targeted uptake into hepatocytes via GalNAc recognition of the Asialoglycoprotein Receptor (ASGPR). With a transient plasma exposure combined with a prolonged liver half-life, GalNAc-siRNA exhibits distinct disposition characteristics. We aimed to develop a generic GalNAc-siRNAs whole-body physiologically based pharmacokinetic-pharmacodynamic (WB-PBPK-PD) model for describing the pharmacokinetic-pharmacodynamic (PK-PD) relationship and overall tissue distribution in the open-source platform Open Systems Pharmacology Suite. Methods: Model development was performed using published studies in mice leveraging the PK-Sim® standard implementation for large molecules with added implementations of ASGPR-mediated liver disposition and downstream target effects. Adequate model performance was achieved across study measurements and included studies adopting a combination of global and compound-specific parameters. Results: The analysis identified significant compound dependencies, e.g., endosomal stability, with direct consequences for the pharmacological effect. Additionally, knowledge gaps in mechanistic understanding related to extravasation and overall tissue distribution were identified during model development. The presented study provides a generic WB-PBPK-PD model for the investigation of GalNAc-siRNAs implemented in a standardized open-source platform.
Collapse
Affiliation(s)
- Emilie Langeskov Salim
- Department of Pharmaceutical Bioscience, Translational Drug Discovery and Development, Uppsala University, SE-75124 Uppsala, Sweden;
- Department of Discovery PKPD & QSP Modelling, Novo Nordisk A/S, DK-2760 Måløv, Denmark;
| | - Kim Kristensen
- Department of Discovery PKPD & QSP Modelling, Novo Nordisk A/S, DK-2760 Måløv, Denmark;
| | - Erik Sjögren
- Department of Pharmaceutical Bioscience, Translational Drug Discovery and Development, Uppsala University, SE-75124 Uppsala, Sweden;
| |
Collapse
|
42
|
Helal MM, Ibrahim AA, Beddor A, Kashbour M. Breaking Barriers in Huntington's Disease Therapy: Focused Ultrasound for Targeted Drug Delivery. Neurochem Res 2025; 50:68. [PMID: 39751928 PMCID: PMC11698766 DOI: 10.1007/s11064-024-04302-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/14/2024] [Accepted: 11/26/2024] [Indexed: 01/04/2025]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disease resulting from a mutation in the huntingtin (HTT) gene and characterized by progressive motor dysfunction, cognitive decline, and psychiatric disturbances. Currently, no disease-modifying treatments are available. Recent research has developed therapeutic agents that may have the potential to directly target the disease pathology, such as gene silencing or clearing the mutant protein. However, these agents are limited by their inability to cross the blood-brain barrier (BBB), preventing optimal therapeutic effects. Although various techniques have been explored to overcome the BBB, focused ultrasound (FUS) has emerged as a promising non-invasive therapeutic modality offering the potential for targeted intervention in neurodegenerative diseases, including HD. Preclinical studies demonstrated the safety and efficacy of FUS in delivering therapeutic agents, such as siRNAs and AAV vector-based gene therapy, resulting in significant reductions in mutant HTT expression and improvements in motor function in HD mouse models. Furthermore, the safety profile of FUS-induced BBB opening has been established in clinical trials on human patients of neurodegenerative diseases other than HD, showing no adverse effects on brain structure or function. This review provides a comprehensive overview of the current state of FUS research in HD and connects existing evidence from neurodegenerative disease studies with its promise in establishing disease-modifying therapies for HD.
Collapse
Affiliation(s)
| | - Arwa Amer Ibrahim
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Ahmad Beddor
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
- Faculty of Medicine, Yarmouk University, Irbid, Jordan
| | - Muataz Kashbour
- Diagnostic Radiology Department, National Cancer Institute, Misrata, Libya
| |
Collapse
|
43
|
Amaeze O, Isoherranen N, Shum S. The absorption, distribution, metabolism and elimination characteristics of small interfering RNA therapeutics and the opportunity to predict disposition in pregnant women. Drug Metab Dispos 2025; 53:100018. [PMID: 39884813 DOI: 10.1124/dmd.123.001383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 04/19/2024] [Accepted: 05/15/2024] [Indexed: 05/31/2024] Open
Abstract
Small interfering RNA (siRNA) therapeutics represent an emerging class of pharmacotherapy with the potential to address previously hard-to-treat diseases. Currently approved siRNA therapeutics include lipid nanoparticle-encapsulated siRNA and tri-N-acetylated galactosamine-conjugated siRNA. These siRNA therapeutics exhibit distinct pharmacokinetic characteristics and unique absorption, distribution, metabolism, and elimination (ADME) properties. As a new drug modality, limited clinical data are available for siRNA therapeutics in specific populations, including pediatrics, geriatrics, individuals with renal or hepatic impairment, and pregnant women, making dosing challenging. In this Minireview, a mechanistic overview of the ADME properties of the 5 currently approved siRNA therapeutics is presented. A concise overview of the clinical data available for therapeutic siRNAs in special populations, focusing on the potential impact of physiologic changes during pregnancy on siRNA disposition, is provided. The utility of physiologically based pharmacokinetic (PBPK) modeling as a tool to elucidate the characteristics and disposition of siRNA therapeutics in pregnant women is explored. Additionally, opportunities to integrate known physiologic alterations induced by pregnancy into PBPK models that incorporate siRNA ADME mechanisms to predict the effects of pregnancy on siRNA disposition are discussed. Clinical data regarding the use of therapeutic siRNA in special populations remain limited. Data for precise parameterization of maternal-fetal siRNA PBPK models are lacking presently and underscore the need for further research in this area. Addressing this gap in knowledge will not only enhance our understanding of siRNA pharmacokinetics during pregnancy but also advance the possible development of siRNA therapeutics to treat pregnancy-related conditions. SIGNIFICANCE STATEMENT: This Minireview proposes a framework on how small interfering RNA (siRNA) disposition can be predicted in pregnancy based on mechanistic absorption, distribution, metabolism, and elimination (ADME) information using physiologically-based pharmacokinetic (PBPK) modeling. The mechanistic ADME information and available clinical data in special populations of currently Food and Drug Administration-approved siRNA therapeutics are summarized. Additionally, how physiological changes during pregnancy may affect siRNA disposition is reviewed, and the opportunities to use PBPK modeling to predict siRNA disposition in pregnant women is explored.
Collapse
Affiliation(s)
- Ogochukwu Amaeze
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Sara Shum
- ReNAgade Therapeutics Management Inc, Cambridge, Massachusetts.
| |
Collapse
|
44
|
Hassan M, Malik A, Yaseen Z, Shahzadi S, Yasir M, Kloczkowski A. A Glimpse of Noncoding RNAs: Secondary Structure, Emerging Trends, and Potential Applications in Human Diseases. Methods Mol Biol 2025; 2867:331-344. [PMID: 39576590 DOI: 10.1007/978-1-0716-4196-5_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2024]
Abstract
An appealing strategy for the treatment of several diseases is the therapeutic targeting of noncoding RNAs (ncRNAs), such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs). Many antisense oligonucleotides and small interfering RNAs have been tested in clinical studies over the past 10 years, and several of these have received FDA approval. However, trial results have thus far been mixed, with some studies reporting strong effects and others showing low effectiveness or side effects, including toxicity. Clinical trials for alternative entities like antimiRNAs are underway, and interest in lncRNA-based therapies is constantly growing. From this perspective, we discuss the basic overview of ncRNAs, their significant role as therapeutic biomarkers against different diseases, and the role of secondary structure in noncoding RNAs.
Collapse
Affiliation(s)
- Mubashir Hassan
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Amal Malik
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Zainab Yaseen
- Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Saba Shahzadi
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Muhammad Yasir
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Andrzej Kloczkowski
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA.
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA.
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
45
|
Daga P, Singh G, Menon T, Sztukowska M, Kalra DK. Emerging RNAi Therapies to Treat Hypertension. Mol Diagn Ther 2025; 29:25-41. [PMID: 39400663 DOI: 10.1007/s40291-024-00747-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 10/15/2024]
Abstract
Hypertension (HTN), often dubbed the "silent killer," poses a significant global health challenge, affecting over 1.3 billion individuals. Despite advances in treatment, effective long-term blood pressure (BP) control remains elusive, necessitating novel therapeutic approaches. Poor control of BP remains a leading cause of cardiovascular morbidity and mortality worldwide and is becoming an even larger global health problem due to the aging population, rising rates of obesity, poorer dietary patterns and overall cardiometabolic health, and suboptimal rates of patient adherence and optimal BP control. Ribonucleic acid interference (RNAi) technology, which leverages the body's natural gene-silencing mechanism, has emerged as a promising strategy for several diseases and has recently been tested for its antihypertensive effects. We systematically reviewed peer-reviewed articles from databases including PubMed, EMBASE, and Scopus for studies examining RNAi's role in managing HTN, focusing on mechanisms, clinical utility, and safety profile. Key early-phase trials of some RNAi-leading candidate drugs are detailed. Also highlighted are challenges such as target specificity, delivery mechanisms, durability of effect, and immunogenicity. We conclude by summarizing how RNAi has a significant potential role in HTN therapy due to their unique benefits, such as long-term duration of action, infrequent dosing, and lack of major side effects.
Collapse
Affiliation(s)
- Pawan Daga
- Department of Internal Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - Gurnoor Singh
- Division of Cardiology, Department of Medicine, Rudd Heart and Lung Center, University of Louisville School of Medicine, 201 Abraham Flexner Way, Suite 600, Louisville, KY, 40202, USA
| | - Tushar Menon
- Division of Cardiology, Department of Medicine, Rudd Heart and Lung Center, University of Louisville School of Medicine, 201 Abraham Flexner Way, Suite 600, Louisville, KY, 40202, USA
| | - Maryta Sztukowska
- Clinical Trials Unit, University of Louisville School of Medicine, Louisville, KY, USA
- University of Information Technology and Management, Rzeszow, Poland
| | - Dinesh K Kalra
- Division of Cardiology, Department of Medicine, Rudd Heart and Lung Center, University of Louisville School of Medicine, 201 Abraham Flexner Way, Suite 600, Louisville, KY, 40202, USA.
| |
Collapse
|
46
|
Sopić M, Vladimirov S, Munjas J, Mitić T, Hall IF, Jusic A, Ruzic D, Devaux Y. Targeting noncoding RNAs to treat atherosclerosis. Br J Pharmacol 2025; 182:220-245. [PMID: 38720437 DOI: 10.1111/bph.16412] [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: 10/26/2023] [Revised: 03/06/2024] [Accepted: 04/05/2024] [Indexed: 12/13/2024] Open
Abstract
Noncoding RNAs (ncRNAs) are pivotal for various pathological processes, impacting disease progression. The potential for leveraging ncRNAs to prevent or treat atherosclerosis and associated cardiovascular diseases is of great significance, especially given the increasing prevalence of atherosclerosis in an ageing and sedentary population. Together, these diseases impose a substantial socio-economic burden, demanding innovative therapeutic solutions. This review explores the potential of ncRNAs in atherosclerosis treatment. We commence by examining approaches for identifying and characterizing atherosclerosis-associated ncRNAs. We then delve into the functional aspects of ncRNAs in atherosclerosis development and progression. Additionally, we review current RNA and RNA-targeting molecules in development or under approval for clinical use, offering insights into their pharmacological potential. The importance of improved ncRNA delivery strategies is highlighted. Finally, we suggest avenues for advanced research to accelerate the use of ncRNAs in treating atherosclerosis and mitigating its societal impact. 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)
- Miron Sopić
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Sandra Vladimirov
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Jelena Munjas
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Tijana Mitić
- BHF/University Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ignacio Fernando Hall
- BHF/University Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Amela Jusic
- HAYA Therapeutics SA, SuperLab Suisse - Bâtiment Serine, Lausanne, Vaud, Switzerland
| | - Dusan Ruzic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Luxembourg, Luxembourg
| |
Collapse
|
47
|
Askarizadeh A, Vahdat-Lasemi F, Karav S, Kesharwani P, Sahebkar A. Lipid nanoparticle-based delivery of small interfering RNAs: New possibilities in the treatment of diverse diseases. Eur Polym J 2025; 223:113624. [DOI: 10.1016/j.eurpolymj.2024.113624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
48
|
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
|
49
|
Anvari S, Nikbakht M, Vaezi M, Amini-Kafiabad S, Ahmadvand M. Immune checkpoints and ncRNAs: pioneering immunotherapy approaches for hematological malignancies. Cancer Cell Int 2024; 24:410. [PMID: 39702293 DOI: 10.1186/s12935-024-03596-8] [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: 03/19/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024] Open
Abstract
Hematological malignancies are typically treated with chemotherapy and radiotherapy as the first-line conventional therapies. However, non-coding RNAs (ncRNAs) are a rapidly expanding field of study in cancer biology that influences the growth, differentiation, and proliferation of tumors by targeting immunological checkpoints. This study reviews the results of studies (from 2012 to 2024) that consider the immune checkpoints and ncRNAs in relation to hematological malignancies receiving immunotherapy. This article provides a summary of the latest advancements in immunotherapy for treating hematological malignancies, focusing on the role of immune checkpoints and ncRNAs in the immune response and their capacity for innovative strategies. The paper also discusses the function of immune checkpoints in maintaining immune homeostasis and how their dysregulation can contribute to developing leukemia and lymphoma. Finally, this research concludes with a discussion on the obstacles and future directions in this rapidly evolving field, emphasizing the need for continued research to fully harness the capacity of immune checkpoints and ncRNAs in immunotherapy for hematological malignancies.
Collapse
Affiliation(s)
- Samira Anvari
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mohsen Nikbakht
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Vaezi
- Hematology, Oncology, and Stem Cell Transplantation Research Center Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Sedigheh Amini-Kafiabad
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran.
| | - Mohammad Ahmadvand
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
50
|
D’Amato A, Prosperi S, Severino P, Myftari V, Correale M, Perrone Filardi P, Badagliacca R, Fedele F, Vizza CD, Palazzuoli A. MicroRNA and Heart Failure: A Novel Promising Diagnostic and Therapeutic Tool. J Clin Med 2024; 13:7560. [PMID: 39768484 PMCID: PMC11728316 DOI: 10.3390/jcm13247560] [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: 11/16/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 01/06/2025] Open
Abstract
Heart failure (HF) has a multifaceted and complex pathophysiology. Beyond neurohormonal, renin-angiotensin-aldosterone system, and adrenergic hyperactivation, a role for other pathophysiological determinants is emerging. Genetic and epigenetic factors are involved in this syndrome. In many maladaptive processes, the role of microRNAs (miRNAs) has been recently demonstrated. MiRNAs are small endogenous non-coding molecules of RNA involved in gene expression regulation, and they play a pivotal role in intercellular communication, being involved in different biological and pathophysiological processes. MiRNAs can modulate infarct area size, cardiomyocytes restoration, collagen deposition, and macrophage polarization. MiRNAs may be considered as specific biomarkers of hypertrophy and fibrosis. MiRNAs have been proposed as a therapeutical tool because their administration can contrast with myocardial pathophysiological remodeling leading to HF. Antimir and miRNA mimics are small oligonucleotides which may be administered in several manners and may be able to regulate the expression of specific and circulating miRNAs. Studies on animal models and on healthy humans demonstrate that these molecules are well tolerated and effective, opening the possibility of a therapeutic use of miRNAs in cases of HF. The application of miRNAs for diagnosis, prognostic stratification, and therapy fits in with the new concept of a personalized and tailored approach to HF.
Collapse
Affiliation(s)
- Andrea D’Amato
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, ‘Sapienza’ University of Rome, Policlinico ‘Umberto I’ of Rome, 00161 Rome, Italy; (A.D.); (S.P.); (P.S.); (V.M.); (R.B.); (C.D.V.)
| | - Silvia Prosperi
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, ‘Sapienza’ University of Rome, Policlinico ‘Umberto I’ of Rome, 00161 Rome, Italy; (A.D.); (S.P.); (P.S.); (V.M.); (R.B.); (C.D.V.)
| | - Paolo Severino
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, ‘Sapienza’ University of Rome, Policlinico ‘Umberto I’ of Rome, 00161 Rome, Italy; (A.D.); (S.P.); (P.S.); (V.M.); (R.B.); (C.D.V.)
| | - Vincenzo Myftari
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, ‘Sapienza’ University of Rome, Policlinico ‘Umberto I’ of Rome, 00161 Rome, Italy; (A.D.); (S.P.); (P.S.); (V.M.); (R.B.); (C.D.V.)
| | - Michele Correale
- Cardiothoracic Department, ‘Policlinico Riuniti’ University Hospital, 71100 Foggia, Italy
| | - Pasquale Perrone Filardi
- Department of Advanced Biomedical Sciences, Section of Cardiology, Federico II University, 80131 Naples, Italy;
| | - Roberto Badagliacca
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, ‘Sapienza’ University of Rome, Policlinico ‘Umberto I’ of Rome, 00161 Rome, Italy; (A.D.); (S.P.); (P.S.); (V.M.); (R.B.); (C.D.V.)
| | | | - Carmine Dario Vizza
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, ‘Sapienza’ University of Rome, Policlinico ‘Umberto I’ of Rome, 00161 Rome, Italy; (A.D.); (S.P.); (P.S.); (V.M.); (R.B.); (C.D.V.)
| | - Alberto Palazzuoli
- Cardio Thoracic and Vascular Department, ‘S. Maria alle Scotte Hospital’, University of Siena, 53100 Siena, Italy;
| |
Collapse
|