1
|
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
|
2
|
Zhang D, Pei S, Feng Z, Xia G. Functions and mechanisms of lncRNAs in immune escape and their application in immunotherapy for colorectal cancer. J Transl Med 2025; 23:689. [PMID: 40537762 DOI: 10.1186/s12967-025-06732-8] [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: 11/25/2024] [Accepted: 06/07/2025] [Indexed: 06/22/2025] Open
Abstract
Recent studies have progressively clarified the critical role of long non-coding RNA (lncRNA) in cancer, especially concerning its involvement in immune processes, which holds significant therapeutic promise. However, the incorporation of lncRNA into immunotherapy has yet to demonstrate adequate clinical efficacy. This shortcoming can be attributed to various challenges, including unclear mechanisms of action and limited prognostic capacity. In colorectal cancer (CRC), this ineffectiveness may predominantly stem from immune escape mechanisms mediated by lncRNAs. Despite some exploration into lncRNA-driven immune escape, a comprehensive overview of its implications for immunotherapy in CRC remains inadequately addressed. In this review, we thoroughly assess the role of lncRNA in immune escape mechanisms associated with CRC, focusing on immune checkpoints, immune cells, and cytokines. We elucidate the relationship between lncRNA-mediated immune escape and CRC progression and discuss strategies for lncRNA-mediated immunotherapy for CRC. Our aim is to provide fresh insights into the potential of targeting lncRNAs to enhance the effectiveness of immunotherapeutic strategies in CRC.
Collapse
Affiliation(s)
- Dechun Zhang
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Shaoxuan Pei
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Zixuan Feng
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Guangfeng Xia
- Department of Hepatobiliary Surgery, Jiujiang University Affiliated Hospital, Jiujiang, 332000, China.
| |
Collapse
|
3
|
Xiong L, Chen S, Li S, He D, Wang Y, Zhang Q, He Z, Li M, He Q. ATP-responsive tumor targeted lipid nanoparticle for enhanced siRNA delivery and improved treatment efficacy in melanoma. J Control Release 2025; 382:113622. [PMID: 40120691 DOI: 10.1016/j.jconrel.2025.113622] [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: 12/02/2024] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/25/2025]
Abstract
Small interfering RNA (siRNA) plays a crucial role in tumor therapy, especially for non-druggable targets with obvious advantages. Nevertheless, its molecular weight, negative charge, and susceptibility to degradation hinder effective delivery to tumor cells for therapeutic action. Lipid nanoparticles (LNPs) serve as an excellent delivery mechanism for siRNA but still face problems such as suboptimal tumor targeting and inefficient intracellular release. To enhance melanoma treatment, we designed lipid nanoparticles modified with phenylboronic acid (PBA) for efficient delivery of siRNA targeting "undruggable" microphthalmia-associated transcription factor (MITF). This nanocarrier successfully encapsulated siRNA and improved tumor targeting by allowing phenylboronic acid to interact with sialic acid residues overexpressed in tumor cells. Furthermore, PBA-modified lipid nanoparticles facilitated the ATP-responsive release of siRNA intracellular. These two aspects enhance gene silencing efficiency. The in vivo targeting and gene silencing capabilities of PBA-modified lipid nanoparticles significantly surpassed those of unmodified LNP. Additionally, PBA-modified nanoparticles exhibited considerable anti-tumor and anti-metastatic effects in animal models, offering an alternative approach for siRNA therapy.
Collapse
Affiliation(s)
- Lin Xiong
- 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, PR China
| | - Shuang Chen
- 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, PR China
| | - Sihui Li
- 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, PR China
| | - Dan He
- 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, PR China
| | - Yashi Wang
- 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, PR China
| | - Qiang Zhang
- 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, PR China
| | - Zhidi He
- 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, PR China
| | - Man Li
- 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, PR China.
| | - Qin He
- 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, PR China.
| |
Collapse
|
4
|
Chen L, Bosmajian C, Woo S. Mechanistic intracellular PK/PD modeling to inform development strategies for small interfering RNA therapeutics. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102516. [PMID: 40242045 PMCID: PMC12002994 DOI: 10.1016/j.omtn.2025.102516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 03/12/2025] [Indexed: 04/18/2025]
Abstract
Small interfering RNA (siRNA) therapeutics provide a targeted approach to silence disease-related genes, with notable success in liver-targeting applications. However, the quantitative effects of siRNA properties, such as stability and affinity, as well as biological factors like cell proliferation, mRNA turnover, and abundance, on gene silencing, particularly for extrahepatic targets, remain poorly understood. To identify determinants influencing gene knockdown extent and duration, we developed a mechanistic intracellular pharmacokinetic/pharmacodynamic (PK/PD) model for RNAiMAX-delivered siRNA, based on cytoplasmic siRNA disposition, RISC-loaded siRNA exposure, and mRNA knockdown across different targets in MCF7 and BT474 cells. The model highlighted the critical roles of cell proliferation in silencing duration and mRNA turnover rates on knockdown extent. In rapid-dividing cells, mRNA half-life drives knockdown profiles, whereas chemical siRNA stabilization extends silencing in slow-dividing cells. Targets with extremely low or high mRNA abundance pose silencing challenges. While sufficient RISC occupancy is essential, increasing RISC exposure has minimal impact on silencing extent; enhancing siRNA-mRNA target engagement is more effective. The model also defined a quantitative relationship for maximal mRNA knockdown, governed by cell proliferation, mRNA half-life, and RISC-mediated cleavage rates. This mechanistic PK/PD modeling provides insights into optimizing siRNA design and target selection in therapeutic development.
Collapse
Affiliation(s)
- Lin Chen
- Division of Pharmacokinetics-Pharmacodynamics and Systems Pharmacology, Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA
| | - Caroline Bosmajian
- Division of Pharmacokinetics-Pharmacodynamics and Systems Pharmacology, Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA
| | - Sukyung Woo
- Division of Pharmacokinetics-Pharmacodynamics and Systems Pharmacology, Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14214, USA
| |
Collapse
|
5
|
Shi Q, Geng J, Han L, Ji X, Li J, Mu Y, Zhao T, Wang L, Jia H. Engineered Salmonella carrying siRNA-PD-1 shrinks orthotopically implanted bladder cancer in rats. J Drug Target 2025:1-9. [PMID: 40418584 DOI: 10.1080/1061186x.2025.2512619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/29/2025] [Accepted: 05/20/2025] [Indexed: 05/28/2025]
Abstract
BACKGROUND Currently, the application of engineered bacteria in tumour treatment has received increasing attention. It has been proved that Bacillus Calmette-Guerin (BCG) can effectively treat bladder cancer (BC). In addition, immune checkpoint blockade is an effective method for tumour treatment. Programmed cell death protein 1 (PD-1), an important immunosuppressive molecule, binds to programmed death ligand receptor 1 (PD-L1) and inhibits the anti-tumour effects of T cells. METHODS The plasmid encoding siRNA-PD-1 was constructed, and the rat BC in situ model was established. After the treatment, morphological changes in tumour tissue were detected by HE staining, and the apoptotic cells in tumour tissue were detected by TUNEL. The expression of related proteins was detected by Western blotting, and the proportion of CD4+ and CD8+ T cells in the spleen was detected by flow cytometry. RESULTS We found that the engineered Salmonella significantly inhibited the growth and incidence of tumours and increased the apoptosis of tumours. Importantly, engineered Salmonella carrying siRNA-PD-1 enhanced the anti-tumour immune response by inhibiting PD-1 expression and increased the CD8+ T cell infiltration in tumour tissue, while elevating the ratio of CD8+/CD4+ in spleens. CONCLUSION We demonstrated that engineered Salmonella siRNA-PD-1 exerted significant anti-tumour effects on BC.
Collapse
Affiliation(s)
- Qizhong Shi
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan, P.R. China
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, P.R. China
| | - Jiaxin Geng
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan, P.R. China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan, P.R. China
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan, P.R. China
| | - Lulu Han
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan, P.R. China
- Department of Urology, Xinxiang Central Hospital, Xinxiang, Henan, P.R. China
| | - Xingchan Ji
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan, P.R. China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan, P.R. China
| | - Jiaoran Li
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan, P.R. China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan, P.R. China
| | - Yonghui Mu
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan, P.R. China
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan, P.R. China
| | - Tiesuo Zhao
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan, P.R. China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan, P.R. China
| | - Lei Wang
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan, P.R. China
- Department of Urology, Xinxiang Central Hospital, Xinxiang, Henan, P.R. China
| | - Huijie Jia
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan, P.R. China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan, P.R. China
| |
Collapse
|
6
|
Yang Y, Zhu L, Xu Y, Liang L, Liu L, Chen X, Li H, Liu H. The progress and prospects of targeting the adenosine pathway in cancer immunotherapy. Biomark Res 2025; 13:75. [PMID: 40390144 PMCID: PMC12090549 DOI: 10.1186/s40364-025-00784-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/26/2025] [Indexed: 05/21/2025] Open
Abstract
Despite the notable success of cancer immunotherapy, its effectiveness is often limited in a significant proportion of patients, highlighting the need to explore alternative tumor immune evasion mechanisms. Adenosine, a key metabolite accumulating in hypoxic tumor regions, has emerged as a promising target in oncology. Inhibiting the adenosinergic pathway not only inhibits tumor progression but also holds potential to enhance immunotherapy outcomes. Multiple therapeutic strategies targeting this pathway are being explored, ranging from preclinical studies to clinical trials. This review examines the complex interactions between adenosine, its receptors, and the tumor microenvironment, proposing strategies to target the adenosinergic axis to boost anti-tumor immunity. It also evaluates early clinical data on pharmacological inhibitors of the adenosinergic pathway and discusses future directions for improving clinical responses.
Collapse
Affiliation(s)
- Yuying Yang
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Lin Zhu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yantao Xu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Long Liang
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Li Liu
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Xiang Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Hui Li
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Hong Liu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
7
|
Zhou S, Zhang L, Duan X, Liu K, Yingnan Y, Ma M, Han B. MiR-425-5p intervenes in autoimmune myocarditis by regulating Treg cell differentiation through NRAS. Front Cell Dev Biol 2025; 13:1600103. [PMID: 40433545 PMCID: PMC12106460 DOI: 10.3389/fcell.2025.1600103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Aim Our Previous research revealed significant differences in exosome-mediated intercellular miR-425a-5p between normal children and those with fulminant myocarditis. We sought to elucidate the molecular underpinnings and functional implications of miR-425a-5p in the context of myocarditis progression. Methods Bioinformatics techniques were employed to predict NRAS as the target gene of miR-425a-5p. We constructed a cellular myocarditis paradigm through LPS-mediated provocation of AC16 cardiomyocyte cultures. MiR-425a-5p was overexpressed, and the expressions of NRAS, cell apoptosis, and proinflammatory cytokine profiles, encompassing IL-1β, IL-6, and TNF-α, were comprehensively quantified. An experimental autoimmune myocarditis (EAM) mouse model was created using adeno-associated virus (AAV) for miR-425a-5p overexpression. Comprehensive histopathological analyses were conducted utilizing multiple staining techniques, including hematoxylin-eosin (HE), immunohistochemical, and Masson trichrome methodologies to characterize tissue responses. Results The study demonstrated that miR-425a-5p alleviated the inflammatory response in both AC16 cells and EAM mice through NRAS mediation. Single-cell data analysis of cardiac immune cells revealed that miR-425a-5p promoted Treg cell differentiation and improved cardiac function. Conclusion MiR-425a-5p plays a crucial role in modulating inflammatory responses in myocarditis, potentially offering a novel therapeutic strategy for managing the disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Bo Han
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
8
|
Esmaeilpour D, Ghomi M, Zare EN, Sillanpää M. Nanotechnology-Enhanced siRNA Delivery: Revolutionizing Cancer Therapy. ACS APPLIED BIO MATERIALS 2025. [PMID: 40354673 DOI: 10.1021/acsabm.5c00489] [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: 05/14/2025]
Abstract
RNA interference (RNAi) has emerged as a transformative approach for cancer therapy, enabling precise gene silencing through small interfering RNA (siRNA). However, the clinical application of siRNA-based treatments faces challenges such as rapid degradation, inefficient cellular uptake, and immune system clearance. Nanotechnology-enhanced siRNA delivery has revolutionized cancer therapy by addressing these limitations, improving siRNA stability, tumor-specific targeting, and therapeutic efficacy. Recent advancements in nanocarrier engineering have introduced innovative strategies to enhance the safety and precision of siRNA-based therapies, offering new opportunities for personalized medicine. This review highlights three key innovations in nanotechnology-enhanced siRNA delivery: artificial intelligence (AI)-driven nanocarrier design, multifunctional nanoparticles for combined therapeutic strategies, and biomimetic nanocarriers for enhanced biocompatibility. AI-driven nanocarriers utilize machine learning algorithms to optimize nanoparticle properties, improving drug release profiles and minimizing off-target effects. Multifunctional nanoparticles integrate siRNA with chemotherapy, immunotherapy, or photothermal therapy, enabling synergistic treatment approaches that enhance therapeutic outcomes and reduce drug resistance. Biomimetic nanocarriers, including exosome-mimicking systems and cell-membrane-coated nanoparticles, improve circulation time, immune evasion, and targeted tumor delivery. These innovations collectively enhance the precision, efficiency, and safety of siRNA-based cancer therapies. The scope and novelty of these advancements lie in their ability to overcome the primary barriers of siRNA delivery while paving the way for clinically viable solutions. This review provides a comprehensive analysis of the latest developments in nanocarrier fabrication, preclinical and clinical studies, and safety assessments. By integrating AI-driven design, multifunctionality, and biomimicry, nanotechnology-enhanced siRNA delivery holds immense potential for the future of precision cancer therapy.
Collapse
Affiliation(s)
- Donya Esmaeilpour
- Center for Nanotechnology in Drug Delivery, School of Pharmacy, Shiraz University of Medical Science, Shiraz 71345-1583, Iran
| | - Matineh Ghomi
- Chemistry Department, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz 6153753843 Iran
| | - Ehsan Nazarzadeh Zare
- School of Chemistry, Damghan University, Damghan 36716-45667, Iran
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India
| | - Mika Sillanpää
- Institute of Research and Development, Duy Tan University, Da Nang 550000, Vietnam
- School of Engineering & Technology, Duy Tan University, Da Nang 550000, Vietnam
| |
Collapse
|
9
|
Varvarà P, Cavallaro G, Mauro N. Poly(2-(diethylamino)ethyl methacrylate)-Functionalized Carbon Nanodots as Theranostic Platforms for siRNA Delivery and Survivin Silencing in Triple-Negative Breast Cancer. Biomacromolecules 2025. [PMID: 40347142 DOI: 10.1021/acs.biomac.5c00267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2025]
Abstract
This study describes the development of carbon nanodot (CDs)-based theranostic nanocarriers that integrate gene silencing with fluorescence imaging. Nitrogen- and sulfur-doped CDs were functionalized through controlled radical surface polymerization of 2-(diethylamino)ethyl methacrylate (DEAEMA), yielding self-tracking, cationic siRNA carriers CDs-pDEAEMA. The functionalization of CDs enhanced their fluorescence, broadening the emission spectrum toward the biologically transparent window. Fluorescent CDs-pDEAEMA effectively bound siRNA, remaining stable under physiological conditions, while in vitro studies proved their hemocompatibility and cytocompatibility on human dermal fibroblasts. Moreover, the ability to deliver BIRC5 siRNA was demonstrated in MDA-MB-231, successfully transfecting triple-negative breast cancer cells and resulting in an 80% reduction in the anti-apoptotic protein survivin. Furthermore, uptake studies demonstrated that the theranostic CDs are efficiently internalized in tumor cells and are clearly detectable by fluorescence imaging in the red region. These findings highlight the potential of CDs-pDEAEMA as an advanced theranostic tool for real-time tracking of siRNA therapy of breast cancer.
Collapse
Affiliation(s)
- Paola Varvarà
- Laboratory of Biocompatible Polymers, Department of "Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche" STEBICEF, University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
- Fondazione Veronesi, Piazza Velasca 5, 20122 Milano, Italy
| | - Gennara Cavallaro
- Laboratory of Biocompatible Polymers, Department of "Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche" STEBICEF, University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Nicolò Mauro
- Laboratory of Biocompatible Polymers, Department of "Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche" STEBICEF, University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| |
Collapse
|
10
|
Zhou S, Han B. Biological disturbance of MiR-425 and its application prospects in cardiovascular diseases. Front Cell Dev Biol 2025; 13:1593241. [PMID: 40417179 PMCID: PMC12098596 DOI: 10.3389/fcell.2025.1593241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 04/28/2025] [Indexed: 05/27/2025] Open
Abstract
MiR-425 is a biological molecule that has potential applications in cardiovascular diseases. It can regulate biological functions by combining with LncRNAs, binding with proteins, and changing the differentiation of immune cells. MiR-425 also has a role as a biomarker of disease. In cardiovascular diseases, it has clinical significance in reducing inflammation and heart repair, inducing angiogenesis, improving the prediction of atherosclerosis, reducing cardiac fibrosis, and regulating atrial natriuretic peptide to affect cardiovascular function. Target gene prediction and KEGG enrichment analysis are also mentioned.
Collapse
Affiliation(s)
- Shan Zhou
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong first Medical University, Jinan, Shandong, China
| | - Bo Han
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong first Medical University, Jinan, Shandong, China
- The Laboratory of Medical Science and Technology Innovation Center (Institute of Biomedical Engineering and Interdisciplinary Studies), Shandong First Medical University, jinan, China
| |
Collapse
|
11
|
Abolhasani S, Ahmadi Y, Fattahi D, Rostami Y, Chollou KM. microRNA-Mediated Regulation of Oxidative Stress in Cardiovascular Diseases. J Clin Lab Anal 2025; 39:e70017. [PMID: 40183484 PMCID: PMC12078765 DOI: 10.1002/jcla.70017] [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: 12/10/2024] [Revised: 02/08/2025] [Accepted: 03/07/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Cardiovascular diseases (CVDs) are the leading cause of mortality globally, often linked to oxidative stress. MicroRNAs (miRNAs) have emerged as significant regulators of oxidative stress within the cardiovascular system. OBJECTIVE This review examines the complex relationship between miRNAs and oxidative stress, clarifying their effects on gene expression pathways related to ROS production and detoxification in CVDs. METHODS From August to October 2024, we conducted a comprehensive search of PubMed, Scopus, Web of Science, and Google Scholar for studies published between 2014 and 2024 investigating the role of miRNAs in oxidative stress and cardiovascular diseases. RESULTS Specific miRNAs have been identified as critical regulators in the pathophysiology of CVDs, with distinct expression patterns correlated with conditions such as hypertension, coronary artery disease, and heart failure. For instance, miR-21 exacerbates oxidative stress by targeting genes essential for redox homeostasis, while miR-210 promotes endothelial cell survival under hypoxic conditions by mitigating ROS levels. CONCLUSION The reciprocal relationship between miRNAs and oxidative stress highlights the potential for therapeutic interventions targeting miRNA expression and activity in managing CVDs. Understanding these molecular mechanisms is vital for developing innovative strategies to address oxidative damage in cardiac tissues and improve cardiovascular health outcomes.
Collapse
Affiliation(s)
- Sakhavat Abolhasani
- Department of Basic Sciences and HealthSarab Faculty of Medical SciencesSarabEast AzerbaijanIran
| | - Yasin Ahmadi
- Department of Medical Laboratory ScienceKomar University of Science and TechnologySulaymaniyahKurdistan RegionIraq
| | - Davood Fattahi
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityLiverpoolUK
| | - Yavar Rostami
- Department of Basic Sciences and HealthSarab Faculty of Medical SciencesSarabEast AzerbaijanIran
| | - Khalil Maleki Chollou
- Department of Basic Sciences and HealthSarab Faculty of Medical SciencesSarabEast AzerbaijanIran
| |
Collapse
|
12
|
Lou Y, Wang Y, Lu J, Chen X. MicroRNA-targeted nanoparticle delivery systems for cancer therapy: current status and future prospects. Nanomedicine (Lond) 2025; 20:1181-1194. [PMID: 40231694 PMCID: PMC12068351 DOI: 10.1080/17435889.2025.2492542] [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: 12/17/2024] [Accepted: 04/09/2025] [Indexed: 04/16/2025] Open
Abstract
Recently, the regulatory effects of microRNAs (miRNAs) on gene expression have been exploited for applications in the diagnosis and treatment of cancer, neurological diseases, and cardiovascular diseases. However, the susceptibility of miRNAs to degradation during somatic circulation and the challenges associated with their delivery to target tissues and cells have limited the clinical application of miRNAs. For application in tumor therapy, it is essential for miRNAs to specifically target cancer cells. Therefore, various novel miRNA delivery systems that protect miRNA against the activity of serum nuclease and deliver miRNA to target cells have been developed and optimized. This review introduces the passive and active targeting strategies of nanoparticles, summarizes the recent progress of miRNA nanocarriers with tumor-targeting ability, and discusses various nanoparticle delivery systems and their antitumor applications. Additionally, this review focuses on the translational challenges and potential strategies for advancing miRNA-based therapies into the clinic.
Collapse
Affiliation(s)
- Yang Lou
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yutian Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Juan Lu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xi Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
13
|
Méndez-García A, García-Mendoza MA, Zárate-Peralta CP, Flores-Perez FV, Carmona-Ramirez LF, Pathak S, Banerjee A, Duttaroy AK, Paul S. Mitochondrial microRNAs (mitomiRs) as emerging biomarkers and therapeutic targets for chronic human diseases. Front Genet 2025; 16:1555563. [PMID: 40352788 PMCID: PMC12061977 DOI: 10.3389/fgene.2025.1555563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/10/2025] [Indexed: 05/14/2025] Open
Abstract
Mitochondria are membrane-bound cell organelles that undertake the majority of the energetic and metabolic processes within the cell. They are also responsible for mediating multiple apoptotic pathways, balancing redox charges, and scavenging reactive oxygen species. MicroRNAs, which are short, non-coding RNAs widely known for regulating gene expression at the post-transcriptional level, regulate many of these processes. The specific microRNAs that directly or indirectly control mitochondrial dynamics are called mitochondrial miRNAs (mitomiRs). The broadest classification of this type of ncRNA encompasses nuclear-encoded miRNAs that interact with cytoplasmatic mRNAs associated with mitochondrial activity. At the same time, a more specific subset comprises nuclear-encoded miRNAs that translocate into the mitochondria to interact with mRNAs inside of this organelle. Finally, the smallest group of mitomiRs includes those codified by mtDNA and can regulate endogenous mitochondrial transcripts or be transported into the cytoplasm to modulate circulating mRNAs. Regardless of the origin or action mechanism, mitomiRs have been recently recognized to have a key role in the progression of a variety of chronic disorders, such as neurodegenerative and cardiovascular diseases, diabetes, asthma, depression, and even cancer. All of these progressive pathologies have been tightly linked to mitochondrial dysregulation. They are further associated with an aberrant expression of specific miRNAs that regulate cellular metabolism, positioning mitomiRs as reliable biomarkers for diagnosing several chronic diseases. These molecular indicators have also provided insights into how these conditions progress, allowing for the development of different miRNA-based treatment strategies that target dysregulated mitochondrial-related genes, reestablishing their baseline activity and restricting further disease progression.
Collapse
Affiliation(s)
| | | | | | | | | | - Surajit Pathak
- Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chennai, India
| | - Antara Banerjee
- Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chennai, India
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Queretaro, Mexico
| |
Collapse
|
14
|
Wang X, Liang Y, Yang F, Shi Y, Shao R, Jing R, Yang T, Chu Q, An D, Zhou Q, Song J, Chen H, Liu C. Molecular mechanisms and targeted therapy of progranulin in metabolic diseases. Front Endocrinol (Lausanne) 2025; 16:1553794. [PMID: 40290306 PMCID: PMC12021630 DOI: 10.3389/fendo.2025.1553794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/17/2025] [Indexed: 04/30/2025] Open
Abstract
Progranulin (PGRN) is a secreted glycoprotein with cytokine-like properties, exerting tripartite mechanisms of inflammation suppression, tissue repair promotion, and metabolic regulation. This multifaceted functionality positions PGRN as a potential "multi-effect therapeutic strategy" for metabolic disorders characterised by cartilage degradation and imbalanced bone remodelling, potentially establishing it as a novel therapeutic target for such conditions. Osteoarthritis, rheumatoid arthritis, intervertebral disc degeneration, osteoporosis, periodontitis, and diabetes-related complications-representing the most prevalent metabolic diseases-currently lack effective treatments due to incomplete understanding of their precise pathogenic mechanisms. Recent studies have revealed that PGRN expression levels are closely associated with the onset and progression of these metabolic disorders. However, the exact regulatory role of PGRN in these diseases remains elusive, partly owing to its tissue-specific actions and context-dependent dual roles (anti-inflammatory vs. pro-inflammatory). In this review, we summarise the structure and functions of PGRN, explore its involvement in neurological disorders, immune-inflammatory diseases, and metabolic conditions, and specifically focus on its molecular mechanisms in metabolic diseases. Furthermore, we consolidate advances in targeting PGRN and the application of its engineered derivative, Atsttrin, in metabolic bone disorders. We also discuss potential unexplored mechanisms through which PGRN may exert influence within this field or other therapeutic domains. Collectively, this work aims to provide a new framework for elucidating PGRN's role in disease pathogenesis and advancing strategies for the prevention and treatment of metabolic disorders.
Collapse
Affiliation(s)
- Xiaxia Wang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yonglin Liang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Fan Yang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yangyang Shi
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Ruiwen Shao
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Ruge Jing
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Tong Yang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Qiao Chu
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Dong An
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Qi Zhou
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Jiayi Song
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Haolan Chen
- TCM Internal Medicine Department, Nanhu Community Health Centre, Pinliang, Gansu, China
| | - Chun Liu
- Library, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
15
|
Bi J, Wang Y, Wang Y. MiR-3613-5p targets AQP4 to promote the progression of chronic atrophic gastritis to gastric cancer. Front Pharmacol 2025; 16:1523689. [PMID: 40255569 PMCID: PMC12006049 DOI: 10.3389/fphar.2025.1523689] [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: 11/06/2024] [Accepted: 03/07/2025] [Indexed: 04/22/2025] Open
Abstract
Introduction: Gastric cancer (GC) exhibits high invasiveness, delayed diagnosis, and poor prognosis. Chronic atrophic gastritis (CAG), an initial stage within the Correa cascade, induces gastric mucosal inflammation and atrophy, promoting genetic and epigenetic alterations. MicroRNAs (miRNAs) dysregulation has been implicated in gastric tumorigenesis, yet their specific roles in CAG progression to GC remain unclear. Methods: Using clinical data from the GEO database, we identified miRNAs differentially expressed in gastric mucosa and serum samples from GC patients. Murine CAG models were established through administration of N-methyl-N-nitrosourea (MNU) and high-salt diet (HSD). In vitro functional assays evaluated proliferation and migration after miRNA modulation in gastric cancer cell lines. MiRNA target validation involved luciferase reporter assays. Results: MiR-3613-5p expression was significantly elevated in gastric mucosal and serum samples of GC patients, mucosal tissues of CAG patients, tumor tissues, and human gastric cancer cell lines. Murine models demonstrated increased miR-3613-5p expression in gastric mucosa following MNU and HSD-induced CAG. Functionally, miR-3613-5p overexpression promoted gastric cancer cell proliferation and migration in vitro, whereas silencing miR-3613-5p alleviated pathological gastric mucosal alterations (atrophy, hyperplasia, inflammatory infiltration) in vivo. Mechanistically, miR-3613-5p inhibited Aquaporin 4 (AQP4) expression by directly targeting its 3'UTR. Discussion: Our findings provide the first evidence that miR-3613-5p facilitates CAG progression toward GC via negative regulation of AQP4. These results highlight miR-3613-5p as a promising biomarker and therapeutic target, suggesting antagomiR-3613-5p as a potential novel strategy to prevent gastric carcinogenesis.
Collapse
Affiliation(s)
- Jian Bi
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yufen Wang
- Department of Digestive Endoscopy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yingde Wang
- Department of Gastroenterology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
16
|
Xu H, Zhao Q, Cai D, Chen X, Zhou X, Gao Y, Wu J, Yuan S, Li D, Zhang R, Peng W, Li G, Nan A. o8G-modified circKIAA1797 promotes lung cancer development by inhibiting cuproptosis. J Exp Clin Cancer Res 2025; 44:110. [PMID: 40176113 PMCID: PMC11963662 DOI: 10.1186/s13046-025-03365-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 03/13/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Lung cancer is a serious threat to human life and health, but effective screening and treatment methods are lacking. Circular RNAs (circRNAs) have important biological functions and are closely related to tumour development. Some studies have shown that the 8-oxo-7,8-dihydroguanosine (o8G) modification plays a key role in the disease process, but the effect of the o8G modification on circRNAs has not been elucidated. Moreover, cuproptosis is a novel mode of cell death in which copper ions directly promote protein aggregation and the disruption of cellular metabolic pathways. The present study revealed that the o8G modification of circKIAA1797 occurs and promotes lung cancer development by inhibiting cuproptosis, which provides new perspectives for epitranscriptomic studies and the development of novel therapeutic approaches for lung cancer. METHODS circRNA differential expression profiles in lung cancer were revealed via RNA high-throughput sequencing, and circKIAA1797 expression in lung cancer cell lines and tissues was detected using qPCR. Experiments such as o8G RNA immunoprecipitation (o8G RIP) and crosslinking immunoprecipitation (CLIP) were performed to explore the presence of o8G on circKIAA1797. The regulation of circKIAA1797 by the o8G reader Y-box binding protein 1 (YBX1) was explored using nuclear-cytoplasmic fractionation, actinomycin D (Act D) stability experiments and other experiments. circKIAA1797 silencing and overexpression systems were constructed for in vivo and in vitro experiments to study the role of circKIAA1797 in lung cancer development. Tagged RNA affinity purification (TRAP), RNA immunoprecipitation (RIP), coimmunoprecipitation (Co-IP), and immunofluorescence (IF) staining were subsequently conducted to reveal the molecular mechanism by which circKIAA1797 regulates cuproptosis and promotes lung cancer development. RESULTS This study is the first to reveal the presence of o8G on circKIAA1797 and that YBX1 is a reader that recognises ROS-induced circKIAA1797 o8G modifications and increases the stability and cytoplasmic expression of circKIAA1797. circKIAA1797, which is associated with the tumour stage and prognosis, has been shown to significantly promote the biological function of lung cancer development both in vivo and in vitro. This study revealed that circKIAA1797 inhibits intracellular cuproptosis by binding to the ferredoxin 1 (FDX1) mRNA, decreasing FDX1 mRNA stability, inhibiting FDX1 expression, and binding to the signal transducer and activator of transcription 1 (STAT1) protein and inhibiting lipoyltransferase 1 (LIPT1) transcription; moreover, circKIAA1797 promotes the closure of the mitochondrial permeability transition pore (mPTP), inhibits cuproptosis, and ultimately promotes lung cancer development. CONCLUSIONS This study revealed the presence of the o8G modification in circKIAA1797, which plays an important role in the development of lung cancer. circKIAA1797 can inhibit cuproptosis by inhibiting key cuproptosis proteins and promoting mPTP closure, ultimately promoting the development of lung cancer. This study provides not only a new theoretical basis for an in-depth understanding of the molecular mechanisms of lung cancer development but also a potential target for lung cancer treatment.
Collapse
Affiliation(s)
- Haotian Xu
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Qingyun Zhao
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Dunyu Cai
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Xingcai Chen
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Xiaodong Zhou
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Yihong Gao
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Jiaxi Wu
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Shengyi Yuan
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Deqing Li
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Ruirui Zhang
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Wenyi Peng
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Gang Li
- School of Public Health, Guangxi Medical University, Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China.
| | - Aruo Nan
- School of Public Health, Guangxi Medical University, Nanning, 530021, China.
- Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
17
|
Yan Y, Zhang Y, Liu J, Chen B, Wang Y. Emerging magic bullet: subcellular organelle-targeted cancer therapy. MEDICAL REVIEW (2021) 2025; 5:117-138. [PMID: 40224364 PMCID: PMC11987508 DOI: 10.1515/mr-2024-0044] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/27/2024] [Indexed: 04/15/2025]
Abstract
The therapeutic efficacy of anticancer drugs heavily relies on their concentration and retention at the corresponding target site. Hence, merely increasing the cellular concentration of drugs is insufficient to achieve satisfactory therapeutic outcomes, especially for the drugs that target specific intracellular sites. This necessitates the implementation of more precise targeting strategies to overcome the limitations posed by diffusion distribution and nonspecific interactions within cells. Consequently, subcellular organelle-targeted cancer therapy, characterized by its exceptional precision, have emerged as a promising approach to eradicate cancer cells through the specific disruption of subcellular organelles. Owing to several advantages including minimized dosage and side effect, optimized efficacy, and reversal of multidrug resistance, subcellular organelle-targeted therapies have garnered significant research interest in recent years. In this review, we comprehensively summarize the distribution of drug targets, targeted delivery strategies at various levels, and sophisticated strategies for targeting specific subcellular organelles. Additionally, we highlight the significance of subcellular targeting in cancer therapy and present essential considerations for its clinical translation.
Collapse
Affiliation(s)
- Yue Yan
- Department of Central Laboratory, Peking University First Hospital, Beijing, China
| | - Yimeng Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jianxiong Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Binlong Chen
- Department of Central Laboratory, Peking University First Hospital, Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Chemical Biology Center, Peking University, Beijing, China
| |
Collapse
|
18
|
Peng X, Pu F, Zhou F, Dai X, Xu F, Wang J, Feng J, Xia P. Has-miR-30c-1-3p inhibits macrophage autophagy and promotes Mycobacterium tuberculosis survival by targeting ATG4B and ATG9B. Sci Rep 2025; 15:10240. [PMID: 40133377 PMCID: PMC11937412 DOI: 10.1038/s41598-025-94452-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Autophagy is a widespread physiological process in the body, which also protects the host by degrading invading pathogens and harmful substances during pathological conditions. Nevertheless, Mycobacterium tuberculosis (MTB), the causative agent of tuberculosis, has evolved strategies to subvert autophagy by modulating microRNA (miRNA) expression, enabling its escape from host defenses. In this study, we established an in vitro model using the human macrophage cell line infected with the highly virulent MTB strain H37Rv. Through RNA sequencing and bioinformatic analysis post H37Rv infection, we screened 14 differentially expressed miRNAs. We predicted and demonstrated that miR-30c-1-3p inhibits autophagy and promotes MTB survival by targeting ATG4B and ATG9B during the infection process. The results showed that miR-30c-1-3p expression was gradually increased before 12 h of H37Rv infection, followed by a decrease. Overexpression of miR-30c-1-3p suppressed autophagic activity. We also identified the targeting of miR-30c-1-3p to ATG4B and ATG9B for the first time, and overexpression of both ATG4B and ATG9B, alone or together, on the basis with upregulation of miR-30c-1-3p reversed the inhibition of autophagy. Autophagy levels were analyzed at different levels by western blot, immunofluorescence, and transmission electron microscopy, all of which showed that upregulation of miR-30c-1-3p inhibited autophagy during H37Rv infection. Additionally, the intervention of miR-30c-1-3p mimics resulted in an increased bacterial load in macrophages, suggesting that MTB achieves immune evasion by upregulating miR-30c-1-3p during infection. In conclusion, our study provides a valuable target for the development of host-directed anti-tuberculosis therapy as well as a new diagnostic marker.
Collapse
Affiliation(s)
- Xianglin Peng
- Department of Orthopedics, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan, 430022, China
- Department of Orthopedics, Wuhan Fourth Hospital, Puai Hospital, Wuhan, 430030, China
| | - Feifei Pu
- Department of Orthopedics, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan, 430022, China
| | - Fangzheng Zhou
- Department of Orthopedics, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan, 430022, China
| | - Xiyong Dai
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan, 430022, China
| | - Feng Xu
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan, 430022, China
| | - Junwen Wang
- Department of Orthopedics, Wuhan Fourth Hospital, Puai Hospital, Wuhan, 430030, China
| | - Jing Feng
- Department of Orthopedics, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan, 430022, China.
| | - Ping Xia
- Department of Orthopedics, Wuhan Fourth Hospital, Puai Hospital, Wuhan, 430030, China.
| |
Collapse
|
19
|
Zhong Y, He JW, Huang CX, Lai HZ, Li XK, Zheng C, Fu X, You FM, Ma Q. The NcRNA/Wnt axis in lung cancer: oncogenic mechanisms, remarkable indicators and therapeutic targets. J Transl Med 2025; 23:326. [PMID: 40087753 PMCID: PMC11907837 DOI: 10.1186/s12967-025-06326-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 02/27/2025] [Indexed: 03/17/2025] Open
Abstract
Early diagnosis of lung cancer (LC) is challenging, treatment options are limited, and treatment resistance leads to poor prognosis and management in most patients. The Wnt/β-catenin signaling pathway plays a vital role in the occurrence, progression, and therapeutic response of LC. Recent studies indicate that non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) function as epigenetic regulators that can promote or inhibit Wnt/β-catenin signaling by interacting with Wnt proteins, receptors, signaling transducers, and transcriptional effectors, thereby affecting LC cell proliferation, metastasis, invasion, and treatment resistance. Deepening our understanding of the regulatory network between ncRNAs and the Wnt/β-catenin signaling pathway will help overcome the limitations of current LC diagnosis and treatment methods. This article comprehensively reviews the regulatory mechanisms related to the functions of ncRNAs and the Wnt/β-catenin pathway in LC, examining their potential as diagnostic and prognostic biomarkers and therapeutic targets, aiming to offer new promising perspectives for LC diagnosis and treatment.
Collapse
Affiliation(s)
- Yang Zhong
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610072, China
| | - Jia-Wei He
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610072, China
| | - Chun-Xia Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610072, China
| | - Heng-Zhou Lai
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610072, China
| | - Xue-Ke Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610072, China
| | - Chuan Zheng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610072, China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610072, China.
| | - Xi Fu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610072, China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610072, China.
| | - Feng-Ming You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610072, China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610072, China.
| | - Qiong Ma
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610072, China.
| |
Collapse
|
20
|
Dai C, Li Q, Wang L, Zhang J, Yang S, Zhang X. Long Noncoding LINC00115 Facilitates Cell Growth and Inhibits Apoptosis by Regulating the miR-4701-5p/P4HB Axis in Bladder Cancer. TOHOKU J EXP MED 2025; 265:69-81. [PMID: 39111879 DOI: 10.1620/tjem.2024.j075] [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: 03/18/2025]
Abstract
Bladder cancer (BCa) is a prevalent urogenital malignancy, imposing a significant burden on health-care systems worldwide. Long noncoding RNAs (lncRNAs) are important regulators of carcinogenesis and affect BCa progression. In this study, the influence of lncRNA LINC00115 on malignant behavior of BCa cells were explored. Bioinformatics method was used for prediction of gene expression and downstream molecules of LIN00115. LINC00115 expression level in BCa cells was measured using RT-qPCR. After LINC00115 depletion, the proportion of viable, proliferative, and apoptotic BCa cells were calculated by methyl thiazolyl tetrazolium (MTT) assays, colony formation assays, and TUNEL staining, respectively. FISH was performed to verify the cellular distribution of LINC00115. The interaction between LINC00115 and miR-4701-5p and the binding between miR-4701-5p and P4HB were confirmed using RNA pulldown, RNA immunoprecipitation (RIP), and luciferase reporter assays. Experimental results showed that LINC00115 was highly expressed in BCa cells. The silencing of LINC00115 restrained BCa cell proliferation and stimulated apoptosis. LINC00115 could directly bind to miR-4701-5p and thus initiate P4HB upregulation in BCa cells. P4HB 3'untranslated region could be targeted by miR-4701-5p. Additionally, Amplification of P4HB expression offset the effects of LINC00115 knockdown on BCa cell proliferative and apoptotic behaviors. In conclusion, LINC00115 facilitates BCa cell growth and inhibits apoptosis via interaction with miR-4701-5p and upregulation of P4HB.
Collapse
Affiliation(s)
- Changyuan Dai
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College
| | - Qingwen Li
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College
| | - Lili Wang
- Department of Emergency medicine, The First Affiliated Hospital of Bengbu Medical College
| | - Jiajun Zhang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College
| | - Shuai Yang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College
| | - Xiaole Zhang
- Department of Urology, The First Affiliated Hospital of Bengbu Medical College
| |
Collapse
|
21
|
Liu M, Wang Y, Zhang Y, Hu D, Tang L, Zhou B, Yang L. Landscape of small nucleic acid therapeutics: moving from the bench to the clinic as next-generation medicines. Signal Transduct Target Ther 2025; 10:73. [PMID: 40059188 PMCID: PMC11891339 DOI: 10.1038/s41392-024-02112-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/23/2024] [Accepted: 12/13/2024] [Indexed: 03/17/2025] Open
Abstract
The ability of small nucleic acids to modulate gene expression via a range of processes has been widely explored. Compared with conventional treatments, small nucleic acid therapeutics have the potential to achieve long-lasting or even curative effects via gene editing. As a result of recent technological advances, efficient small nucleic acid delivery for therapeutic and biomedical applications has been achieved, accelerating their clinical translation. Here, we review the increasing number of small nucleic acid therapeutic classes and the most common chemical modifications and delivery platforms. We also discuss the key advances in the design, development and therapeutic application of each delivery platform. Furthermore, this review presents comprehensive profiles of currently approved small nucleic acid drugs, including 11 antisense oligonucleotides (ASOs), 2 aptamers and 6 siRNA drugs, summarizing their modifications, disease-specific mechanisms of action and delivery strategies. Other candidates whose clinical trial status has been recorded and updated are also discussed. We also consider strategic issues such as important safety considerations, novel vectors and hurdles for translating academic breakthroughs to the clinic. Small nucleic acid therapeutics have produced favorable results in clinical trials and have the potential to address previously "undruggable" targets, suggesting that they could be useful for guiding the development of additional clinical candidates.
Collapse
Affiliation(s)
- Mohan Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yusi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yibing Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Die Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bailing Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
22
|
Li Y, Gan J, Lei J, Qi S, Yu X, Zhang W, Feng Y, Zhang Y, Cheng M, Ma L, Mao Z, Liu Z, Yu G. Catalytic Hybrid Lipid Nanoparticles Potentiate Circle RNA-Based Cytokine Immunotherapy. ACS NANO 2025; 19:7864-7876. [PMID: 39977421 DOI: 10.1021/acsnano.4c14517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Cytokine therapeutics in cancer immunotherapy are greatly limited by their short half-time, serious toxicity, and frequent administration, which can possibly be addressed by ribonucleic acid (RNA) technology through the expression of targeting cytokines in situ. However, the intracellular translation of RNA remains restricted due to the generation of excessive reactive oxygen species (ROS) and overconsumption of adenosine triphosphate (ATP) within the transfected cells. Herein, hybrid lipid nanoparticles (Mn-LNPs) are developed by incorporating small-sized trimanganese tetraoxide nanoparticles within conventional lipid nanoparticles, showing the ability to generate oxygen, eliminate ROS, and boost intracellular ATP, thus greatly enhancing the translation efficiency. This hybrid platform is employed to encapsulate interleukin 12 (IL-12)-encoding circular RNA (Mn-LNPs@RNAIL-12) for tumor immunotherapy, exhibiting unparalleled advantages in the proliferation of cytotoxic T cells and stimulation of antitumor immunity. Moreover, the antitumor efficacy of Mn-LNPs@RNAIL-12 is further strengthened by synergizing with immune checkpoint blockade therapy to achieve durable and potent antitumor performances.
Collapse
Affiliation(s)
- Yongcan Li
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Jinqun Gan
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Jiaqi Lei
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Shaolong Qi
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Xinyang Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Weibing Zhang
- Shanxi Academy of Advanced Research and Innovation, Taiyuan 030032, P. R. China
| | - Yunxuan Feng
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Yundong Zhang
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Meiqi Cheng
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Lie Ma
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Zhida Liu
- Shanxi Academy of Advanced Research and Innovation, Taiyuan 030032, P. R. China
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
23
|
Li M, Wu L, Si H, Wu Y, Liu Y, Zeng Y, Shen B. Engineered mitochondria in diseases: mechanisms, strategies, and applications. Signal Transduct Target Ther 2025; 10:71. [PMID: 40025039 PMCID: PMC11873319 DOI: 10.1038/s41392-024-02081-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/30/2024] [Accepted: 11/17/2024] [Indexed: 03/04/2025] Open
Abstract
Mitochondrial diseases represent one of the most prevalent and debilitating categories of hereditary disorders, characterized by significant genetic, biological, and clinical heterogeneity, which has driven the development of the field of engineered mitochondria. With the growing recognition of the pathogenic role of damaged mitochondria in aging, oxidative disorders, inflammatory diseases, and cancer, the application of engineered mitochondria has expanded to those non-hereditary contexts (sometimes referred to as mitochondria-related diseases). Due to their unique non-eukaryotic origins and endosymbiotic relationship, mitochondria are considered highly suitable for gene editing and intercellular transplantation, and remarkable progress has been achieved in two promising therapeutic strategies-mitochondrial gene editing and artificial mitochondrial transfer (collectively referred to as engineered mitochondria in this review) over the past two decades. Here, we provide a comprehensive review of the mechanisms and recent advancements in the development of engineered mitochondria for therapeutic applications, alongside a concise summary of potential clinical implications and supporting evidence from preclinical and clinical studies. Additionally, an emerging and potentially feasible approach involves ex vivo mitochondrial editing, followed by selection and transplantation, which holds the potential to overcome limitations such as reduced in vivo operability and the introduction of allogeneic mitochondrial heterogeneity, thereby broadening the applicability of engineered mitochondria.
Collapse
Affiliation(s)
- Mingyang Li
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Limin Wu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Haibo Si
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuangang Wu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuan Liu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yi Zeng
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Bin Shen
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
24
|
Bellavita R, Braccia S, Piccolo M, Bialecki P, Ferraro MG, Graziano SF, Esposito E, Donadio F, Bryszewska M, Irace C, Pedziwiatr-Werbicka E, Falanga A, Galdiero S. Shielding siRNA by peptide-based nanofibers: An efficient approach for turning off EGFR gene in breast cancer. Int J Biol Macromol 2025; 292:139219. [PMID: 39733890 DOI: 10.1016/j.ijbiomac.2024.139219] [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: 08/01/2024] [Revised: 12/24/2024] [Accepted: 12/24/2024] [Indexed: 12/31/2024]
Abstract
Peptide-based self-assembled nanosystems show great promise as non-viral gene and siRNA delivery vectors. In the current study, we designed and functionalized nanofibers for the delivery of siRNA, targeting and silencing EGFR gene overexpressed in triple-negative breast cancer. The nanofiber-mediated siRNA delivery was characterized in terms of zeta potential, morphology, and structural stability by circular dichroism spectroscopy. In cytotoxicity studies, nanofibers presented high biocompatibility showing a negligible effect on cell viability both on healthy and cancer cell lines. The binding between nanofibers and EGFR-siRNA was investigated and ascertained by performing different biophysical studies. The complex siRNA:NF was stable over time, under fetal bovine serum, temperature and ionic strength effects. Moreover, nanofibers effectiveness in stabilizing and delivering an ad hoc selected siRNA for EGFR gene expression silencing was verified in a preclinical model of triple-negative breast cancer. Specifically, a significant gene knockdown was obtained with the complex siRNA:NF, that is comparable with the effect obtained by lipofectamine/siRNA transfection. This effective gene silencing derived from the successful internalization of nanofibers by cancer cells as observed by confocal microscopy. These results suggested that this peptide-based nanofiber could be an effective and safe systemic siRNA delivery system for application in biomedical areas.
Collapse
Affiliation(s)
- Rosa Bellavita
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Simone Braccia
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Marialuisa Piccolo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Piotr Bialecki
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143 st., 90-236 Lodz, Poland
| | - Maria Grazia Ferraro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Sossio Fabio Graziano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Emanuela Esposito
- Institute of Applied Sciences and Intelligent Systems (ISASI), Naples Cryo Electron Microscopy Laboratory - EYE LAB, National Research Council (CNR), Via Pietro Castellino 111, 80131 Naples, Italy
| | - Federica Donadio
- Institute of Applied Sciences and Intelligent Systems (ISASI), Naples Cryo Electron Microscopy Laboratory - EYE LAB, National Research Council (CNR), Via Pietro Castellino 111, 80131 Naples, Italy
| | - Maria Bryszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143 st., 90-236 Lodz, Poland
| | - Carlo Irace
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Elzbieta Pedziwiatr-Werbicka
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143 st., 90-236 Lodz, Poland
| | - Annarita Falanga
- Department of Agricultural Science, University of Naples Federico II, Via Università 100, Portici, 80055 Portici, Italy
| | - Stefania Galdiero
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy.
| |
Collapse
|
25
|
Gandhi N, Modi S, Soni S, Andey T. Modular self-emulsifying drug delivery platform to enhance cellular uptake activity in triple-negative breast cancer. Eur J Pharm Sci 2025; 206:106993. [PMID: 39708842 DOI: 10.1016/j.ejps.2024.106993] [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: 07/22/2023] [Revised: 11/08/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Triple-negative breast cancer (TNBC) presents with resistance phenotypes to certain therapies, such as cisplatin, often requiring higher dosing, with associated acquired tumor resistance, renal toxicity, and variable patient responses. A self-emulsifying drug delivery (SEDD) formulation approach was proposed to overcome the limitations of cisplatin in TNBC, focusing on improving intracellular cisplatin and control siRNA uptake as a proof-of-principle of dual drug delivery. Four SEDD formulations were prepared and optimized for cisplatin (o/w) emulsion and FITC-siRNA (w/o) emulsion using pseudo-ternary phase diagrams to facilitate the formation of water-in-oil-water (w/o/w) emulsions. Formulations were characterized by size, polydispersity (PDI), and surface charge and tested in vitro. Cellular uptake via triplex staining of drug-loaded SEDDs was investigated. SEDDs showed enhanced internalization and promoted selective TNBC cellular uptake. The current study is a proof-of-principle for the successful co-delivery of cisplatin (small molecule) and siRNA (large molecule) via the SEDDs platform.
Collapse
Affiliation(s)
- Nandini Gandhi
- Massachusetts College of Pharmacy and Health Sciences (MCPHS University) Department of Pharmaceutical Sciences, School of Pharmacy, 19 Foster St., Worcester, MA 01608, USA
| | - Shail Modi
- Massachusetts College of Pharmacy and Health Sciences (MCPHS University) Department of Pharmaceutical Sciences, School of Pharmacy, 19 Foster St., Worcester, MA 01608, USA
| | - Shailvi Soni
- Massachusetts College of Pharmacy and Health Sciences (MCPHS University) Department of Pharmaceutical Sciences, School of Pharmacy, 19 Foster St., Worcester, MA 01608, USA
| | - Terrick Andey
- Massachusetts College of Pharmacy and Health Sciences (MCPHS University) Department of Pharmaceutical Sciences, School of Pharmacy, 19 Foster St., Worcester, MA 01608, USA.
| |
Collapse
|
26
|
Yuan F, Tang Y, Liang H, Cao M, Ren Y, Li Y, Yang G, Zhong Z, Xiong Z, He Z, Deng M, Yao Z. CircPIK3C3 inhibits hepatocellular carcinoma progression and lenvatinib resistance by suppressing the Wnt/β-catenin pathway via the miR-452-5p/SOX15 axis. Genomics 2025; 117:110999. [PMID: 39863187 DOI: 10.1016/j.ygeno.2025.110999] [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: 04/22/2024] [Revised: 12/28/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025]
Abstract
INTRODUCTION Resistance to lenvatinib limits the effectiveness of the targeted treatments for HCC. However, the exact mechanism behind this resistance remains elusive. Current research suggests that circular RNA (circRNA) is pivotal in mediating drug resistance during targeted treatments. OBJECTIVES To investigate the influence of circRNA on HCC progression and its resistance to lenvatinib. METHODS We identified the crucial circRNA hsa_circ_0005711 (circPIK3C3) through bioinformatics. Study (in-vitro and in-vivo) on the expression of circPIK3C3 (measured by qRT-PCR) and its association with progress of HCC patients including lenvatinib resistance were performed. Techniques such as dual-luciferase reporter assays, RNA FISH, RAP, and AGO2-RIP were employed for discerning circPIK3C3's specific mechanisms related to progression of HCC and its lenvatinib resistance. RESULTS Study (in-vitro and in-vivo) revealed that circPIK3C3 exhibited reduced expression and lenvatinib resistance in HCC, which was intimately tied to patient outcomes. Moreover, circPIK3C3 elevated SOX15 expression while suppressing the signaling pathway related to Wnt/β-catenin via inhibition of miR-452-5p through a competitive endogenous RNA (ceRNA) network. This, in turn, mitigated HCC progression and its resistance to lenvatinib. CONCLUSION CircPIK3C3 is instrumental in the disease progression and resistance to Lenvatinib in HCC. It presents a potential therapeutic avenue for patients with lenvatinib-resistant HCC and could serve as a valuable molecular marker for forecasting lenvatinib resistance in HCC patients.
Collapse
Affiliation(s)
- Feng Yuan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China; Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yongchang Tang
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Department of General Surgery, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Hao Liang
- Department of General Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Mingbo Cao
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yupeng Ren
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yuxuan Li
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Gaoyuan Yang
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Zhaozhong Zhong
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China; Department of Kidney Transplantation, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Zhiyong Xiong
- Department of Kidney Transplantation, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Zhiwei He
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Meihai Deng
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Zhicheng Yao
- Department of General Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| |
Collapse
|
27
|
Zhou JH, Cai C, Zhou XF, Xu D. Progress in research of non-coding RNAs in colorectal cancer and their application in early diagnosis. Shijie Huaren Xiaohua Zazhi 2025; 33:96-105. [DOI: 10.11569/wcjd.v33.i2.96] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/03/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025] Open
Abstract
Colorectal cancer is one of the common malignant tumors in the digestive system worldwide, with its incidence and mortality rates ranking high among various diseases. Although certain advancements have been achieved in the diagnosis and treatment techniques of colorectal cancer in recent years, the existing diagnostic means and treatment methods still present numerous limitations, significantly influencing the early detection, precise diagnosis, and individualized treatment of colorectal cancer. With the in-depth research of molecular biology, non-coding RNAs (ncRNAs) have gained increasing attention in the development and prognosis evaluation of colorectal cancer. This paper summarizes some studies related to the expression of different types of ncRNAs in colorectal cancer and selects a portion of ncRNAs that are expected to serve as new diagnostic indicators for this malignancy. The emergence of new biological indicators will contribute to the early diagnosis of colorectal cancer, facilitating its early detection and even prevention.
Collapse
Affiliation(s)
- Jin-Hang Zhou
- People's Hospital of Wucheng District, Jinhua City, Jinhua 321000, Zhejiang Province, China
| | - Cheng Cai
- Department of Colorectal and Anal Surgery, Jinhua Central Hospital, Jinhua 321000, Zhejiang Province, China
| | - Xiao-Feng Zhou
- People's Hospital of Wucheng District, Jinhua City, Jinhua 321000, Zhejiang Province, China
| | - Dan Xu
- People's Hospital of Wucheng District, Jinhua City, Jinhua 321000, Zhejiang Province, China
| |
Collapse
|
28
|
Yi J, Du J, Chen X, Nie RC, Hu GS, Wang L, Zhang YY, Chen S, Wen XS, Luo DX, He H, Liu W. A circRNA-mRNA pairing mechanism regulates tumor growth and endocrine therapy resistance in ER-positive breast cancer. Proc Natl Acad Sci U S A 2025; 122:e2420383122. [PMID: 40233410 PMCID: PMC11874584 DOI: 10.1073/pnas.2420383122] [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: 10/04/2024] [Accepted: 01/13/2025] [Indexed: 04/17/2025] Open
Abstract
The molecular mechanisms underlying estrogen receptor (ER)-positive breast carcinogenesis and drug resistance remain incompletely understood. Elevated expression of CCND1 is linked to enhanced invasiveness, poorer prognosis, and resistance to drug therapies in ER-positive breast cancer. In this study, we identify a highly expressed circular RNA (circRNA) derived from FOXK2, called circFOXK2, which plays a key role in stabilizing CCND1 mRNA, thereby promoting cell cycle progression, cell growth, and endocrine therapy resistance in ER-positive breast cancer cells. Mechanistically, circFOXK2 binds directly to CCND1 mRNA via RNA-RNA pairing and recruits the RNA-binding protein ELAVL1/HuR, stabilizing the CCND1 mRNA and enhancing CCND1 protein levels. This results in activation of the CCND1-CDK4/6-p-RB-E2F signaling axis, driving the transcription of downstream E2F target genes and facilitating the G1/S transition during cell cycle progression. Notably, targeting circFOXK2 with antisense oligonucleotide (ASO-circFOXK2) suppresses ER-positive breast cancer cell growth both in vitro and in vivo. Moreover, combination therapy with ASO-circFOXK2 and tamoxifen exhibits synergistic effects and restores tamoxifen sensitivity in tamoxifen-resistant cells. Clinically, high circFOXK2 expression is positively correlated with CCND1 levels in both ER-positive breast cancer cell lines and patient tumor tissues. Overall, our findings reveal the critical role of circFOXK2 in stabilizing the oncogene CCND1 and promoting cancer progression, positioning circFOXK2 as a potential therapeutic target for ER-positive breast cancer in clinical settings.
Collapse
MESH Headings
- Humans
- Breast Neoplasms/genetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Female
- Drug Resistance, Neoplasm/genetics
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Cyclin D1/genetics
- Cyclin D1/metabolism
- Animals
- Mice
- Cell Line, Tumor
- Cell Proliferation/genetics
- Cell Proliferation/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/genetics
- Gene Expression Regulation, Neoplastic
- Antineoplastic Agents, Hormonal/pharmacology
- ELAV-Like Protein 1/metabolism
- ELAV-Like Protein 1/genetics
- Tamoxifen/pharmacology
- Mice, Nude
- MCF-7 Cells
Collapse
Affiliation(s)
- Jia Yi
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Yu-Yue Pathology Scientific Research Center, Chongqing400039, China
- Jinfeng Laboratory, Chongqing401329, China
| | - Jiao Du
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
| | - Xue Chen
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
| | - Rui-chao Nie
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen361102, Fujian, China
| | - Guo-sheng Hu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
| | - Lei Wang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
| | - Yue-ying Zhang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
| | - Shang Chen
- Laboratory Medicine Centre, Shenzhen Nanshan People’s Hospital, Shenzhen518052, Guangdong, China
| | - Xiao-sha Wen
- Laboratory Medicine Centre, Shenzhen Nanshan People’s Hospital, Shenzhen518052, Guangdong, China
| | - Di-xian Luo
- The Third Affiliated Hospital (Luohu Hospital), Shenzhen University, Shenzhen518000, Guangdong, China
| | - Hua He
- Department of Neurosurgery, Third Affiliated Hospital, Naval Medical University, Shanghai200438, China
| | - Wen Liu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Xiamen University, Xiamen361102, Fujian, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen361102, Fujian, China
| |
Collapse
|
29
|
Yang J, Luo Y, Yao Z, Wang Z, Jiang K. Theoretical perspectives and clinical applications of non-coding RNA in lung cancer metastasis: a systematic review. Discov Oncol 2025; 16:169. [PMID: 39937377 PMCID: PMC11822152 DOI: 10.1007/s12672-025-01919-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
Lung cancer is one of the deadliest malignancies worldwide, with distant metastasis being a major cause of death. However, the specific mechanisms of lung cancer metastasis remain unclear. NcRNAs, a widely present type of non-coding RNAs in the body, constitute about 98% of the human genome, lacking protein-coding capacity but involved in various cellular processes such as proliferation, apoptosis, invasion, and migration. Studies have shown that ncRNAs play a crucial role in the metastasis of lung cancer, although research in this area is limited. This review summarizes the biological origins and functions of ncRNAs, their specific roles and mechanisms in lung cancer metastasis, and discusses their potential for early screening and therapeutic applications in lung cancer. Furthermore, it outlines the challenges in translating basic advancements of ncRNAs in lung cancer metastasis into clinical practice.
Collapse
Affiliation(s)
- Jie Yang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, People's Republic of China
| | - Yi Luo
- The Clinical Medical College, Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Zuhuan Yao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, People's Republic of China
| | - Zhaokai Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, People's Republic of China
| | - Ke Jiang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, Hubei, People's Republic of China.
| |
Collapse
|
30
|
Zeng J, Tong S, Liu J, Liu S, Mungur R, Chen S. MiR-433 inhibits cell invasion of glioblastoma via direct targeting TRPM8 based on bioinformatic analysis and experimental validation. Gene 2025; 936:149121. [PMID: 39581355 DOI: 10.1016/j.gene.2024.149121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Understanding the essential role of miRNA in regulating cell invasion in glioblastoma opens up new avenues for targeted therapeutic interventions in the future. By screening out eligible miRNA expression data sets from the GEO database, the WGCNA package based on the R language is further used to construct a co-expression network model of the chip data set, to identify modules related to disease states and perform pivotal miRNA screening on the related modules. The target relationship between miRNA and TRPM8 was verified by bioinformatics and luciferase gene report, and the effect of miRNA overexpression on TRPM8 protein level was analyzed by Western blot. The result of miR-433 overexpression on the invasion ability of glioblastoma cells in vitro was examined by scratch test and Transwell invasion test. The results of this study indicate that the selected target miR-433 has a strong binding relationship with TRPM8 and can effectively regulate its expression. Furthermore, overexpression of miR-433 was found to inhibit the invasion ability of glioblastoma cells by targeting TRPM8. These data demonstrate that miR-433 can target TRPM8 to inhibit glioblastoma cell invasion.
Collapse
Affiliation(s)
- Jianping Zeng
- Department of Neurosurgery, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University. Nanchang 330006, Jiangxi Province, PR China.
| | - Shoufang Tong
- Department of Transfusion Medicine, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang Provincial People's Hospital) Hangzhou Medical College, Taizhou, Zhejiang, PR China
| | - Jing Liu
- Department of Pharmacy, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University. Nanchang 330006, Jiangxi Province, PR China
| | - Shuai Liu
- Department of Neurosurgery, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University. Nanchang 330006, Jiangxi Province, PR China
| | - Rajneesh Mungur
- Department of Neurosurgery, The First Affiliated Hospital of Zhejiang University, Hangzhou 310000, Zhejiang Province, PR China
| | - Shangshi Chen
- Department of Neurosurgery, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University. Nanchang 330006, Jiangxi Province, PR China.
| |
Collapse
|
31
|
Tomita T. Non-vesicular extracellular RNA: A potential drug target to intervene cell-cell communication. Pharmacol Ther 2025; 266:108774. [PMID: 39644926 DOI: 10.1016/j.pharmthera.2024.108774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
The importance of non-vesicular extracellular RNA in the mammalian system is becoming increasingly apparent. Non-vesicular extracellular RNA is defined as RNA molecules not included in a lipid bilayer such as exosomes. Because non-vesicular extracellular RNA is not protected from RNases and is therefore rapidly degraded, they were not easily captured by conventional biofluid analyses. Recent publications showed that some non-vesicular extracellular RNAs are relatively stable in biofluids or tissue culture media, and they have unique biological functions. Major RNAs (rRNA, mRNA, and tRNA) and other non-cording RNAs play important roles in transcription or translation in the cell. In contrast, non-vesicular extracellular RNA has functions related to intercellular communication rather than protein synthesis. This review discusses the basics of non-vesicular extracellular RNA, including its definition, purification, receptors, and future prospects as a drug target.
Collapse
Affiliation(s)
- Takeshi Tomita
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, School of Medicine, Japan; Department of Biochemistry and Molecular Biology, Shinshu University, School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan.
| |
Collapse
|
32
|
Zhang H, Zhang L. Live-Cell RNA Imaging with a DNA-Functionalized Metal-Organic Framework-Based Fluorescent Probe. Methods Mol Biol 2025; 2875:59-70. [PMID: 39535639 DOI: 10.1007/978-1-0716-4248-1_5] [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: 11/16/2024]
Abstract
Live-cell fluorescence imaging of tumor-associated miRNAs is significant for understanding the cancer onset and progression and the diagnosis and prognosis of clinical diseases. In this protocol, we describe the construction of a DNA-functionalized metal-organic framework-based fluorescent probe and demonstrate that the probe can be used to detect miRNA in vitro and live-cell imaging. The formed DNA-MOF probe can specifically target miRNA with high sensitivity and specificity. The detection limit of the extracellular assay is as low as the picomolar level. In addition, we found that the probe could permeate cells and can be successfully applied in intracellular miRNA imaging, even achieving the distinction of cancer cells and normal cells, as well as the cancer cell lines with different miRNA expression levels.
Collapse
Affiliation(s)
- Hongyan Zhang
- Department of Chemistry, Tianjin Key Laboratory of Molecular Optoelectronic Sciences, School of Science, Tianjin University, Tianjin, People's Republic of China
| | - Libing Zhang
- Department of Chemistry, Tianjin Key Laboratory of Molecular Optoelectronic Sciences, School of Science, Tianjin University, Tianjin, People's Republic of China.
| |
Collapse
|
33
|
Peng X, Wang T, Dai B, Zhu Y, Ji M, Yang P, Zhang J, Liu W, Miao Y, Liu Y, Wang S, Sun J. Gene Therapy for Inflammatory Cascade in Intrauterine Injury with Engineered Extracellular Vesicles Hybrid Snail Mucus-enhanced Adhesive Hydrogels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410769. [PMID: 39454114 PMCID: PMC11714243 DOI: 10.1002/advs.202410769] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Indexed: 10/27/2024]
Abstract
Early hyper-inflammation caused by intrauterine injury triggered subsequent intrauterine adhesion (IUA). STAT1-mediated M1 macrophages are confirmed to secrete pro-inflammatory cytokines to accelerate inflammatory cascade and IUA formation by multi-omics analysis and experimental verification. However, clinically used hyaluronic acid (HA) hydrogels are prone to slip out of injury sites due to poor bio-adhesion properties. Therefore, there are still challenges in applying hydrogels for M1 macrophage intervention in IUA treatment. Herein, an engineered extracellular vesicles (EVs) hybrid snail mucus (SM)-enhanced adhesive hydrogels to improve bio-adhesion property is fabricated and M1 macrophage intervention through targeting delivery and STAT1 silencing is achieved. First, inspired by the high bio-adhesion capacity of SM, SM and gelatin methacrylate (GelMA) solution are mixed to construct GelMA/SM (GS) hydrogel. Then, folic acid-modified extracellular vesicles (FA-EVs) are synthesized for targeting the delivery of STAT1-siRNA. Upon injection of FA-EVs hybrid GS hydrogel into the uterine cavity, a protective hydrogel layer forms on the surface of injury sites and sustains the release of STAT1-siRNA-loaded FA-EVs to curtail M1 macrophages generation through inhibiting STAT1 phosphorylation, resulting in reduction of myofibroblasts activation and collagen deposition. In addition, the pregnancy rate and the number of fetuses in rats treated with this hydrogel were much higher than those in other groups, suggesting that the hydrogel could promote functional endometrial regeneration and restore fertility. Overall, this study presents a promising strategy for employing FA-EVs hybrid adhesive hydrogel with superior bio-adhesion properties and M1 macrophage targeting delivery for IUA treatment and uterus recovery.
Collapse
Affiliation(s)
- Xiaotong Peng
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Tao Wang
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Bo Dai
- Department of HematologyHuashan HospitalFudan UniversityShanghai200040China
| | - Yiping Zhu
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Mei Ji
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Pusheng Yang
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Jiaxin Zhang
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Wenwen Liu
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Yaxin Miao
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| | - Yonghang Liu
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Shuo Wang
- Department of OrthopaedicsShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200233China
| | - Jing Sun
- Department of GynecologyShanghai Key Laboratory of Maternal Fetal MedicineShanghai Institute of Maternal‐Fetal Medicine and Gynecologic OncologyShanghai First Maternity and Infant HospitalSchool of MedicineTongji UniversityShanghai200092China
| |
Collapse
|
34
|
Kole E, Jadhav K, Singh R, Mandpe S, Abhang A, Verma RK, Naik J. Recent Developments in Tyrosine Kinase Inhibitor-based Nanotherapeutics for EGFR-resistant Non-small Cell Lung Cancer. Curr Drug Deliv 2025; 22:249-260. [PMID: 38275043 DOI: 10.2174/0115672018278617231207051907] [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: 08/23/2023] [Revised: 10/18/2023] [Accepted: 11/20/2023] [Indexed: 01/27/2024]
Abstract
The advent of drug resistance in response to epidermal growth factor receptor (EGFR)- tyrosine kinase inhibitor (TKI) targeted therapy represents a serious challenge in the management of non-small cell lung cancer (NSCLC). These acquired resistance mutations, attributed to several advanced EGFR mutations and, necessitated the development of new-generation TKIs. Nanomedicine approaches provide a plausible way to address these problems by providing targeted delivery and sustained release, which have demonstrated success in preclinical trials. This review article provides a summary of nano-formulations designed for EGFR-TKI-resistant NSCLC, highlighting their efficacy in both in vitro and in vivo models. These findings reveal insights into the design of nanoparticles and multifunctional nanosystems, offering a potential avenue for efficacious treatment of EGFR-TKIresistant NSCLC.
Collapse
Affiliation(s)
- Eknath Kole
- Department of Pharmaceutical Technology, University Institute of Chemical Technology, KBC North Maharashtra University, Jalgaon M.S., 425001, India
| | - Krishna Jadhav
- Pharmaceutical Nanotechnology Lab, Institute of Nano Science and Technology, Sahibzada Ajit Singh Nagar (Mohali), Punjab, 140306, India
| | - Raghuraj Singh
- Pharmaceutical Nanotechnology Lab, Institute of Nano Science and Technology, Sahibzada Ajit Singh Nagar (Mohali), Punjab, 140306, India
| | - Shilpa Mandpe
- Department of Pharmaceutical Technology, University Institute of Chemical Technology, KBC North Maharashtra University, Jalgaon M.S., 425001, India
| | - Ashwin Abhang
- Department of Biopharmaceutics, Biocon Bristol-Myers Squibb R&D Center (BBRC), Bangalore, India
| | - Rahul K Verma
- Pharmaceutical Nanotechnology Lab, Institute of Nano Science and Technology, Sahibzada Ajit Singh Nagar (Mohali), Punjab, 140306, India
| | - Jitendra Naik
- Department of Pharmaceutical Technology, University Institute of Chemical Technology, KBC North Maharashtra University, Jalgaon M.S., 425001, India
| |
Collapse
|
35
|
Zhang Y, Zhang M, Song H, Dai Q, Liu C. Tumor Microenvironment-Responsive Polymer-Based RNA Delivery Systems for Cancer Treatment. SMALL METHODS 2025; 9:e2400278. [PMID: 38803312 DOI: 10.1002/smtd.202400278] [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: 02/25/2024] [Revised: 03/30/2024] [Indexed: 05/29/2024]
Abstract
Ribonucleic acid (RNA) therapeutics offer a broad prospect in cancer treatment. However, their successful application requires overcoming various physiological barriers to effectively deliver RNAs to the target sites. Currently, a number of RNA delivery systems based on polymeric nanoparticles are developed to overcome these barriers in RNA delivery. This work provides an overview of the existing RNA therapeutics for cancer gene therapy, and particularly summarizes those that are entering the clinical phase. This work then discusses the core features and latest research developments of tumor microenvironment-responsive polymer-based RNA delivery carriers which are designed based on the pathological characteristics of the tumor microenvironment. Finally, this work also proposes opportunities for the transformation of RNA therapies into cancer immunotherapy methods in clinical applications.
Collapse
Affiliation(s)
- Yahan Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Ming Zhang
- Department of Pathology, Peking University International Hospital, Beijing, 102206, China
| | - Haiqin Song
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025, China
| | - Qiong Dai
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
36
|
Pallathadka H, Jabir M, Rasool KH, Hanumanthaiah M, Sharma N, Pramanik A, Rab SO, Jawad SF, Oghenemaro EF, Mustafa YF. siRNA-based therapy for overcoming drug resistance in human solid tumours; molecular and immunological approaches. Hum Immunol 2025; 86:111221. [PMID: 39700968 DOI: 10.1016/j.humimm.2024.111221] [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: 08/10/2024] [Revised: 11/25/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024]
Abstract
RNA interference (RNAi) is a primordial biological process that protects against external intrusion. SiRNA has the potential to selectively silence disease-related genes in a sequence-specific way, thus offering a promising therapeutic approach. The efficacy of siRNA-based therapies in cancer treatment has gained significant recognition due to multiple studies demonstrating its ability to effectively suppress cancer cells' growth and multiplication. Moreover, siRNA-based medicines have shown considerable promise in enhancing the sensitivity of cancer cells to chemotherapy and other treatment methods by suppressing genes that play a role in the development of drug resistance. Exploring and identifying functional genes linked to cancer cell characteristics and drug resistance is crucial for developing effective siRNAs for cancer treatment and advancing targeted and personalized therapeutics. Targeting and silencing genes in charge of resistance mechanisms, such as those involved in drug efflux, cell survival, or DNA repair, is possible with siRNA therapy in the context of drug resistance, especially cancer. Through inhibiting these genes, siRNA therapy can prevent resistance and restore the efficacy of traditional medications. This review addresses the potential of siRNAs in addressing drug resistance in human tumours, opening up new possibilities in cancer therapy. This review article offers a non-systematic summary of how different siRNA types contribute to cancer cells' treatment resistance. Using pertinent keywords, sources were chosen from reliable databases, including PubMed, Scopus, and Google Scholar. The review covered essential papers in this area and those that mainly addressed the function of siRNA in drug resistance. The articles examined in connection with the title of this review were primarily published from 2020 onward and are based on in vitro studies. Furthermore, this article examines the potential barriers and prospective perspectives of siRNA therapies.
Collapse
Affiliation(s)
| | - Majid Jabir
- Department of Applied Sciences, University of Technology, Iraq
| | | | - Malathi Hanumanthaiah
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Neha Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri - 140307, Mohali, Punjab, India
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University Dehradun, Uttarakhand, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Sabrean Farhan Jawad
- Department of Biochemistry, College of Science, Al-Mustaqbal University, 51001 Babil, Iraq.
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Delta State University, Faculty of Pharmacy, PMB 1 Abraka, Delta State, Nigeria
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| |
Collapse
|
37
|
Zhao X, Qi X, Liu D, Che X, Wu G. A Novel Approach for Bladder Cancer Treatment: Nanoparticles as a Drug Delivery System. Int J Nanomedicine 2024; 19:13461-13483. [PMID: 39713223 PMCID: PMC11662911 DOI: 10.2147/ijn.s498729] [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: 10/01/2024] [Accepted: 12/05/2024] [Indexed: 12/24/2024] Open
Abstract
Bladder cancer represents one of the most prevalent malignant neoplasms of the urinary tract. In the Asian context, it represents the eighth most common cancer in males. In 2022, there were approximately 613,791 individuals diagnosed with bladder cancer worldwide. Despite the availability of efficacious treatments for the two principal forms of bladder cancer, namely non-invasive and invasive bladder cancer, the high incidence of recurrence following treatment and the suboptimal outcomes observed in patients with high-grade and advanced disease represent significant concerns in the management of bladder cancer at this juncture. Nanoparticles have gained attention for their excellent properties, including stable physical properties, a porous structure that can be loaded with a variety of substances, and so on. The in-depth research on nanoparticles has led to their emergence as a new class of nanoparticles for combination therapy, due to their advantageous properties. These include the extension of the drug release window, the enhancement of drug bioavailability, the improvement of drug targeting ability, the reduction of local and systemic toxicity, and the simultaneous delivery of multiple drugs for combination therapy. As a result, nanoparticles have become a novel agent of the drug delivery system. The advent of nanoparticles has provided a new impetus for the development of non-surgical treatments for bladder cancer, including chemotherapy, immunotherapy, gene therapy and phototherapy. The unique properties of nanoparticles have facilitated the combination of diverse non-surgical therapeutic modalities, enhancing their overall efficacy. This review examines the recent advancements in the use of nanoparticles in non-surgical bladder cancer treatments, encompassing aspects such as delivery, therapeutic efficacy, and the associated toxicity of nanoparticles, as well as the challenges encountered in clinical applications.
Collapse
Affiliation(s)
- Xinming Zhao
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People’s Republic of China
| | - Xiaochen Qi
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People’s Republic of China
| | - Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People’s Republic of China
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People’s Republic of China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People’s Republic of China
| |
Collapse
|
38
|
Liu Y, Guo Q, Shi Y, Cui M, Jing F. Research progress of novel anti-tumor drug formulations. Front Oncol 2024; 14:1507958. [PMID: 39737395 PMCID: PMC11683012 DOI: 10.3389/fonc.2024.1507958] [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: 10/08/2024] [Accepted: 12/02/2024] [Indexed: 01/01/2025] Open
Abstract
Cancers have become the second leading cause of death worldwide, following cardiovascular diseases.Traditional anti- cancer strategies, including radiotherapy chemotherapy, surgery, and targeted therapies, have been widely used but are often reassessed due to their significant side effects and relatively low cure rate. Recently, the development of novel formulations for anti-tumor drugs has gained considerable attention, marking a pivotal step forward in cancer treatment advancements. These innovative formulations aim to enhance the therapeutic efficacy of anti-tumor drugs by employing advanced drug formulation technologies and delivery systems. In particular, nano-drug delivery systems (NDDS) have emerged as a promising approach to improve drug targeting, reduce side effects, and overcome drug resistance. This review highlights recent progress in NDDS for anti-tumor drug development and explores the future prospects of these advanced formulations in improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Yan Liu
- Department of Pharmacology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Qie Guo
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - YunYan Shi
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - MengNa Cui
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - FanBo Jing
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
39
|
Wang Y, Li D, Lu Y, Du C, Zou J, Lu Z, Jiang K. A Targeted Octahedral DNA Nanostructure Co-delivers siME3 and Doxorubicin to Enhance Collateral Lethality in ME2-Deficient Pancreatic Cancer. NANO LETTERS 2024; 24:15926-15932. [PMID: 39602246 DOI: 10.1021/acs.nanolett.4c05123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The genetic characteristics of pancreatic cancer (PC) are being revealed, but treatment strategies based on these profiles are developing slowly. About one-third of PC patients harbor SMAD4 mutations, with its homozygous deletions often accompanied by deletions of the malic enzyme 2 (ME2) gene, leading to upregulation of malic enzyme 3 (ME3) to eliminate reactive oxygen species (ROS). We designed an aptamer-modified octahedral DNA nanostructure for targeted co-delivery of siRNA targeting ME3 (siME3) and doxorubicin (DOX). This nanostructure targets the epidermal growth factor receptor (EGFR) on the membrane of PC cells. Upon internalization, siME3 and DOX are released intracellularly. The siME3 effectively inhibited ME3 expression, diminishing the tumor cells' capacity to clear ROS. Moreover, DOX further increases the level of cellular ROS, and the sustained accumulation of ROS ultimately leads to apoptosis of ME2-deficient PC cells. This targeting nanostructure shows potential for enhancing collateral lethality in this PC subgroup.
Collapse
Affiliation(s)
- Yazhou Wang
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Danrui Li
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yichao Lu
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chong Du
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jiajia Zou
- Beijing Intell Nanomedicine, No. 9, Chengwan Street, Haidian District, Beijing 100000, China
| | - Zipeng Lu
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Kuirong Jiang
- Pancreas Centre, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
40
|
Xiong Z, Wang Y, Li Z, Li C, Tu C, Li Z. A review on the crosstalk between non-coding RNAs and the cGAS-STING signaling pathway. Int J Biol Macromol 2024; 283:137748. [PMID: 39566795 DOI: 10.1016/j.ijbiomac.2024.137748] [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: 08/14/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024]
Abstract
In the innate immune system, the cyclic GMP-AMP synthase (cGAS)-interferon gene stimulator (STING) pathway activates the type I interferon (IFN) response and the NF-κB pathway by recognizing double-stranded DNAs, the imbalance of which plays a pivotal role in human diseases, including cancer, autoimmune and inflammatory diseases. Non-coding RNAs (ncRNAs) are a diverse group of transcripts that do not code for proteins but regulate various targets and signaling pathways in physiological and pathological processes. Recently, there has been increasing interest in investigating the interplay between the cGAS-STING pathway and ncRNAs. In this review, we provide a concise overview of the cGAS-STING pathway and ncRNAs. Then, we specifically delve into the regulation of the cGAS-STING pathway by long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs), the three major classes of ncRNAs, and the influence of the cGAS-STING pathway on the expression of ncRNAs. Furthermore, we introduce the therapeutic applications targeting the cGAS-STING pathway and ncRNA therapy, and propose the utilization of drug delivery systems to deliver ncRNAs that influence the cGAS-STING pathway. Overall, this review highlights the emerging understanding of the intricate relationship between the cGAS-STING pathway and ncRNAs, shedding light on their potential as therapeutic targets in various diseases.
Collapse
Affiliation(s)
- Zijian Xiong
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Yu Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhaoqi Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Chenbei Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Shenzhen Research Institute of Central South University, Guangdong 518063, China; Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| | - Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Shenzhen Research Institute of Central South University, Guangdong 518063, China; Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
41
|
Gan Y, Hao Q, Han T, Tong J, Yan Q, Zhong H, Gao B, Li Y, Xuan Z, Li P, Yao L, Xu Y, Jiang YZ, Shao ZM, Deng J, Chen J, Zhou X. Targeting BRIX1 via Engineered Exosomes Induces Nucleolar Stress to Suppress Cancer Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407370. [PMID: 39475053 DOI: 10.1002/advs.202407370] [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: 07/01/2024] [Revised: 10/16/2024] [Indexed: 12/19/2024]
Abstract
Elevated ribosome biogenesis correlates with the rapid growth and progression of cancer. Targeted blockade of ribosome biogenesis induces nucleolar stress, which preferentially leads to the elimination of malignant cells. In this study, it is reported that the nucleolar protein BRIX1 is a critical regulator for the homeostasis between ribosome biogenesis and p53 activation. BRIX1 facilitated the processing of pre-rRNA by supporting the formation of the PeBoW complex. In addition, BRIX1 prevented p53 activation in response to nucleolar stress by impairing the interactions between MDM2 and the ribosomal proteins, RPL5, and RPL11, thereby triggering the resistance of cancer cells to chemotherapy. Conversely, depletion of BRIX1 induced nucleolar stress, which in turn activated p53 through RPL5 and RPL11, consequently inhibiting the growth of tumors. Moreover, engineered exosomes are developed, which are surface-decorated with iRGD, a tumor-homing peptide, and loaded with siRNAs specific to BRIX1, for the treatment of cancer. iRGD-Exo-siBRIX1 significantly suppressed the growth of colorectal cancer and enhanced the efficacy of 5-FU chemotherapy in vivo. Overall, the study uncovers that BRIX1 functions as an oncoprotein to promote rRNA synthesis and dampen p53 activity, and also implies that targeted inhibition of BRIX1 via engineered exosomes can be a potent approach for cancer therapy.
Collapse
Affiliation(s)
- Yu Gan
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China
| | - Qian Hao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China
| | - Tao Han
- Institutes of Health Central Plains, Xinxiang Key laboratory for Molecular Oncology, Xinxiang Medical University, Xinxiang, Henan, 453003, P. R. China
| | - Jing Tong
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China
| | - Qingya Yan
- Institutes of Health Central Plains, Xinxiang Key laboratory for Molecular Oncology, Xinxiang Medical University, Xinxiang, Henan, 453003, P. R. China
| | - Hongguang Zhong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi, 330006, P. R. China
| | - Bo Gao
- Umibio Co. Ltd., Shanghai, 201210, P. R. China
| | - Yanan Li
- Umibio Co. Ltd., Shanghai, 201210, P. R. China
| | | | - Pengfei Li
- Laboratory of Animal Center, Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, 712046, P. R. China
| | - Litong Yao
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, P. R. China
| | - Yingying Xu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, P. R. China
| | - Yi-Zhou Jiang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, P. R. China
| | - Zhi-Ming Shao
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, P. R. China
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, Jiangxi, 330006, P. R. China
| | - Jiaxiang Chen
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, P. R. China
| | - Xiang Zhou
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, P. R. China
- Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, P. R. China
| |
Collapse
|
42
|
Jia X, Wang Y, Qiao Y, Jiang X, Li J. Nanomaterial-based regulation of redox metabolism for enhancing cancer therapy. Chem Soc Rev 2024; 53:11590-11656. [PMID: 39431683 DOI: 10.1039/d4cs00404c] [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: 10/22/2024]
Abstract
Altered redox metabolism is one of the hallmarks of tumor cells, which not only contributes to tumor proliferation, metastasis, and immune evasion, but also has great relevance to therapeutic resistance. Therefore, regulation of redox metabolism of tumor cells has been proposed as an attractive therapeutic strategy to inhibit tumor growth and reverse therapeutic resistance. In this respect, nanomedicines have exhibited significant therapeutic advantages as intensively reported in recent studies. In this review, we would like to summarize the latest advances in nanomaterial-assisted strategies for redox metabolic regulation therapy, with a focus on the regulation of redox metabolism-related metabolite levels, enzyme activity, and signaling pathways. In the end, future expectations and challenges of such emerging strategies have been discussed, hoping to enlighten and promote their further development for meeting the various demands of advanced cancer therapies. It is highly expected that these therapeutic strategies based on redox metabolism regulation will play a more important role in the field of nanomedicine.
Collapse
Affiliation(s)
- Xiaodan Jia
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
| | - Yue Wang
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
| | - Yue Qiao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Xiue Jiang
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Jinghong Li
- Beijing Institute of Life Science and Technology, Beijing 102206, P. R. China
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, P. R. China.
| |
Collapse
|
43
|
Guo F, Ji X, Xiong C, Sun H, Liang Z, Yan-Do R, Gai B, Gao F, Huang L, Li Z, Kuang BY, Shi P. Single-cell encoded gene silencing for high-throughput combinatorial siRNA screening. Nat Commun 2024; 15:9985. [PMID: 39562763 PMCID: PMC11576956 DOI: 10.1038/s41467-024-53419-7] [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: 03/14/2024] [Accepted: 10/09/2024] [Indexed: 11/21/2024] Open
Abstract
The use of combinatorial siRNAs shows great promise for drug discovery, but the identification of safe and effective siRNA combinations remains challenging. Here, we develop a massively multiplexed technology for systematic screening of siRNA-based cocktail therapeutics. We employ composite micro-carriers that are responsive to near infrared light and magnetic field to achieve photoporation-facilitated siRNA transfection to individual cells. Thus, randomized gene silencing by different siRNA formulations can be performed with high-throughput single-cell-based analyses. For screening anti-cancer siRNA cocktails, we test more than 1300 siRNA combinations for knocking down multiple genes related to tumor growth, discovering effective 3-siRNA formulations with an emphasis on the critical role of inhibiting Cyclin D1 and survivin, along with their complementary targets for synergic efficacy. This approach enables orders of magnitude reduction in time and cost associated with largescale siRNA screening, and resolves key insights to siRNA pharmacology that are not permissive to existing methods.
Collapse
Affiliation(s)
- Feng Guo
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Science Park, Hong Kong SAR, 999077, China
| | - Xianglin Ji
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
| | - Chuxiao Xiong
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
| | - Hailiang Sun
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
| | - Zhenghua Liang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, 999077, China
| | - Richard Yan-Do
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Science Park, Hong Kong SAR, 999077, China
| | - Baowen Gai
- Department of Colorectal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Feng Gao
- Department of Colorectal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Linfeng Huang
- Wang-Cai Biochemistry Lab, Division of Natural and Applied Sciences & Global Health Research Center, Duke Kunshan University, Kunshan, Jiangsu, China
| | - Zhongping Li
- Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Becki Yi Kuang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, 999077, China
| | - Peng Shi
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China.
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Science Park, Hong Kong SAR, 999077, China.
- Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China.
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, 518000, China.
| |
Collapse
|
44
|
Xu J, Tang Z. Progress on angiogenic and antiangiogenic agents in the tumor microenvironment. Front Oncol 2024; 14:1491099. [PMID: 39629004 PMCID: PMC11611712 DOI: 10.3389/fonc.2024.1491099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/31/2024] [Indexed: 12/06/2024] Open
Abstract
The development of tumors and their metastasis relies heavily on the process of angiogenesis. When the volume of a tumor expands, the resulting internal hypoxic conditions trigger the body to enhance the production of various angiogenic factors. These include vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), and transforming growth factor-α (TGF-α), all of which work together to stimulate the activation of endothelial cells and catalyze angiogenesis. Antiangiogenic therapy (AAT) aims to normalize tumor blood vessels by inhibiting these angiogenic signals. In this review, we will explore the molecular mechanisms of angiogenesis within the tumor microenvironment, discuss traditional antiangiogenic drugs along with their limitations, examine new antiangiogenic drugs and the advantages of combination therapy, and consider future research directions in the field of antiangiogenic drugs. This comprehensive overview aims to provide insights that may aid in the development of more effective anti-tumor treatments.
Collapse
Affiliation(s)
| | - Zhihua Tang
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing, China
| |
Collapse
|
45
|
Polash SA, Poddar A, Ahmady F, Kannourakis G, Jayachandran A, Shukla R. Impact of Ligand Concentration on the Properties of Nucleic-Acid-Encapsulated MOFs and Inflammation Modulation in Prostate Cancer Cells. ACS APPLIED BIO MATERIALS 2024; 7:7635-7645. [PMID: 39497260 DOI: 10.1021/acsabm.4c01185] [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: 11/19/2024]
Abstract
The zeolitic imidazolate framework (ZIF) is one of the most explored metal-organic-framework-based systems for nucleic acid delivery to cancer cells. Current nucleic acid delivery tools exhibit several drawbacks, such as high manufacturing costs, endosomal entrapment, toxicity, and immunogenicity. However, the biomimetic mineralization of Zn-based ZIFs offers a low-cost and facile encapsulation of nucleic acids at room temperature in aqueous conditions. The efficiency of nucleic acid delivery and its subsequent impact on inflammation in cells are influenced by the physicochemical properties of the material. The imidazole content determines the formation and crystallinity of ZIF, and an optimal ratio ensures the formation of well-defined and highly crystalline structures. In this study, a series of siRNA-encapsulated ZIFs (siRNA@ZIF) were systematically prepared by varying ligand-to-metal (L/M) molar ratios. Our study demonstrates that variations in ligand concentrations influence the crystalline structures, particle size, and shape of siRNA@ZIF particles. At low L/M, two-dimensional siRNA@ZIF particles form with a size of 1 μm. As the L/M ratio increases gradually, the particle size decreases, resulting in three-dimensional particles ∼200 nm in size. We also observed better stability of siRNA@ZIF in water prepared using high L/M values and time-dependent cellular uptake by the cells. Additionally, no significant impact of the biocomposites on inflammation was found, indicating the lack of an unwanted immune response and nonimmunotoxic nature over longer periods (96 h). These findings highlight the necessity of fine-tuning ligand concentrations and synthesis chemistry in designing efficient and optimal ZIF-based systems as versatile delivery platforms for nucleic acids.
Collapse
Affiliation(s)
- Shakil Ahmed Polash
- Ian Potter NanoBiosensing Facility, NanoBiotechnology Research Laboratory, School of Science, RMIT University, Melbourne, VIC 3001, Australia
- Centre for Advanced Materials and Industrial Chemistry, RMIT University, Melbourne, VIC 3001, Australia
| | - Arpita Poddar
- Ian Potter NanoBiosensing Facility, NanoBiotechnology Research Laboratory, School of Science, RMIT University, Melbourne, VIC 3001, Australia
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia
| | - Farah Ahmady
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia
| | | | | | - Ravi Shukla
- Ian Potter NanoBiosensing Facility, NanoBiotechnology Research Laboratory, School of Science, RMIT University, Melbourne, VIC 3001, Australia
- Centre for Advanced Materials and Industrial Chemistry, RMIT University, Melbourne, VIC 3001, Australia
| |
Collapse
|
46
|
Dilmac S, Hamurcu Z, Ozpolat B. Therapeutic Landscape of FOXM1 in Triple-Negative Breast Cancer and Aggressive Solid Cancers. Cancers (Basel) 2024; 16:3823. [PMID: 39594778 PMCID: PMC11593102 DOI: 10.3390/cancers16223823] [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: 10/19/2024] [Revised: 11/11/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the most aggressive forms of breast cancer, lacking common treatment targets such as estrogen (ER), progesterone (PR), and HER2 receptors. This subtype is associated with significant heterogeneity, chemoresistance, early recurrence, metastasis, and poor patient survival. FOXM1 is a cancer-promoting transcription factor that plays a critical role in TNBC and other highly aggressive cancers by driving cell proliferation, invasion, metastasis, and drug resistance. In TNBC, mutations in the TP53 gene-detected in approximately 80% of patients-lead to the overexpression of FOXM1, making it a promising therapeutic target. Beyond TNBC, FOXM1 is implicated in other solid cancers, such as brain (glioblastoma), lung, and pancreatic cancers, and is considered an Achilles' heel of aggressive cancers. Despite its potential as a therapeutic target, there are currently no FDA-approved FOXM1 inhibitors, and none have advanced to clinical trials. This review explores the role of FOXM1 in cancer progression and highlights the current status of efforts to develop effective FOXM1 inhibitors.
Collapse
Affiliation(s)
- Sayra Dilmac
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA;
| | - Zuhal Hamurcu
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri 38030, Turkey;
| | - Bulent Ozpolat
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA;
| |
Collapse
|
47
|
Peng Z, Zhao T, Gao P, Zhang G, Wu X, Tian H, Qu M, Tan X, Zhang Y, Zhao X, Qi X. Tumor-Derived Extracellular Vesicles Enable Tumor Tropism Chemo-Genetherapy for Local Immune Activation in Triple-Negative Breast Cancer. ACS NANO 2024; 18:30943-30956. [PMID: 39474658 PMCID: PMC11562804 DOI: 10.1021/acsnano.3c12967] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 10/11/2024] [Accepted: 10/22/2024] [Indexed: 11/13/2024]
Abstract
Triple-negative breast cancer (TNBC) is highly heterogeneous, lacks accessible therapeutic targets, and features an immunosuppressive tumor microenvironment (TME). Anthracycline-based chemotherapy remains the primary treatment method for TNBC, while the current popular immune checkpoint inhibitors persistently encounter therapeutic resistance. Therefore, there is an urgent need to explore combined therapeutic strategies to remodel the TME and improve the treatment response. Considering the highly specific homing ability of tumor cell-derived vesicles and the key role of the signal transduction and activation of the transcription factor 3 (STAT3) pathway in TNBC, we propose a synergistic therapeutic strategy that integrates gene therapy, chemotherapy, and immunotherapy based on STAT3 short interfering RNA (siSTAT3) and doxorubicin (DOX)-functionalized tumor-derived extracellular vesicles (TEVs) (siSTAT3-DOX@TEV). The in vitro and in vivo results demonstrate that siSTAT3-DOX@TEV target tumor tissues precisely, downregulate STAT3 expression, and synergistically and efficiently induce immunogenic death, thereby reversing the immunosuppressive TME. Moreover, mass cytometry and immunohistochemistry reveal the local immune activation effect of siSTAT3-DOX@TEV, with a significant increase in M1 macrophages, CD4+ T cells, and CD8+ T cells in tumor tissues. These results provide strong hints for the development of TEV-based chemo-gene therapeutic agents for TNBC treatment at the clinical level.
Collapse
Affiliation(s)
- Zaihui Peng
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tingting Zhao
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Pingping Gao
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Guozhi Zhang
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiujuan Wu
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hao Tian
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Man Qu
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xuanni Tan
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yi Zhang
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiang Zhao
- Department
of Oncology, Southwest Hospital, Third Military
Medical University (Army Medical University), Chongqing 400038, China
| | - Xiaowei Qi
- Department
of Breast and Thyroid Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
48
|
Jiao J, Qian Y, Lv Y, Wei W, Long Y, Guo X, Buerliesi A, Ye J, Han H, Li J, Zhu Y, Zhang W. Overcoming limitations and advancing the therapeutic potential of antibody-oligonucleotide conjugates (AOCs): Current status and future perspectives. Pharmacol Res 2024; 209:107469. [PMID: 39433169 DOI: 10.1016/j.phrs.2024.107469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024]
Abstract
As cancer incidence rises due to an aging population, the importance of precision medicine continues to grow. Antibody-drug conjugates (ADCs) exemplify targeted therapies by delivering cytotoxic agents to specific antigens. Building on this concept, researchers have developed antibody-oligonucleotide conjugates (AOCs), which combine antibodies with oligonucleotides to regulate gene expression. This review highlights the mechanism of AOCs, emphasizing their unique ability to selectively target and modulate disease-causing proteins. It also explores the components of AOCs and their application in tumor therapy while addressing key challenges such as manufacturing complexities, endosomal escape, and immune response. The article underscores the significance of AOCs in precision oncology and discusses future directions, highlighting their potential in treating cancers driven by genetic mutations and abnormal protein expression.
Collapse
Affiliation(s)
- Jinlan Jiao
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Yun Qian
- Dermatologic Surgery Department, Institute of Dermatology, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing 210042, China
| | - Yinhua Lv
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Wenqian Wei
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Yongxuan Long
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Xiaoling Guo
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Anya Buerliesi
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Jiahui Ye
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Hao Han
- Department of Ultrasound, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jinbo Li
- State Key Laboratory of Analytical Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China.
| | - Yun Zhu
- Department of Pharmacy, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, China.
| | - Weijie Zhang
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China.
| |
Collapse
|
49
|
Wang Y, Chen B. Nanowire-mediated miRNA delivery. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2532-2534. [PMID: 39190127 DOI: 10.1007/s11427-024-2641-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/07/2024] [Indexed: 08/28/2024]
Affiliation(s)
- Yuan Wang
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Boston Children's Hospital, Harvard Medical School, Boston, 02115, USA.
| | - Bing Chen
- College of Electronic and Optical Engineering and College of Flexible Electronics (Future Technology), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China.
| |
Collapse
|
50
|
Youssef A, Sahgal A, Das S. Radioresistance and brain metastases: a review of the literature and applied perspective. Front Oncol 2024; 14:1477448. [PMID: 39540151 PMCID: PMC11557554 DOI: 10.3389/fonc.2024.1477448] [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: 08/08/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Intracranial metastatic disease is a serious complication of cancer, treated through surgery, radiation, and targeted therapies. The central role of radiation therapy makes understanding the radioresistance of metastases a priori a key interest for prognostication and therapeutic development. Although historically defined clinic-radiographically according to tumour response, developments in new techniques for delivering radiation treatment and understanding of radioprotective mechanisms led to a need to revisit the definition of radioresistance in the modern era. Factors influencing radioresistance include tumour-related factors (hypoxia, cancer stem cells, tumour kinetics, tumour microenvironment, metabolic alterations, tumour heterogeneity DNA damage repair, non-coding RNA, exosomes, methylomes, and autophagy), host-related factors (volume effect & dose-limiting non-cancerous tissue, pathophysiology, and exosomes), technical factors, and probabilistic factors (cell cycle and random gravity of DNA damage). Influences on radioresistance are introduced and discussed in the context of brain metastases.
Collapse
Affiliation(s)
- Andrew Youssef
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Arjun Sahgal
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Hospital, Toronto, ON, Canada
| | - Sunit Das
- Division of Neurosurgery, St. Michael’s Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|