1
|
Jia Z, Zhang Y, Cao L, Wang J, Liang H. Research hotspots and trends of immunotherapy and melanoma: A bibliometric analysis during 2014-2024. Hum Vaccin Immunother 2025; 21:2464379. [PMID: 40012099 PMCID: PMC11869780 DOI: 10.1080/21645515.2025.2464379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/22/2025] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
Over the last decade, the increasing global prevalence of melanoma has sparked growing interest in immunotherapies, which show significant potential against this form of skin cancer. This research aims to offer a framework to guide future studies and inspire new research directions. In this study, we used the Web of Science Core Collection to collect papers on immunotherapy and melanoma published between 2014 and 2024. With Excel and visualization tools like VOSviewer, COOC 13.2, Citespace, and Bibliometrix (R-Tool of R-Studio), we analyzed the data to spot trends and new focuses in the research. Our findings indicate a substantial surge in research activity concerning immunotherapy and melanoma between 2014 and 2024. The USA and China emerged as leading contributors, engaging in extensive and close collaborative efforts with European counterparts. Furthermore, seven of the top 10 research institutions are located in the USA, with the MD Anderson Cancer Center in Texas being the most productive. In addition, the Journal of Cancer Immunotherapy is the journal with the most articles published in the field. Professor Georgina V. Long from the Melanoma Institute at the University of Sydney was one of the most productive scholars. Keyword analysis shows that immune checkpoint inhibitors, tumor microenvironment and targeted therapies are key areas of interest for the research community. This paper uses bibliometric analysis to outline research trends and key points in immunotherapy and melanoma from 2014 to 2024, which helps understand the current research and guides future research directions.
Collapse
Affiliation(s)
- Zixuan Jia
- Department of Urology, People’s Hospital of Longhua, Shenzhen, Guangdong, China
- School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Youao Zhang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, Guangdong, China
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Luyan Cao
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, Scotland
| | - Jieyan Wang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, Guangdong, China
| | - Hui Liang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, Guangdong, China
| |
Collapse
|
2
|
Wang H, Zhao Y, Liu J, Yuan R, Xiang Y. Aptamer-based and highly sensitive electrochemical label-free gliotoxin biosensor via a dual recycling signal amplification cascade strategy. Talanta 2025; 290:127789. [PMID: 40010121 DOI: 10.1016/j.talanta.2025.127789] [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: 12/24/2024] [Revised: 02/10/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
Gliotoxin (GT), a mycotoxin produced by Aspergillus fumigatus, exerts immunosuppressive and pro-apoptotic effects on mammalian cells, posing severe health threat upon human. Ultrasensitively and selectively detecting GT is therefore of great significance. In this work, on the basis of a new GT-specific aptamer, we describe construction of electrochemical label-free biosensor for GT with high sensitivity via exonuclease III (Exo III)-aided dual recycling signal amplification strategy. Target GT analyte combines with the aptamer recognition probe in the aptamer/ssDNA duplex to liberate the ssDNA, which hybridizes with the assistant hairpin to trigger the dual recycling cleavage of the G-quadruplex strand-containing signal hairpin on the electrode with the presence of Exo III. As a result, a substantial number of free G-quadruplex strands are generated. Consequently, aided by K+ ions, these G-quadruplexes bind and confine many hemin molecules on the electrode, which are subjected by electro-reduction for the generation of highly amplified current for label-free GT assay with 3.14 pM detection limit. In addition, such aptamer biosensor is also demonstrated with high selectivity and amenable for detecting GT in diluted human serums, highlighting its promising potentials for the convenient diagnosis of GT-associated diseases.
Collapse
Affiliation(s)
- Hao Wang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Yi Zhao
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Juan Liu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Ruo Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China
| | - Yun Xiang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, PR China.
| |
Collapse
|
3
|
Lai BQ, Wu RJ, Wu CR, Yu HY, Xu J, Yang SB, Chen ZH, Li X, Guo YN, Yang Y, Che MT, Wu TT, Fu GT, Yang YH, Chen Z, Hua N, Liu R, Zheng QJ, Chen YF. DON-Apt19S bioactive scaffold transplantation promotes in situ spinal cord repair in rats with transected spinal cord injury by effectively recruiting endogenous neural stem cells and mesenchymal stem cells. Mater Today Bio 2025; 32:101753. [PMID: 40275960 PMCID: PMC12019207 DOI: 10.1016/j.mtbio.2025.101753] [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: 11/14/2024] [Revised: 03/28/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
The spinal cord's limited regeneration is attributed to the scarcity of endogenous stem cells and a poor post-injury microenvironment in adult mammals. To overcome these challenges, we transplanted a DNA aptamer 19S (Apt19S) sustained-release decellularized optic nerve (DON) scaffold (DON-A) into completely transected spinal cord injury (SCI) site in rats and investigated its effect on endogenous stem cell recruitment and differentiation, which subsequently contributed to in situ SCI repair. It has been demonstrated that Apt19S specifically binds to the membrane receptor alkaline phosphatase highly expressed on neural stem cells (NSCs) and mesenchymal stem cells (MSCs), and our study further proved that Apt19S can simultaneously recruit endogenous NSCs and MSCs to the lesion of SCI. In our study, the DON-A promoted stem cell proliferation in the early stage of the injury, followed by the rapid neurogenesis through NSCs and revascularization via MSCs. Synaptic connections between corticospinal tracts and calcitonin gene-related peptide positive nerve fibers with newborn neurons confirmed the formation of endogenous neuronal relays at the injury site, which improved the rats' motor and sensory functions. This study offers a new strategy for recruiting both NSCs and MSCs to synergistically overcome low spinal cord self-repair ability, holding a high potential for clinical translation.
Collapse
Affiliation(s)
- Bi-Qin Lai
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, China
| | - Rong-Jie Wu
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Chuang-Ran Wu
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hai-Yang Yu
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jing Xu
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Shang-Bin Yang
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Zheng-Hong Chen
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, China
- Rehabilitation Medicine Department, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xing Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi-Nan Guo
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Yue Yang
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Ming-Tian Che
- Cedars Sinai Biobank & Research Pathology Resource, Cedars-Sinai, Los Angeles, CA, 90048, USA
| | - Ting-Ting Wu
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Guang-Tao Fu
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yu-Hui Yang
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhen Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Nan Hua
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Rui Liu
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Qiu-Jian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yuan-Feng Chen
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
4
|
Qiu F, Xie D, Chen H, Wang Z, Huang J, Cao C, Liang Y, Yang X, He DY, Fu X, Lu A, Liang C. Generation of cytotoxic aptamers specifically targeting fibroblast-like synoviocytes by CSCT-SELEX for treatment of rheumatoid arthritis. Ann Rheum Dis 2025; 84:726-745. [PMID: 39237134 DOI: 10.1136/ard-2024-225565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is an autoimmune disease characterised by aggressive fibroblast-like synoviocytes (FLSs). Very few RA patients-derived FLSs (RA-FLSs)-specific surface signatures have been identified, and there is currently no approved targeted therapy for RA-FLSs. This study aimed to screen therapeutic aptamers with cell-targeting and cytotoxic properties against RA-FLSs and to uncover the molecular targets and mechanism of action of the screened aptamers. METHODS We developed a cell-specific and cytotoxic systematic evolution of ligands by exponential enrichment (CSCT-SELEX) method to screen the therapeutic aptamers without prior knowledge of the surface signatures of RA-FLSs. The molecular targets and mechanisms of action of the screened aptamers were determined by pull-down assays and RNA sequencing. The therapeutic efficacy of the screened aptamers was examined in arthritic mouse models. RESULTS We obtained an aptamer SAPT8 that selectively recognised and killed RA-FLSs. The molecular target of SAPT8 was nucleolin (NCL), a shuttling protein overexpressed on the surface and involved in the tumor-like transformation of RA-FLSs. Mechanistically, SAPT8 interacted with the surface NCL and was internalised to achieve lysosomal degradation of NCL, leading to the upregulation of proapoptotic p53 and downregulation of antiapoptotic B-cell lymphoma 2 (Bcl-2) in RA-FLSs. When administrated systemically to arthritic mice, SAPT8 accumulated in the inflamed FLSs of joints. SAPT8 monotherapy or its combination with tumour necrosis factor (TNF)-targeted biologics was shown to relieve arthritis in mouse models. CONCLUSIONS CSCT-SELEX could be a promising strategy for developing cell-targeting and cytotoxic aptamers. SAPT8 aptamer selectively ablates RA-FLSs via modulating NCL-p53/Bcl-2 signalling, representing a potential alternative or complementary therapy for RA.
Collapse
Affiliation(s)
- Fang Qiu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Duoli Xie
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Hongzhen Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zhuqian Wang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jie Huang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Chunhao Cao
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | | | - Xu Yang
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Dong-Yi He
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuekun Fu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China; Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.
| |
Collapse
|
5
|
Yu CH, He X, Acero REP, Han X, Wang Y, Sczepanski JT. Interrogation of mirror-image l-RNA-protein interactions reveals key mechanisms of single-stranded G-rich l-RNA cytotoxicity and a potential mitigation strategy. Chem Sci 2025; 16:7560-7572. [PMID: 40171033 PMCID: PMC11955919 DOI: 10.1039/d5sc00596e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/11/2025] [Indexed: 04/03/2025] Open
Abstract
l-Oligonucleotides (ONs), the synthetic enantiomers of native d-nucleic acids, are being increasingly utilized in the development of diverse biomedical technologies, including molecular imaging tools, diagnostic biosensors, and aptamer-based therapeutics. Nevertheless, our understanding of how l-ONs behave in living systems falls far short of native d-ONs. In particular, despite the potential for an abundant l-ON-protein interactome, the extent to which l-ONs bind to endogenous proteins and the consequences of these interactions are unknown, posing a major hurdle towards engineering functional l-ONs with predictable intracellular behaviours. Towards closing this knowledge gap, we now report the first l-ON-protein interactome, revealing that a wide-range of nuclear proteins have the potential to bind l-RNA. Importantly, by focusing our study on cytotoxic single-stranded G-rich l-RNA sequences, our data reveal key protein interactions that contribute to the cytotoxicity of these sequences. Furthermore, we show that introducing 2'-O-methyl modifications into single-stranded G-rich l-RNA can decrease its cytotoxicity through reducing l-RNA-protein interactions, thereby demonstrating that a well-established strategy for mitigating the cytotoxic effects of antisense ONs may translate across the chiral mirror. Overall, these findings greatly deepen our understanding of the intracellular behavior of l-ONs and provide valuable guidance for the future development of safe and effective l-ON-based biomedical technologies.
Collapse
Affiliation(s)
- Chen-Hsu Yu
- Department of Chemistry, Texas A&M University College Station Texas 77843 USA
| | - Xiaomei He
- Department of Chemistry, University of California Riverside Riverside California 92521-0403 USA
- Department of Biology, East Carolina University Greenville North Carolina 27858 USA
| | | | - Xuan Han
- Department of Chemistry, Texas A&M University College Station Texas 77843 USA
| | - Yinsheng Wang
- Department of Chemistry, University of California Riverside Riverside California 92521-0403 USA
| | - Jonathan T Sczepanski
- Department of Chemistry, Texas A&M University College Station Texas 77843 USA
- Department of Biochemistry and Biophysics, Texas A&M University College Station Texas 77843 USA
| |
Collapse
|
6
|
Li L, Xie S, Zhou J, Ran J. Utilizing aptamers in targeted protein degradation strategies for disease therapy. J Pathol 2025. [PMID: 40207978 DOI: 10.1002/path.6422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/27/2025] [Accepted: 02/26/2025] [Indexed: 04/11/2025]
Abstract
Targeted protein degradation (TPD) has emerged as a promising therapeutic strategy, offering the potential to reduce disease-causing proteins that have traditionally been challenging to target using conventional small molecules. Despite significant advances made with TPD technologies, challenges such as high molecular weight, difficulties in identifying suitable ligands, suboptimal absorption, and metabolic instability remain unresolved. Recently, aptamers - single-stranded DNA or RNA oligonucleotides known for their high specificity and affinity for protein targets - have introduced novel opportunities to expand the scope of TPD, a strategy now referred to as aptamer-based TPD. This approach has demonstrated considerable promise in treating various diseases, such as cancer and ocular disorders. For example, an aptamer-proteolysis-targeting chimera (PROTAC) conjugate (APC) improved tumor targeting and reduced toxicity in a breast cancer model, and a vascular endothelial growth factor-degrading (VED)-lysosome-targeting chimera (LYTAC) molecule effectively inhibited abnormal vascular growth in vascular retinal diseases. These examples highlight the practical relevance and potential in advancing drug discovery efforts. In this review we provide a comprehensive overview of the latest advances in aptamer-based TPD strategies, including proteolysis-targeting and lysosome-targeting chimeras, emphasizing their applications, potential therapeutic benefits, as well as the challenges that must be overcome to fully harness their clinical potential. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Lin Li
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, PR China
| | - Songbo Xie
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, PR China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, PR China
- Department of Genetics and Cell Biology, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, Nankai University, Tianjin, PR China
| | - Jie Ran
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, PR China
| |
Collapse
|
7
|
Cesarini V, Appleton SL, de Franciscis V, Catalucci D. The recent blooming of therapeutic aptamers. Mol Aspects Med 2025; 102:101350. [PMID: 39933246 DOI: 10.1016/j.mam.2025.101350] [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: 09/11/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
In the dynamic landscape of biomedical research, therapeutic RNA aptamers have recently come to the forefront, showing significant potential in diagnostics and therapeutics. This review aims to raise awareness of aptamer technology within the scientific community by exploring the progress made in the therapeutic field, from the lessons learned in research to the future opportunities and impact that these innovative molecules are increasingly having on society to meet current health needs, i.e. targeted and personalized therapies.
Collapse
Affiliation(s)
- Valeriana Cesarini
- National Research Council (CNR), Institute of Genetic and Biomedical Research (IRGB), Milan, Italy
| | - Silvia Lucia Appleton
- National Research Council (CNR), Institute of Genetic and Biomedical Research (IRGB), Milan, Italy
| | - Vittorio de Franciscis
- National Research Council (CNR), Institute of Genetic and Biomedical Research (IRGB), Milan, Italy.
| | - Daniele Catalucci
- National Research Council (CNR), Institute of Genetic and Biomedical Research (IRGB), Milan, Italy; Humanitas Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, (Milan), Italy.
| |
Collapse
|
8
|
He J, Ren X, Zhang Q, Wang S, Li Z, Cai K, Li M, Hu Y, Ran Q, Luo Z. Nanoradiosentizers with X ray-actuatable supramolecular aptamer building units for programmable immunostimulatory T cell engagement. Biomaterials 2025; 315:122924. [PMID: 39489019 DOI: 10.1016/j.biomaterials.2024.122924] [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: 08/05/2024] [Revised: 10/02/2024] [Accepted: 10/27/2024] [Indexed: 11/05/2024]
Abstract
The insufficient activation and impaired effector functions of T cells in the immunosuppressive tumor microenvironment (TME) substantially reduces the immunostimulatory effects of radiotherapy. Herein, a multifunctional nanoradiosensitizer is established by integrating molecularly engineered aptamer precursors into cisplatin-loaded liposomes for enhancing radio-immunotherapy of solid tumors. Exposure to ionizing radiation (IR) following the nanoradiosensitizer treatment would induce pronounced immunogenic death (ICD) of tumor cells through cisplatin-mediated radiosensitization while also trigger the detachment of the aptamer precursors, which further self-assemble into PD-L1/PD-1-bispecific aptamer-based T cell engagers (CA) through the bridging effect of tumor-derived ATP to direct T cell binding onto tumor cells in the post-IR TME in a spatial-temporally programmable manner. The CA-mediated post-IR tumor-T cell engagement could override the immunosuppressive barriers in TME and enhance T cell-mediated recognition and elimination of tumor cells while minimizing systemic toxicities. Overall, this work offers an innovative approach to enhance the radio-immunotherapeutic efficacy in the clinics.
Collapse
Affiliation(s)
- Jinming He
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Xijiao Ren
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Qiqi Zhang
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Shuang Wang
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Zhongjun Li
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 400044, China.
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Qian Ran
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China.
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
9
|
Bege M, Ghanem Kattoub R, Borbás A. The 20th Anniversary of Pegaptanib (MacugenTM), the First Approved Aptamer Medicine: History, Recent Advances and Future Prospects of Aptamers in Therapy. Pharmaceutics 2025; 17:394. [PMID: 40143057 PMCID: PMC11944999 DOI: 10.3390/pharmaceutics17030394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
In addition to classic small-molecule drugs and modern protein-based biologics, an intriguing class of medicines is the therapeutic oligonucleotides. Most approved drugs in this category are antisense oligomers or those acting via RNA interference, both of which use base hybridization. Aptamers, also known as chemical antibodies form a smaller, yet equally interesting group of oligonucleotides that can recognize a wide range of molecular targets. Despite their high potential, only two aptamers have been approved to date, pegaptanib (MacugenTM) and avacincaptad pegol (IzervayTM), both for the treatment of age-related macular degeneration (AMD). Targeting vascular endothelial growth factor (VEGF), which plays an important role in the pathogenesis of many eye diseases, pegaptanib emerged as the first anti-VEGF agent and was used in various indications, further inspiring the development of other anti-VEGF therapies. In this review, we summarize the history of the first approved aptamer medicine, pegaptanib. We describe its chemistry and track its development from the earliest stages to the preclinical phase, clinical trials, and eventual regulatory approval. Additionally, we evaluate its position among other therapeutic agents and provide a comprehensive overview of pegaptanib's efficacy, safety, and cost-effectiveness, comparing these aspects with those of monoclonal antibodies with similar indications, bevacizumab and ranibizumab.
Collapse
Affiliation(s)
- Miklós Bege
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary; (M.B.); (R.G.K.)
| | - Rasha Ghanem Kattoub
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary; (M.B.); (R.G.K.)
- Doctoral School of Pharmaceutical Sciences, Faculty of Pharmacy, University of Debrecen, Nagyerdei krt. 98., 4032 Debrecen, Hungary
| | - Anikó Borbás
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary; (M.B.); (R.G.K.)
| |
Collapse
|
10
|
Yu H, Canoura J, Byrd C, Alkhamis O, Bacon A, Yan A, Sullenger BA, Xiao Y. Improving Aptamer Affinity and Determining Sequence-Activity Relationships via Motif-SELEX. J Am Chem Soc 2025; 147:9472-9486. [PMID: 40053911 DOI: 10.1021/jacs.4c17041] [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: 03/09/2025]
Abstract
The affinity of nucleic acid aptamers isolated in vitro via Systematic Evolution of Ligands by Exponential Enrichment (SELEX) is often limited because the entire potential sequence space cannot be screened. In this study, we introduce Motif-SELEX, a novel method that enables the optimization of existing underperforming aptamers by generating libraries that broadly represent both the sequence and length variations of the parent sequence. This approach enables the isolation of sequences with improved affinity without the biases and limitations of traditional mutagenesis methods like doped SELEX and error-prone PCR. As a demonstration, we applied Motif-SELEX to a DNA-based morphine aptamer and a 2' fluoro- and methoxy-RNA-based apixaban aptamer, discovering new, better-performing sequences with differing random domain lengths from their parents and up to 10-fold improvements in affinity. These new sequences would be inaccessible to traditional post-SELEX methods. Critically, our analysis of Motif-SELEX pools also enabled us to identify sequence and structural elements crucial for target binding and to predict secondary and tertiary structures for a given aptamer family─even when those structures involve noncanonical nucleotide interactions. We believe that Motif-SELEX offers an effective and generalizable solution for optimizing the structure and binding properties of functional nucleic acid molecules for diverse applications.
Collapse
Affiliation(s)
- Haixiang Yu
- Department of Surgery, Duke University Medical Center, 2 Genome Ct., Durham, North Carolina 27710, United States
| | - Juan Canoura
- Department of Chemistry, North Carolina State University, 2620 Yarbrough Dr., Raleigh, North Carolina 27695, United States
| | - Caleb Byrd
- Department of Chemistry, North Carolina State University, 2620 Yarbrough Dr., Raleigh, North Carolina 27695, United States
| | - Obtin Alkhamis
- Department of Chemistry, North Carolina State University, 2620 Yarbrough Dr., Raleigh, North Carolina 27695, United States
| | - Adara Bacon
- Department of Chemistry, North Carolina State University, 2620 Yarbrough Dr., Raleigh, North Carolina 27695, United States
| | - Amy Yan
- Department of Surgery, Duke University Medical Center, 2 Genome Ct., Durham, North Carolina 27710, United States
| | - Bruce A Sullenger
- Department of Surgery, Duke University Medical Center, 2 Genome Ct., Durham, North Carolina 27710, United States
| | - Yi Xiao
- Department of Chemistry, North Carolina State University, 2620 Yarbrough Dr., Raleigh, North Carolina 27695, United States
| |
Collapse
|
11
|
Didarian R, Bargh S, Gülerman A, Özalp VC, Erel Ö, Yildirim-Tirgil N. Selection of DNA Aptamers Against Parathyroid Hormone for Electrochemical Impedimetric Biosensor System Development. Biotechnol Appl Biochem 2025:e2745. [PMID: 40108760 DOI: 10.1002/bab.2745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 03/01/2025] [Indexed: 03/22/2025]
Abstract
This work presents the pioneering development of an aptamer-based electrochemical biosensor for real-time monitoring of parathyroid hormone (PTH) levels, with a focus on intraoperative assessment during parathyroid surgery. It introduces, for the first time, the selection and characterization of aptamers targeting distinct segments of the PTH peptide. The study demonstrates the feasibility and efficacy of the biosensing platform through a precisely designed experimental framework, including SELEX-based aptamer selection, aptamer-peptide interaction analysis, and biosensor fabrication. The SELEX process yields aptamers with notable binding affinities to different fragments of PTH, with the PTH (53-84) aptamer showing particularly sensitive binding to the hormone's C terminus, allowing for precise PTH analysis. Electrochemical characterization reveals significant changes in electrochemical impedance spectroscopy (EIS) signals upon exposure to varying PTH concentrations, highlighting the sensor's sensitivity and selectivity. The increase in charge transfer resistance (Rct) values with rising PTH concentrations underscores the biosensor's capability to detect PTH-induced structural changes, validating its potential for accurate measurement. The biosensor shows remarkable selectivity in the presence of common interferents in serum samples, ensuring precise PTH detection. Stability assessments over a 45-day storage period demonstrate the biosensor's robustness and long-term reliability, affirming its practical suitability. In summary, the developed aptamer-based biosensor represents a promising tool for sensitive and selective PTH detection, with potential applications in biomedical research and clinical diagnostics, particularly for intraoperative PTH analysis during parathyroidectomy. Continued research and optimization efforts hold promise for enhancing its performance and expanding its utility in diverse healthcare settings.
Collapse
Affiliation(s)
- Reza Didarian
- Department of Biomedical Engineering, Ankara Yıldırım Beyazıt University, Ankara, Türkiye
| | - Saharnaz Bargh
- Department of Radiopharmacy, Faculty of Pharmacy, Hacettepe University, Ankara, Türkiye
| | - Almina Gülerman
- Department of Metallurgy and Materials Engineering, Ankara Yıldırım Beyazıt University, Ankara, Türkiye
| | - Veli Cengiz Özalp
- Department of Medical Biology, School of Medicine, Atılım University, Ankara, Türkiye
| | - Özcan Erel
- Department of Biovchemistry, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara, Türkiye
| | - Nimet Yildirim-Tirgil
- Department of Biomedical Engineering, Ankara Yıldırım Beyazıt University, Ankara, Türkiye
- Department of Metallurgy and Materials Engineering, Ankara Yıldırım Beyazıt University, Ankara, Türkiye
| |
Collapse
|
12
|
Huang Z, Li P, Li Y, Duan X, Li M, Jiang D, Li J. SYL3C Aptamer-DNA Tetrahedra Conjugates Enable Near-Infrared Fluorescent Imaging of Colorectal Cancer. Int J Nanomedicine 2025; 20:3595-3606. [PMID: 40125435 PMCID: PMC11930263 DOI: 10.2147/ijn.s510964] [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: 12/07/2024] [Accepted: 03/08/2025] [Indexed: 03/25/2025] Open
Abstract
Purpose SYL3C is an optimized DNA aptamer with high selectivity and affinity for the epithelial cell adhesion molecule (EpCAM), an overexpressed tumor antigen in colorectal cancer (CRC). While its cellular affinity has been validated, in vivo studies are lacking. Methods This study modifies SYL3C with the fluorescent motif Cy7 to evaluate its metabolism and diagnostic potential in EpCAM-positive HT-29 xenograft mice using near-infrared fluorescence (NIRF). We also employ DNA Tetrahedra (DTN) to load the Cy7-DTN-SYL3C probe and assess whether this strategy improves circulation and tumor uptake of SYL3C. Results Cy7-SYL3C is primarily metabolized by the kidneys and enables targeted imaging of HT-29 tumors, outperforming untargeted Cy7-DTN. The DTN coupling strategy prolongs SYL3C metabolism and enhances tumor probe uptake about twice higher than Cy7-SYL3C over 24 hours. Conclusion This study presents preliminary evidence for the SYL3C aptamer's potential in vivo imaging of EpCAM-positive CRC. The DTN conjugation strategy may extend the aptamer's metabolic stability and improve tumor uptake, expanding its applications in CRC diagnosis and treatment.
Collapse
Affiliation(s)
- Zhidie Huang
- Inner Mongolia Medical University, Hohhot, People’s Republic of China
| | - Pinghui Li
- Inner Mongolia Medical University, Hohhot, People’s Republic of China
| | - Yiwen Li
- Inner Mongolia Medical University, Hohhot, People’s Republic of China
| | - Xiaoyan Duan
- Department of Nuclear Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, People’s Republic of China
- Inner Mongolia Key Laboratory of Molecular Imaging, Hohhot, People’s Republic of China
| | - Mengting Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People’s Republic of China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People’s Republic of China
| | - Jianbo Li
- Department of Nuclear Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, People’s Republic of China
- Inner Mongolia Key Laboratory of Molecular Imaging, Hohhot, People’s Republic of China
| |
Collapse
|
13
|
Yang Y, Yang C, Deng K, Xiao Y, Liu X, Du Z. Nucleic Acid Drugs in Radiotherapy. Chembiochem 2025; 26:e202400854. [PMID: 39903093 DOI: 10.1002/cbic.202400854] [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: 10/15/2024] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/06/2025]
Abstract
Radiotherapy remains a cornerstone of cancer treatment, using high-energy radiation to induce DNA damage in tumor cells, leading to cell death. However, its efficacy is often hindered by challenges such as radiation resistance and side effects. As a powerful class of functional molecules, nucleic acid drugs (NADs) present a promising solution to these limitations. Engineered to target key pathways like DNA repair and tumor hypoxia, NADs can enhance radiotherapy sensitivity. NADs can also serve as delivery vehicles for radiotherapy agents such as radionuclides, improving targeting accuracy and minimizing side effects. This review explores the role of NADs in optimizing radiotherapy, highlighting their mechanisms, clinical applications, and synergies with radiotherapy, ultimately offering a promising strategy for improving patient outcomes in cancer therapy.
Collapse
Affiliation(s)
- Yuying Yang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Cai Yang
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Kai Deng
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Yating Xiao
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, Universities and Colleges Admissions Service (UCAS), Hangzhou, 310024, China
| | - Xiangsheng Liu
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Zhen Du
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| |
Collapse
|
14
|
Xia B, Shaheen N, Chen H, Zhao J, Guo P, Zhao Y. RNA aptamer-mediated RNA nanotechnology for potential treatment of cardiopulmonary diseases. Pharmacol Res 2025; 213:107659. [PMID: 39978660 DOI: 10.1016/j.phrs.2025.107659] [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: 09/13/2024] [Revised: 01/14/2025] [Accepted: 02/14/2025] [Indexed: 02/22/2025]
Abstract
Ribonucleic acid (RNA) aptamers are single-stranded RNAs that bind to target proteins or other molecules with high specificity and affinity, modulating biological functions through distinct mechanisms. These aptamers can act n as antagonists to block pathological interactions, agonists to activate signaling pathways, or delivery vehicles for therapeutic cargos such as siRNAs and miRNAs. The advances in RNA nanotechnology further enhances the versatility of RNA aptamers, offering scalable platforms for engineering. In this review, we have summarized recent developments in RNA aptamer-mediated RNA nanotechnology and provide an overview of its potential in treating cardiovascular and respiratory disorders, including atherosclerosis, acute coronary syndromes, heart failure, lung cancer, pulmonary hypertension, asthma, chronic obstructive pulmonary disease (COPD), acute lung injury, viral respiratory infections, and pulmonary fibrosis. By integrating aptamer technologies with innovative delivery systems, RNA aptamers hold the potential to revolutionize the treatment landscape for cardiopulmonary diseases.
Collapse
Affiliation(s)
- Boyu Xia
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Nargis Shaheen
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Huilong Chen
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jing Zhao
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Peixuan Guo
- Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Yutong Zhao
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
15
|
Hsu YW, Ma L, Tang Y, Li M, Zhou C, Geng Y, Zhang C, Wang T, Guo W, Li M, Wang Y. The application of aptamers in the repair of bone, nerve, and vascular tissues. J Mater Chem B 2025; 13:1872-1889. [PMID: 39760465 DOI: 10.1039/d4tb02180k] [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: 01/07/2025]
Abstract
Aptamers represent a distinct category of short nucleotide sequences or peptide molecules characterized by their ability to bind to specific targets with high precision. These molecules are predominantly synthesized through SELEX (Systematic Evolution of Ligands by Exponential Enrichment) technology. Recent findings indicate that aptamers may have significant applications in regenerative medicine, particularly in the domain of tissue repair. In comparison to other bioactive agents, aptamers exhibit superior specificity and affinity, are more readily accessible, and can be chemically modified, thereby presenting a promising avenue for the functionalization of tissue engineering materials in tissue repair applications. This review delineates the properties of aptamers and examines the methodologies and advancements related to aptamer-functionalized hydrogels, nanoparticles, and electrospun materials. It categorizes the four primary functions of aptamers in tissue repair, namely regeneration, delivery systems, anti-inflammatory actions, and pro-coagulation effects. Furthermore, the review explores the utilization of aptamer-functionalized tissue engineering materials in the repair of bone, nerve, and vascular tissues, highlighting the mechanisms by which aptamers facilitate tissue growth and repair through regenerative properties and their role in transporting substances that promote repair. Lastly, the review addresses the future prospects and challenges associated with the application of aptamers in tissue repair, offering novel insights and directions for further research and application in this domain.
Collapse
Affiliation(s)
- Yu-Wei Hsu
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
- Emergency Department, Peking University People's Hospital, Beijing, 100044, China.
| | - Le Ma
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
| | - Ye Tang
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing, 100044, China
| | - Mengen Li
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing, 100044, China
| | - Chengkai Zhou
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
| | - Yan Geng
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
| | - Chenxi Zhang
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
| | - Tianbing Wang
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
| | - Wei Guo
- Emergency Department, Peking University People's Hospital, Beijing, 100044, China.
| | - Ming Li
- Trauma Medicine Center, Peking University People's Hospital, Beijing, 100044, China.
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
| | - Yanhua Wang
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, 100044, China.
- National Center for Trauma Medicine, Beijing, 100044, China
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing, 100044, China
| |
Collapse
|
16
|
Bettinsoli V, Melzi G, Marchese I, Pantaleoni S, Passoni FC, Corsini E. New approach methodologies to assess wanted and unwanted drugs-induced immunostimulation. Curr Res Toxicol 2025; 8:100222. [PMID: 40027547 PMCID: PMC11872130 DOI: 10.1016/j.crtox.2025.100222] [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: 03/14/2024] [Revised: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
This review examines various classes of drugs, focusing on their therapeutic and adverse effects, particularly in relation to immunostimulation. We emphasize the potential of new approach methodologies (NAMs) to study both expected and unexpected immunostimulatory effects. By evaluating the modes of action of different immunostimulatory drugs, we aim to provide insights into effectively assessing unwanted immunostimulatory responses. The review begins by exploring drugs that stimulate the immune system-including immunostimulants, monoclonal antibodies, chemotherapeutics, and nucleic acid-based drugs-to outline NAMs that could be employed to evaluate immunostimulation.
Collapse
Affiliation(s)
- Valeria Bettinsoli
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9 20133 Milan, Italy
- Department of Pharmacy, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Gloria Melzi
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9 20133 Milan, Italy
| | - Irene Marchese
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9 20133 Milan, Italy
| | - Sofia Pantaleoni
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9 20133 Milan, Italy
| | - Francesca Carlotta Passoni
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9 20133 Milan, Italy
| | - Emanuela Corsini
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9 20133 Milan, Italy
| |
Collapse
|
17
|
Zhang Y, Li Y, Fang B, Du Y, Peng P. Framework Nucleic Acids: Innovative Tools for Cellular Sensing and Therapeutics. Chembiochem 2025; 26:e202400810. [PMID: 39653648 DOI: 10.1002/cbic.202400810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/07/2024] [Indexed: 12/18/2024]
Abstract
As emerging biomaterials, framework nucleic acids (FNAs) have recently demonstrated great potential in the biomedical field due to their high programmability, biocompatibility, unique structural diversity, and precise molecular design capabilities. This review focuses on the applications of FNAs in cellular sensing and disease treatment. First, we systematically introduce the applications of FNAs in cellular sensing, including their precise recognition and response to the extracellular tumor microenvironment, cell membrane proteins, and intracellular biomarkers. Subsequently, we review the potential of FNAs in disease treatment, covering their applications and development in drug delivery, regulation of cell behavior, and immunomodulation. We also discuss the limitations and potential role of FNAs in personalized medicine, precision diagnostics, and advanced therapies. The broad application of FNAs is expected to drive significant breakthroughs in future biomedical technological innovations and clinical translation.
Collapse
Affiliation(s)
- Yihan Zhang
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Yuting Li
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Bowen Fang
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Yi Du
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Pai Peng
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| |
Collapse
|
18
|
Liu S, Li X, Gao H, Chen J, Jiang H. Progress in Aptamer Research and Future Applications. ChemistryOpen 2025:e202400463. [PMID: 39901496 DOI: 10.1002/open.202400463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/08/2025] [Indexed: 02/05/2025] Open
Abstract
Aptamers are short, single-stranded DNA, RNA or synthetic XNA molecules that bind to target molecules with high specificity and affinity. These intrinsically structured RNA or DNA oligonucleotides are not only substitutes for antibodies, but also show great potential for applications in diagnostics, specific drug delivery, and treatment of certain diseases. While the process of aptamer identification and its core functional mechanism known as systematic evolution of exponentially enriched ligands (SELEX), SELEX involves a number of single processes, each contributing to the success or failure of aptamer generation. Today, aptamers are widely used to facilitate basic research discoveries and clinical diagnostics. In addition, aptamers play a promising role as clinical diagnostic and therapeutic agents. This review provides recent advances in this rapidly growing field of research, with special emphasis on aptamer generation and screening, small molecule aptamers, the development of aptamer applications, and applications in clinical medicine. And it also discusses the problems that still exist today with aptamers.
Collapse
Affiliation(s)
- Song Liu
- Beijing Anzhen Hospital, Capital Medical University, Experimental Research Center, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| | - Xiaolu Li
- Beijing Anzhen Hospital, Capital Medical University, Experimental Research Center, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| | - Huyang Gao
- Guangxi Medical University, Life Sciences Institute, Nanning, China
| | - Jing Chen
- Beijing Anzhen Hospital, Capital Medical University, Experimental Research Center, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| | - Hongfeng Jiang
- Beijing Anzhen Hospital, Capital Medical University, Experimental Research Center, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, China
| |
Collapse
|
19
|
Ismail E, Liu Y, Wang Y, Yazdanparast Tafti S, Zhang XF, Cheng X. Aptamer-based biotherapeutic conjugate for shear responsive release of Von Willebrand factor A1 domain. NANOSCALE 2025; 17:1246-1259. [PMID: 39412758 DOI: 10.1039/d4nr02715a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Smart polymers that mimic and even surpass the functionality of natural responsive materials have been actively researched. This study explores the design and characterization of a Single-MOlecule-based material REsponsive to Shear (SMORES) for the targeted release of A1, the platelet binding domain of the blood clotting protein von Willebrand factor (VWF). Each SMORES construct employs an aptamer molecule as the flow transducer and a microparticle to sense and amplify the hydrodynamic force. Within the construct, the aptamer, ARC1172, undergoes conformational changes beyond a shear stress threshold, mimicking the shear-responsive behavior of VWF. This conformational alteration modulates the bioavailability of its target, the VWF-A1 domain, ultimately releasing it at elevated shear. Through optical tweezer-based single-molecule force measurement, ARC1172s role as a force transducer was assessed by examining its unfolding under constant pulling force. We also investigated its refolding rate as a function of force under varied relaxation periods. These analyses revealed a narrow range of threshold forces (3-7 pN) governing the transition between folded and unfolded states. We subsequently constructed the SMORES material by conjugating ARC1172 and a microbead, and immobilizing the other end of the aptamer on a substrate. Single-molecule flow experiments on immobilized SMORES constructs revealed a peak A1 domain release within a flow rate range of (40-70 μL min-1). A COMSOL Multiphysics model translated these flow rates to total forces of 3.10 pN-5.63 pN experienced by the aptamers, aligning with single-molecule force microscopy predictions. Evaluation under variable flow conditions showed a peak binding of A1 to the platelet glycoprotein Ib (GPIB) within the same force range, confirming released payload functionality. Building on knowledge of aptamer biomechanics, this study presents a new strategy to create shear-stimulated biomaterials based on single biomolecules.
Collapse
Affiliation(s)
- Esraa Ismail
- Department of Bioengineering, P. C. Rossin College of Engineering and Applied Science, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Yi Liu
- Department of Bioengineering, P. C. Rossin College of Engineering and Applied Science, Lehigh University, Bethlehem, Pennsylvania, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, USA
| | - Yi Wang
- Department of Materials Science and Engineering, P. C. Rossin College of Engineering and Applied Science, Lehigh University, Bethlehem, Pennsylvania, USA.
- Current Address: Analytical Research and Development, Merck & Co., Inc. Greater Philadelphia, Pennsylvania, USA
| | - Sajedehalsadat Yazdanparast Tafti
- Department of Bioengineering, P. C. Rossin College of Engineering and Applied Science, Lehigh University, Bethlehem, Pennsylvania, USA
| | - X Frank Zhang
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, USA
| | - Xuanhong Cheng
- Department of Bioengineering, P. C. Rossin College of Engineering and Applied Science, Lehigh University, Bethlehem, Pennsylvania, USA
- Department of Materials Science and Engineering, P. C. Rossin College of Engineering and Applied Science, Lehigh University, Bethlehem, Pennsylvania, USA.
| |
Collapse
|
20
|
Feng Z, Xie D, Qiu F, Huang J, Wang Z, Liang C. Development of hybrid aptamers-engineered PROTACs for degrading VEGF165 in both tumor- and vascular endothelial cells. Eur J Med Chem 2025; 281:117027. [PMID: 39504794 DOI: 10.1016/j.ejmech.2024.117027] [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: 09/18/2024] [Revised: 10/20/2024] [Accepted: 11/02/2024] [Indexed: 11/08/2024]
Abstract
Tumors and angiogenesis are connected through a complex interplay. VEGF165, generated from both tumor and vascular endothelial cells, serves as a mutual benefit for both cell types. Therapeutic approaches modulating VEGF165 have been proposed as promising antitumor therapies. PROTACs are bifunctional molecules that exploit the intracellular ubiquitin-proteasome system to degrade specific proteins. To date, there are no targeted PROTACs designed to degrade VEGF165 in both tumor and vascular endothelial cells. The aptamer AS1411 is notable for its ability to selectively recognize and enter both tumor and vascular endothelial cells by targeting the cell surface nucleolin (NCL). Moreover, AS1411 has also been repurposed as an intracellular recruiter of E3 ligase MDM2 via leveraging NCL as a molecular bridge. In this study, we conjugated AS1411 with a VEGF165-specific aptamer V7t1, creating hybrid aptamers-engineered PROTACs. The PROTACs demonstrate remarkable selectivity for both tumor and vascular endothelial cells and facilitate the ubiquitination and proteasomal degradation of VEGF165. The PROTACs inhibit the growth of tumor cells and also impede angiogenesis, without causing toxicity to normal tissues. The hybrid aptamers-engineered PROTACs provide an avenue for disrupting the tumor-angiogenesis interplay through modulation of VEGF165 in both tumor and vascular endothelial cells.
Collapse
Affiliation(s)
- Ziting Feng
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Duoli Xie
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Fang Qiu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Jie Huang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Zhuqian Wang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China.
| |
Collapse
|
21
|
Dansereau SJ, Cui H, Dartawan RP, Sheng J. The Plethora of RNA-Protein Interactions Model a Basis for RNA Therapies. Genes (Basel) 2025; 16:48. [PMID: 39858595 PMCID: PMC11765398 DOI: 10.3390/genes16010048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 12/25/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
The notion of RNA-based therapeutics has gained wide attractions in both academic and commercial institutions. RNA is a polymer of nucleic acids that has been proven to be impressively versatile, dating to its hypothesized RNA World origins, evidenced by its enzymatic roles in facilitating DNA replication, mRNA decay, and protein synthesis. This is underscored through the activities of riboswitches, spliceosomes, ribosomes, and telomerases. Given its broad range of interactions within the cell, RNA can be targeted by a therapeutic or modified as a pharmacologic scaffold for diseases such as nucleotide repeat disorders, infectious diseases, and cancer. RNA therapeutic techniques that have been researched include, but are not limited to, CRISPR/Cas gene editing, anti-sense oligonucleotides (ASOs), siRNA, small molecule treatments, and RNA aptamers. The knowledge gleaned from studying RNA-centric mechanisms will inevitably improve the design of RNA-based therapeutics. Building on this understanding, we explore the physiological diversity of RNA functions, examine specific dysfunctions, such as splicing errors and viral interactions, and discuss their therapeutic implications.
Collapse
Affiliation(s)
| | | | | | - Jia Sheng
- Department of Chemistry, The RNA Institute, University at Albany, SUNY, 1400 Washington Ave Extension, Albany, NY 12222, USA; (S.J.D.); (H.C.)
| |
Collapse
|
22
|
Liu ZX, Chen G, Yu ZL. Advances in subpopulation separation and detection of extracellular vesicles: for liquid biopsy and downstream research. Theranostics 2025; 15:1135-1155. [PMID: 39776815 PMCID: PMC11700854 DOI: 10.7150/thno.106459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Extracellular vesicles (EVs) are carriers of a diverse array of bioactive molecules, making them valuable clinical tools for liquid biopsy in disease diagnosis and prognosis evaluation. These molecules play critical roles in various physiological and pathological conditions, and effective separation of EVs is essential to achieve these objectives. Due to the high heterogeneity of EVs, particularly with regard to their cargo molecules, merely isolating the general EV population is inadequate for liquid biopsy and biological function studies. Therefore, separating EV subpopulations becomes crucial. Traditional separation methods, such as differential ultracentrifugation and size exclusion chromatography, along with burgeoning techniques like classical microfluidic chips and covalent chemistry, often prove time-consuming, yield low purity, and have limited ability to address cargo heterogeneity. Thus, precise separation of EV subpopulations is of utmost importance. Additionally, detecting subpopulation-specific cargo is vital for validating the effectiveness of separation methods and supporting clinical biopsy applications. However, reviews that focus specifically on detection methods for EV subpopulations are limited. This paper provides a comprehensive overview of the methods for separating and detecting EV subpopulations with surface marker heterogeneity, comparing the advantages and limitations of each technique. Furthermore, it discusses challenges and future prospects for these methods in the context of liquid biopsy and downstream research. Collectively, this review aims to offer innovative insights into the separation and detection of EV subpopulations, guiding researchers to avoid common pitfalls and refine their investigative approaches.
Collapse
Affiliation(s)
- Zi-Xiu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Gang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Zi-Li Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
23
|
Jain N, Singh A, Bhatia D. DNA-amphiphilic nanostructures: synthesis, characterization and applications. NANOSCALE 2024; 17:18-52. [PMID: 39560070 DOI: 10.1039/d4nr03236e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
DNA's extraordinary potential reaches far beyond its role as a carrier of genetic information. It serves as a remarkably adaptable structural foundation for constructing intricate nanostructures with a diverse range of functionalities. This inherent programmability sets DNA apart from other biomolecules like peptides, proteins, and small molecules. By covalently attaching DNA to synthetic hydrophobic moieties, researchers create DNA amphiphiles capable of interacting with artificial lipid bilayers and cell membranes. These hybrid structures have rapidly gained prominence due to their promising potential in the medical field. This review provides a comprehensive overview of the latest advancements in the synthesis of DNA amphiphiles and their assembly into well-defined nanostructures. It explores the diverse applications of these nanostructures across various medical domains, including targeted drug delivery, innovative immunotherapies, and gene-silencing techniques. Moreover, the review delves into the current challenges and prospects of this rapidly evolving field, highlighting the potential of DNA hybrid materials to revolutionize medical treatments and diagnostics. By addressing the limitations and exploring new avenues of research, scientists aim to unlock the full potential of DNA nanotechnology for the benefit of human health.
Collapse
Affiliation(s)
- Nishkarsh Jain
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Prem Nagar, Patiala, Punjab 147004, India
| | - Ankur Singh
- Department of Biological Sciences & Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Dhiraj Bhatia
- Department of Biological Sciences & Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| |
Collapse
|
24
|
Duan H, Zhang Y, Otis MR, Drolet DW, Geisbrecht BV. The Inhibitory Effects of a Factor B-Binding DNA Aptamer Family Supersede the Gain of Function of Factor B Variants Associated with Atypical Hemolytic Uremic Syndrome. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1691-1702. [PMID: 39431879 PMCID: PMC11573645 DOI: 10.4049/jimmunol.2400420] [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: 07/18/2024] [Accepted: 09/30/2024] [Indexed: 10/22/2024]
Abstract
Aptamers are short, single-stranded oligonucleotides that selectively bind to target biomolecules. Although they generally exhibit good binding specificity, their affinities are often limited because of the relative lack of hydrophobic groups in nucleic acids. Chemically modified nucleotides incorporating hydrophobic structures into uracil have been synthesized to address this obstacle. Modified DNA aptamers containing such nonstandard nucleotides have been developed for >20 different complement proteins. These modified aptamers show increased affinity and enhanced serum stability and have potential value as therapeutic agents. We recently conducted a structure/function study on a family of modified DNA aptamers that bind specifically to complement Factor B (FB). This work revealed that these aptamers selectively inhibit the complement alternative pathway (AP) by preventing the formation of the AP complement component C3 (C3) proconvertase complex, C3bB. Certain patients with atypical hemolytic uremic syndrome express gain-of-function variants of FB that enhance the formation of the proconvertase complex and/or decrease the efficacy of endogenous regulators against the C3 convertases they form. To investigate whether these FB-binding aptamers could override the effects of disease-causing mutations in FB, we examined how they interacted with several FB variants, including D279G, F286L, K323E, and K350N, in various assays of complement function. We found that the inhibitory effect of the FB-binding aptamers superseded the gain-of-function mutations in FB, although the aptamers could not dissociate preformed C3 convertases. These findings suggest that FB-binding aptamers could be further developed as a potential treatment for certain atypical hemolytic uremic syndrome patients or those with other diseases characterized by excessive complement activity.
Collapse
Affiliation(s)
- Huiquan Duan
- Department of Biochemistry & Molecular Biophysics, Kansas State University; Manhattan, KS 66506 U.S.A
| | - Ying Zhang
- Department of Biochemistry & Molecular Biophysics, Kansas State University; Manhattan, KS 66506 U.S.A
| | | | | | - Brian V. Geisbrecht
- Department of Biochemistry & Molecular Biophysics, Kansas State University; Manhattan, KS 66506 U.S.A
| |
Collapse
|
25
|
Leng D, Cao K, Hao Q, Peng Z, Pan G, Liu J, Yu J, Tang J, Li J, Chen H, Chen H, Tang H. CD133-targeted afatinib nanomicelles for enhanced lung cancer theranostics. Nanomedicine (Lond) 2024; 19:2605-2617. [PMID: 39601387 DOI: 10.1080/17435889.2024.2422804] [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: 07/11/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
AIMS To develop a novel nanomicelle system to target and eradicate CD133-expressing lung cancer stem cells (CSCs) while imaging lung cancer. METHODS Averatinib nanomicelles with CD133 aptamers incorporated with gadolinium imaging reagents (M-Afa&Gd-CD133) were synthesized. The anticancer and imaging activities of M-Afa&Gd-CD133 were evaluated both in vitro and in vivo. RESULTS M-Afa&Gd-CD133 efficiently targeted CD133+ lung CSCs and showed significant antitumor efficacy both in vitro and in vivo. Furthermore, M-Afa&Gd-CD33, as a T1 contrast agent, offers superior and sustained visualization of tumors over an extended period. CONCLUSION M-Afa&Gd-CD133 represents a promising strategy for the theranostics of lung cancer.
Collapse
Affiliation(s)
- Dewen Leng
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, China
| | - Kai Cao
- Department of Radiology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Qiang Hao
- Department of Radiology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhu Peng
- College of Life Science, Mudanjiang Medical University, Mudanjiang, China
| | - Gaofeng Pan
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, China
| | - Jing Liu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, China
| | - Jie Yu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, China
| | - Junyi Tang
- Department of Radiology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jing Li
- Department of Radiology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Han Chen
- Department of General Surgery, 905th Hospital of People's Liberation Army Navy, Naval Medical University, Shanghai, China
| | - Huaiwen Chen
- Department of Radiology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
- Sunlipo Biotech Research Center for Nanomedicine, Shanghai, China
| | - Hao Tang
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, China
| |
Collapse
|
26
|
Zhao S, Di Y, Fan H, Xu C, Li H, Wang Y, Wang W, Li C, Wang J. Targeted delivery of extracellular vesicles: the mechanisms, techniques and therapeutic applications. MOLECULAR BIOMEDICINE 2024; 5:60. [PMID: 39567444 PMCID: PMC11579273 DOI: 10.1186/s43556-024-00230-x] [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: 08/14/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024] Open
Abstract
Extracellular vesicles (EVs) are cell-derived vesicles with a phospholipid bilayer measuring 50-150 nm in diameter with demonstrated therapeutic potentials. Limitations such as the natural biodistribution (mainly concentrated in the liver and spleen) and short plasma half-life of EVs present significant challenges to their clinical translation. In recent years, growing research indicated that engineered EVs with enhanced targeting to lesion sites have markedly promoted therapeutic efficacy. However, there is a dearth of systematic knowledge on the recent advances in engineering EVs for targeted delivery. Herein, we provide an overview of the targeting mechanisms, engineering techniques, and clinical translations of natural and engineered EVs in therapeutic applications. Enrichment of EVs at lesion sites may be achieved through the recognition of tissue markers, pathological changes, and the circumvention of mononuclear phagocyte system (MPS). Alternatively, external stimuli, including magnetic fields and ultrasound, may also be employed. EV engineering techniques that fulfill targeting functions includes genetic engineering, membrane fusion, chemical modification and physical modification. A comparative statistical analysis was conducted to elucidate the discrepancies between the diverse techniques on size, morphology, stability, targeting and therapeutic efficacy in vitro and in vivo. Additionally, a summary of the registered clinical trials utilizing EVs from 2010 to 2023 has been provided, with a full discussion on the perspectives. This review provides a comprehensive overview of the mechanisms and techniques associated with targeted delivery of EVs in therapeutic applications to advocate further explorations of engineered EVs and accelerate their clinical applications.
Collapse
Affiliation(s)
- Shuang Zhao
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yunfeng Di
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Huilan Fan
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Chengyan Xu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Haijing Li
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yong Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
- Key Laboratory of Traditional Chinese Medicine Syndrome and Formula, Ministry of Education, Beijing, 100029, China
| | - Wei Wang
- Key Laboratory of Traditional Chinese Medicine Syndrome and Formula, Ministry of Education, Beijing, 100029, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Chun Li
- Key Laboratory of Traditional Chinese Medicine Syndrome and Formula, Ministry of Education, Beijing, 100029, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jingyu Wang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
27
|
Wang Z, Yue S, Chen X, Li J, Zhu P, Chen H, Qiu F, Xie D, Liang Y, Li D, Lu A, Liang C. Design of Murine Double Minute 2 Proteolysis Targeting Chimera Degraders with a Built-In Tumor-Targeting Ability. J Med Chem 2024; 67:18865-18882. [PMID: 39437434 DOI: 10.1021/acs.jmedchem.4c01228] [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: 10/25/2024]
Abstract
Proteolysis targeting chimeras (PROTACs) are heterobifunctional molecules to induce the proteasomal degradation of target proteins. Currently, there are no tumor-targeting PROTACs for modulating oncogenic murine double minute 2 (MDM2). AS1411 is a tumor-targeting aptamer that specifically recognizes nucleolin (NCL) overexpressed on the surface of tumor cells. We recently repurposed AS1411 as an MDM2 recruiter since it could form an NCL-bridged ternary complex with MDM2. In this study, we design a PROTAC molecule AS1411-VH032 via conjugating AS1411 with a recruiter of von Hippel-Lindau (VHL) ligase VH032. AS1411-VH032 facilitates tumor-selective degradation of MDM2, leading to tumor shrinkage with no detectable toxicity. Besides being a molecular target, MDM2 also serves as an E3 ligase harnessed by PROTACs. Thus, we developed an AS1411-based homo-PROTAC homoAS1411, which induces tumor-specific suicide degradation of MDM2 and prevents tumor progression without causing side effects. Both AS1411-VH032 and homoAS1411 are promising MDM2 degraders with built-in tumor-targeting ability, which balances the antitumor efficacy with a favorable safety profile.
Collapse
Affiliation(s)
- Zhuqian Wang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Siran Yue
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Xinxin Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Jin Li
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Peixi Zhu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hongzhen Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Fang Qiu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Duoli Xie
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Yiying Liang
- Shenzhen LingGene Biotech Co., Ltd., Shenzhen 518055, China
| | - Defang Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai 264003, China
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510006, China
- Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China
| |
Collapse
|
28
|
Brylev VA, Ryabukhina EV, Nazarova EV, Samoylenkova NS, Gulyak EL, Sapozhnikova KA, Dzarieva FM, Ustinov AV, Pronin IN, Usachev DY, Kopylov AM, Golovin AV, Pavlova GV, Ryazantsev DY, Korshun VA. Towards Aptamer-Targeted Drug Delivery to Brain Tumors: The Synthesis of Ramified Conjugates of an EGFR-Specific Aptamer with MMAE on a Cathepsin B-Cleavable Linker. Pharmaceutics 2024; 16:1434. [PMID: 39598559 PMCID: PMC11597439 DOI: 10.3390/pharmaceutics16111434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Targeted delivery of chemotherapeutic agents is a well-established approach to cancer therapy. Antibody-drug conjugates (ADCs) typically carry toxic payloads attached to a tumor-associated antigen-targeting IgG antibody via an enzyme-cleavable linker that releases the drug inside the cell. Aptamers are a promising alternative to antibodies in terms of antigen targeting; however, their polynucleotide nature and smaller size result in a completely different PK/PD profile compared to an IgG. This may prove advantageous: owing to their lower molecular weight, aptamer-drug conjugates may achieve better penetration of solid tumors compared to ADCs. Methods: On the way to therapeutic aptamer-drug conjugates, we aimed to develop a versatile and modular approach for the assembly of aptamer-enzymatically cleavable payload conjugates of various drug-aptamer ratios. We chose the epidermal growth factor receptor (EGFR), a transmembrane protein often overexpressed in brain tumors, as the target antigen. We used the 46 mer EGFR-targeting DNA sequence GR-20, monomethylauristatin E (MMAE) on the cathepsin-cleavable ValCit-p-aminobenzylcarbamate linker as the payload, and pentaerythritol-based tetraazide as the branching point for the straightforward synthesis of aptamer-drug conjugates by means of a stepwise Cu-catalyzed azide-alkyne cycloaddition (CuAAC) click reaction. Results: Branched aptamer conjugates of 1:3, 2:2, and 3:1 stoichiometry were synthesized and showed higher cytotoxic activity compared to a 1:1 conjugate, particularly on several glioma cell lines. Conclusions: This approach is convenient and potentially applicable to any aptamer sequence, as well as other payloads and cleavable linkers, thus paving the way for future development of aptamer-drug therapeutics by easily providing a range of branched conjugates for in vitro and in vivo testing.
Collapse
Affiliation(s)
- Vladimir A. Brylev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
| | - Ekaterina V. Ryabukhina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
| | | | - Nadezhda S. Samoylenkova
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
| | - Evgeny L. Gulyak
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
| | - Ksenia A. Sapozhnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
| | - Fatima M. Dzarieva
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
- Institute of Higher Nervous Activity and Neurophysiology, Butlerova 5A, 117485 Moscow, Russia
| | - Alexey V. Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
| | - Igor N. Pronin
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
| | - Dmitry Y. Usachev
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
| | - Alexey M. Kopylov
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1-3, 119991 Moscow, Russia; (A.M.K.)
| | - Andrey V. Golovin
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1-3, 119991 Moscow, Russia; (A.M.K.)
- Department of Microbiology, Virology and Immunology, Sechenov First Moscow State Medical University, Trubetskaya 8, 119991 Moscow, Russia
| | - Galina V. Pavlova
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
- Institute of Higher Nervous Activity and Neurophysiology, Butlerova 5A, 117485 Moscow, Russia
- Department of Medical Genetics, Sechenov First Moscow State Medical University, Trubetskaya 8, 119991 Moscow, Russia
| | - Dmitry Yu. Ryazantsev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
| | - Vladimir A. Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
| |
Collapse
|
29
|
Stangherlin S, Ding Y, Liu J. Dissociation Constant (K d) Measurement for Small-Molecule Binding Aptamers: Homogeneous Assay Methods and Critical Evaluations. SMALL METHODS 2024:e2401572. [PMID: 39511863 DOI: 10.1002/smtd.202401572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/27/2024] [Indexed: 11/15/2024]
Abstract
Since 1990, numerous aptamers have been isolated and discovered for use in various analytical, biomedical, and environmental applications. This trend continues to date. A critical step in the characterization of aptamer binding is to measure its binding affinity toward both target and non-target molecules. Dissociation constant (Kd) is the most commonly used value in characterizing aptamer binding. In this article, homogenous assays are reviewed for aptamers that can bind small-molecule targets. The reviewed methods include label-free methods, such as isothermal titration calorimetry, intrinsic fluorescence of target molecules, DNA staining dyes, and nuclease digestion assays, and labeled methods, such as the strand displacement reaction. Some methods are not recommended, such as those based on the aggregation of gold nanoparticles and the desorption of fluorophore-labeled DNA from nanomaterials. The difference between the measured apparent Kd and the true Kd of aptamer binding is stressed. In addition, avoiding the titration regime and paying attention to the time required to reach equilibrium are discussed. Finally, it is important to include mutated non-binding sequences as controls.
Collapse
Affiliation(s)
- Stefen Stangherlin
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Yuzhe Ding
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Juewen Liu
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| |
Collapse
|
30
|
Chatterjee D, Bhattacharya S, Kumari L, Datta A. Aptamers: ushering in new hopes in targeted glioblastoma therapy. J Drug Target 2024; 32:1005-1028. [PMID: 38923419 DOI: 10.1080/1061186x.2024.2373306] [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: 04/16/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024]
Abstract
Glioblastoma, a formidable brain cancer, has remained a therapeutic challenge due to its aggressive nature and resistance to conventional treatments. Recent data indicate that aptamers, short synthetic DNA or RNA molecules can be used in anti-cancer therapy due to their better tumour penetration, specific binding affinity, longer retention in tumour sites and their ability to cross the blood-brain barrier. With the ability to modify these oligonucleotides through the selection process, and using rational design to modify them, post-SELEX aptamers offer several advantages in glioblastoma treatment, including precise targeting of cancer cells while sparing healthy tissue. This review discusses the pivotal role of aptamers in glioblastoma therapy and diagnosis, emphasising their potential to enhance treatment efficacy and also highlights recent advancements in aptamer-based therapies which can transform the landscape of glioblastoma treatment, offering renewed hope to patients and clinicians alike.
Collapse
Affiliation(s)
- Debarpan Chatterjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| | - Srijan Bhattacharya
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| | - Leena Kumari
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| | - Aparna Datta
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| |
Collapse
|
31
|
Shearer V, Yu CH, Han X, Sczepanski JT. The clinical potential of l-oligonucleotides: challenges and opportunities. Chem Sci 2024; 15:d4sc05157b. [PMID: 39479156 PMCID: PMC11514577 DOI: 10.1039/d4sc05157b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/19/2024] [Indexed: 11/02/2024] Open
Abstract
Chemically modified nucleotides are central to the development of biostable research tools and oligonucleotide therapeutics. In this context, l-oligonucleotides, the synthetic enantiomer of native d-nucleic acids, hold great promise. As enantiomers, l-oligonucleotides share the same physical and chemical properties as their native counterparts, yet their inverted l-(deoxy)ribose sugars afford them orthogonality towards the stereospecific environment of biology. Notably, l-oligonucleotides are highly resistant to degradation by cellular nucleases, providing them with superior biostability. As a result, l-oligonucleotides are being increasingly utilized for the development of diverse biomedical technologies, including molecular imaging tools, diagnostic biosensors, and aptamer-based therapeutics. Herein, we present recent such examples that highlight the clinical potential of l-oligonucleotides. Additionally, we provide our perspective on the remaining challenges and practical considerations currently associated with the use of l-oligonucleotides and explore potential solutions that will lead to the broader adoption of l-oligonucleotides in clinical applications.
Collapse
Affiliation(s)
- Victoria Shearer
- Department of Chemistry, Texas A&M University College Station Texas 77843 USA
| | - Chen-Hsu Yu
- Department of Chemistry, Texas A&M University College Station Texas 77843 USA
| | - Xuan Han
- Department of Chemistry, Texas A&M University College Station Texas 77843 USA
| | | |
Collapse
|
32
|
Leng G, Duan B, Liu J, Li S, Zhao W, Wang S, Hou G, Qu J. The advancements and prospective developments in anti-tumor targeted therapy. Neoplasia 2024; 56:101024. [PMID: 39047659 PMCID: PMC11318541 DOI: 10.1016/j.neo.2024.101024] [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: 02/02/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
Cancer poses a major threat to human health worldwide. The development of anti-tumor materials provides new modalities for cancer diagnosis and treatment. In this review, we comprehensively summarize the research progress and clinical applications of anti-tumor materials. First, we introduce the etiology and pathogenesis of cancer, and the significance and challenges of anti-tumor materials research. Then, we classify anti-tumor materials and discuss their mechanisms of action. After that, we elaborate the research advances and clinical applications of anti-tumor materials, including those targeting tumor cells and therapeutic instruments. Finally, we discuss the future perspectives and challenges in the field of anti-tumor materials. This review aims to provide an overview of the current status of anti-tumor materials research and application, and to offer insights into future directions in this rapidly evolving field, which holds promise for more precise, efficient and customized treatment of cancer.
Collapse
Affiliation(s)
- Guorui Leng
- School of Special Education and Rehabilitation, Binzhou Medical University, Yantai 264003, China
| | - Baorong Duan
- Research Center for Leather and Protein of College of Chemistry & Chemical Engineering, Yantai University, Yantai 264005, China
| | - Junjie Liu
- Department of Physics, Binzhou Medical University, Yantai 264003, China
| | - Song Li
- School of Special Education and Rehabilitation, Binzhou Medical University, Yantai 264003, China
| | - Wenwen Zhao
- School of Special Education and Rehabilitation, Binzhou Medical University, Yantai 264003, China
| | - Shanshan Wang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Guige Hou
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China.
| | - Jiale Qu
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China; Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS); Shandong Provincial Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, China.
| |
Collapse
|
33
|
Lu ZX, Khimani AH, Tong Y. Importance of a reference ladder in analytical testing of CRISPR HDR donor ssDNA templates. Biotechniques 2024; 76:479-484. [PMID: 39632511 DOI: 10.1080/07366205.2024.2429308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024] Open
|
34
|
Mallawarachchi S, Cebecioglu RE, Althumayri M, Beker L, Fernando S, Ceylan Koydemir H. Systematic design and evaluation of aptamers for VEGF and PlGF biomarkers of Preeclampsia. BMC Biotechnol 2024; 24:64. [PMID: 39334133 PMCID: PMC11428563 DOI: 10.1186/s12896-024-00891-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Preeclampsia is a potentially life-threatening condition for both mother and baby, characterized by hypertension and potential organ damage. Early diagnosis is crucial to mitigate its adverse health effects. Traditional diagnostic methods, which focus on late-manifesting symptoms like hypertension and proteinuria, underscore the need for molecular diagnostic approaches for timely detection. This study successfully designs and evaluates novel aptamers with high specificity and affinity for Vascular Endothelial Growth Factor (VEGF) and Placental Growth Factor (PlGF), biomarkers closely associated with preeclampsia. Using molecular docking, molecular dynamics simulations, and BioLayer Interferometry (BLI), we identified aptamers that demonstrated strong binding affinities, comparable or superior to traditional antibodies. Our findings suggest that these aptamers have the potential to be integrated into cost-effective, point-of-care diagnostic tools, significantly improving early detection and intervention strategies for preeclampsia. The robust performance of these aptamers marks a pivotal step toward the development of more reliable and accessible diagnostic solutions, with implications for better maternal and fetal health outcomes.
Collapse
Affiliation(s)
- Samavath Mallawarachchi
- Department of Biological and Agricultural Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Rümeysa E Cebecioglu
- Department of Biomedical Sciences and Engineering, Koç University, Istanbul, 34450, Turkey
- Medical Laboratory Techniques, Health Services of Vocational School, Kent University, Istanbul, 34333, Turkey
| | - Majed Althumayri
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
- Center for Remote Health Technologies and Systems, Texas A&M Engineering Experiment Station, College Station, TX, 77843, USA
| | - Levent Beker
- Department of Mechanical Engineering, Koç University, Istanbul, 34450, Turkey
| | - Sandun Fernando
- Department of Biological and Agricultural Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Hatice Ceylan Koydemir
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA.
- Center for Remote Health Technologies and Systems, Texas A&M Engineering Experiment Station, College Station, TX, 77843, USA.
| |
Collapse
|
35
|
Erkal-Aytemur A, Mülazımoğlu İE, Üstündağ Z, Caglayan MO. A novel aptasensor platform for the detection of carcinoembryonic antigen using quartz crystal microbalance. Talanta 2024; 277:126376. [PMID: 38852341 DOI: 10.1016/j.talanta.2024.126376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/13/2024] [Accepted: 06/05/2024] [Indexed: 06/11/2024]
Abstract
In this study, a quartz crystal microbalance (QCM) aptasensor for carcinoembryonic antigen (CEA), a well-known biomarker for various cancer types, was reported, utilizing two different aptamers. To achieve this, a nanofilm of 4-mercaptophenyl was electrochemically attached to gold-coated QCM crystal surfaces via the reduction of 4-mercaptobenzenediazonium salt (4 MB-DAT) using cyclic voltammetry. Subsequently, gold nanoparticles (AuNP) were affixed to this structure, and then aptamers (antiCEA1 and antiCEA2) modified with SH-functional ends bound to AuNPs completed the modification. The analytical performance of the CEA sensor was evaluated through simultaneous QCM measurements employing CEA solutions ranging from 0.1 ng/mL to 25 ng/mL. The detection limit (LOD) for CEA was determined to be 102 pg/mL for antiCEA1 and 108 pg/mL for antiCEA2 aptamers. Interday and intraday precision and accuracy tests yielded maximum results of 4.3 and + 3.8, respectively, for both aptasensors, as measured by relative standard deviation (RSD%) and relative error (RE%). The kinetic data of the aptasensors resulted in affinity values (KD) of 0.43 ± 0.14 nM for antiCEA1 and 0.75 ± 0.42 nM for antiCEA2. These values were lower than the reported values of 3.9 nM and 37.8 nM for both aptamers, respectively. The selectivity of the aptasensor was evaluated by measuring the signal changes caused by alpha-fetoprotein (AFP), cancer antigen (CA-125), and vascular endothelial growth factor (VEGF-165) individually and together at a concentration of 500 ng/mL, resulting in a maximum 4.1 % change, which was comparable to precision and accuracy values reported in the literature. After confirming the selectivity of the aptamers, recovery experiments were conducted using spiked commercial serum samples to simulate real samples, and the lowest recovery value obtained was 95.4 %. It was determined that two different aptasensors could be successfully used for the QCM-based detection of CEA in this study.
Collapse
Affiliation(s)
- Aslı Erkal-Aytemur
- Alanya Alaaddin Keykubat University, R.K. Faculty of Engineering, Fundamental Science, Antalya, Turkey
| | | | - Zafer Üstündağ
- Kütahya Dumlupınar University, Faculty of Arts and Science, Department of Chemistry, Kütahya, Turkey
| | - Mustafa Oguzhan Caglayan
- Bilecik Seyh Edebali University, Faculty of Engineering, Department of Bioengineering, Bilecik, Turkey.
| |
Collapse
|
36
|
Uinarni H, Oghenemaro EF, Menon SV, Hjazi A, Ibrahim FM, Kaur M, Zafarjonovna AZ, Deorari M, Jabir MS, Zwamel AH. Breaking Barriers: Nucleic Acid Aptamers in Gastrointestinal (GI) Cancers Therapy. Cell Biochem Biophys 2024; 82:1763-1776. [PMID: 38916791 DOI: 10.1007/s12013-024-01367-w] [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] [Accepted: 06/13/2024] [Indexed: 06/26/2024]
Abstract
Conventional cancer therapies can have significant adverse effects as they are not targeted to cancer cells and may damage healthy cells. Single-stranded oligonucleotides assembled in a particular architecture, known as aptamers, enable them to attach selectively to target areas. Usually, they are created by Systematic Evolution of Ligand by Exponential enrichment (SELEX), and they go through a rigorous pharmacological revision process to change their therapeutic half-life, affinity, and specificity. They could thus offer a viable substitute for antibodies in the targeted cancer treatment market. Although aptamers can be a better choice in some situations, antibodies are still appropriate for many other uses. The technique of delivering aptamers is simple and reasonable, and the time needed to manufacture them is relatively brief. Aptamers do not require animals or an immune response to be produced, in contrast to antibodies. When used as a medication, aptamers can directly suppress tumor cells. As an alternative, they can be included in systems for targeted drug delivery that administer medications specifically to tumor cells while reducing toxicity to healthy cells. The most recent and cutting-edge methods for treating gastrointestinal (GI) tract cancer with aptamers will be covered in this review, with a focus on targeted therapy as a means of conquering resistance to traditional medicines.
Collapse
Affiliation(s)
- Herlina Uinarni
- Department of Anatomy, School of Medicine and Health Sciences Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia.
- Radiology department of Pantai Indah Kapuk Hospital Jakarta, Jakarta, Indonesia.
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria
| | - Soumya V Menon
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Fatma Magdi Ibrahim
- Assisstant professor, Community Health Nursing, RAK Medical and Health Sciences University, Ras Al Khaimah, UAE
- Lecturer, geriatric nursing, Mansoura University, Mansoura, Egypt
| | - Mandeep Kaur
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | | | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Majid S Jabir
- Department of applied sciences, University of technology, Baghdad, Iraq
| | - Ahmed Hussein Zwamel
- Medical laboratory technique college, the Islamic University, Najaf, Iraq
- Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
37
|
Sutariya V, Bhatt P, Saini A, Miller A, Badole SL, Tur J, Gittinger M, Kim JW, Manickam R, Tipparaju SM. Development and testing of nanoparticles delivery for P7C3 small molecule using injury models. Mol Cell Biochem 2024; 479:2429-2445. [PMID: 37787834 DOI: 10.1007/s11010-023-04865-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 09/21/2023] [Indexed: 10/04/2023]
Abstract
The use of nanoparticles (NPs) has emerged as a potential tool for safe and effective drug delivery. In the present study, we developed small molecule P7C3-based NPs and tested its efficacy and toxicity along with the tissue specific aptamer-modified P7C3 NPs. The P7C3 NPs were prepared using poly (D, L-lactic-co-glycolic acid) carboxylic acid (PLGA-COOH) polymer, were conjugated with skeletal muscle-specific RNA aptamer (A01B P7C3 NPs) and characterized for its cytotoxicity, cellular uptake, and wound healing in vitro. The A01B P7C3 NPs demonstrated an encapsulation efficiency of 30.2 ± 2.6%, with the particle size 255.9 ± 4.3 nm, polydispersity index of 0.335 ± 0.05 and zeta potential of + 10.4 ± 1.8mV. The FTIR spectrum of P7C3 NPs displayed complete encapsulation of the drug in the NPs. The P7C3 NPs and A01B P7C3 NPs displayed sustained drug release in vitro for up to 6 days and qPCR analysis confirmed A01B aptamer binding to P7C3 NPs. The C2C12 cells viability assay displayed no cytotoxic effects of all 3 formulations at 48 and 72 h. In addition, the cellular uptake of A01B P7C3 NPs in C2C12 myoblasts demonstrated higher uptake. In vitro assay mimicking wound healing showed improved wound closure with P7C3 NPs. In addition, P7C3 NPs significantly decreased TNF-α induced NF-κB activity in the C2C12/NF-κB reporter cells after 24-hour treatment. The P7C3 NPs showed 3-4-fold higher efficacy compared to P7C3 solutions in both wound-closure and inflammation assays in C2C12 cells. Furthermore, the P7C3 NPs showed 3-4-fold higher efficacy in reducing the infarct size and protected mouse hearts from ex vivo ischemia-reperfusion injury. Overall, this study demonstrates the safe and effective delivery of P7C3 NPs.
Collapse
Affiliation(s)
- Vijaykumar Sutariya
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Priyanka Bhatt
- Department of Pharmaceutical Sciences, Wegmans School of Pharmacy, St. John Fisher University, Rochester, NY, 14618, USA
| | - Aren Saini
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Abraian Miller
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Sachin L Badole
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Jared Tur
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Mackenzie Gittinger
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Joung Woul Kim
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Ravikumar Manickam
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Srinivas M Tipparaju
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA.
| |
Collapse
|
38
|
Rezaee MA, Shobeiri SS, Moghadam M, Mashayekhi K, Sankian M. In vitro identification of single-stranded DNA aptamers targeting human IL-23 using the protein-SELEX strategy. J Pharm Biomed Anal 2024; 247:116245. [PMID: 38810331 DOI: 10.1016/j.jpba.2024.116245] [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: 04/09/2024] [Revised: 05/15/2024] [Accepted: 05/19/2024] [Indexed: 05/31/2024]
Abstract
Interleukin (IL)-23 inhibitor monoclonal antibodies shown significant efficacy in treating autoimmune diseases. DNA or RNA aptamers exhibit comparable specificity to antibodies, are cost-effective, non-immunogenic, and do not have batch to batch variation. This study aimed to characterize a single-stranded DNA (ssDNA) aptamer targeting human IL-23. The alpha subunit of IL-23 (P19) and intact IL-23 were cloned, expressed, and the proteins finally were purified through Ni2+-iminodiacetic acid affinity chromatography. The selection and characterization of ssDNA aptamer against P19 were conducted using the protein-systematic evolution of ligands by exponential enrichment (SELEX). Dot blot assay was carried out to monitor binding of the aptamer output of SELEX rounds, to P19 protein. The dissociation constant (Kd) of aptamers with positive results in dot blot assay, determined based on their binding to IL-23 using an ELISA method. Recombinant P19 and IL-23 proteins were 26 and 72 kDa, respectively, observed on SDS-PAGE .12 %. The aptamers output from 7, 8, 9, 10, 11, and 12 rounds of the SELEX was monitored by dot blot assay, revealing that the aptamer from the round 8 has stronger luminescent signal and was selected for TA-cloning. After analyzing the biotinylated aptamers from clones, positive clones in dot blot assay and ELISA were sequenced. Finally, the Kd calculation revealed three aptamers with high affinity, named A23P3, A23P6, and A23P15 with Kd values of 1.37, 2.139, and 2.88 nM, respectively. Results of this study introduced three specific anti-IL-23 ssDNA aptamers with high affinity, which could be utilized for therapeutic and diagnostic purposes.
Collapse
Affiliation(s)
- Mohammad Ali Rezaee
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Laboratory Sciences, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Saeideh Sadat Shobeiri
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Malihe Moghadam
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kazem Mashayekhi
- Department of Immunology, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mojtaba Sankian
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
39
|
Barros MM, Castro J, Araújo D, Oliveira R, Campos AM, Silva S, Outor-Monteiro D, Almeida C. Application of DNA aptamers to block enterotoxigenic Escherichia coli toxicity in a Galleria mellonella larval model. Front Chem 2024; 12:1425903. [PMID: 39268007 PMCID: PMC11390681 DOI: 10.3389/fchem.2024.1425903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is the major bacterial cause of diarrheal diseases in pigs, particularly at young ages, resulting in significant costs to swine farming. The pathogenicity of ETEC is largely dependent on the presence of fimbriae and the ability to produce toxins. Fimbriae are responsible for their initial adhesion to the intestinal epithelial cells, leading to the onset of infection. In particular, the F4 type (K88) fimbriae are often attributed to neonatal infections and have also been associated with post-weaning diarrheal infections. This disease is traditionally prevented or treated with antibiotics, but their use is being severely restricted due to the emergence of resistant bacteria and their impact on human health. Emerging approaches such as aptamers that target the F4-type fimbriae and block the initial ETEC adhesion are a promising alternative. The aim of this study is to assess the effectiveness of two aptamers, Apt31 and Apt37, in controlling ETEC infection in the G. mellonella in vivo model. Initially, the dissociation constant (KD) of each aptamer against ETEC was established using real-time quantitative PCR methodology. Subsequently, different concentrations of the aptamers were injected into Galleria mellonella to study their toxicity. Afterwards, the anti-ETEC potential of Apt31 and Apt37 was assessed in the larvae model. The determined KD was 81.79 nM (95% CI: 31.21-199.4 nM) and 50.71 nM (95% CI: 26.52-96.15 nM) for the Apt31 and Apt37, respectively, showing no statistical difference. No toxicity was observed in G. mellonella following injection with both aptamers at any concentration. However, the administration of Apt31 together with ETEC-F4+ in G. mellonella resulted in a significant improvement of approximately 30% in both larvae survival and health index compared to ETEC-F4+ alone. These findings suggest that aptamers have promising inhibitory effect against ETEC infections and pave the way for additional in vivo studies.
Collapse
Affiliation(s)
- Maria Margarida Barros
- National Institute for Agrarian and Veterinariay Research (INIAV), Vairão, Portugal
- Veterinary and Animal Research Centre (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Joana Castro
- National Institute for Agrarian and Veterinariay Research (INIAV), Vairão, Portugal
- Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Daniela Araújo
- National Institute for Agrarian and Veterinariay Research (INIAV), Vairão, Portugal
- Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS-Associate Laboratory, Braga, Portugal
| | - Ricardo Oliveira
- National Institute for Agrarian and Veterinariay Research (INIAV), Vairão, Portugal
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Ana Maria Campos
- National Institute for Agrarian and Veterinariay Research (INIAV), Vairão, Portugal
| | - Sónia Silva
- National Institute for Agrarian and Veterinariay Research (INIAV), Vairão, Portugal
- Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS-Associate Laboratory, Braga, Portugal
| | - Divanildo Outor-Monteiro
- Veterinary and Animal Research Centre (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Carina Almeida
- National Institute for Agrarian and Veterinariay Research (INIAV), Vairão, Portugal
- Centre of Biological Engineering, University of Minho, Braga, Portugal
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| |
Collapse
|
40
|
Kosel J, Ropret P. The potential of aptamers for the analysis of ceramic bound proteins found within pottery. Sci Rep 2024; 14:19947. [PMID: 39198509 PMCID: PMC11358422 DOI: 10.1038/s41598-024-70048-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/12/2024] [Indexed: 09/01/2024] Open
Abstract
Archaeological pottery are the most numerous objects found during excavations and reflect the culinary practices of the past. However, their functionality for cooking/storing specific foods or drinks cannot be deduced solely from comparing their shapes and sizes. Analysis of protein residues bound to ceramics can reveal the protein/animal type through their amino acid sequence, thus enabling direct identification of food types. Therefore, the aim of our experimental study was to test sixteen aptamers for the analysis of proteinaceous organic residues found within the porous structure of pottery. Traditionally prepared archaeological ceramic replicas were cooked for 5 days in various food/protein suspensions, were UV aged, buried for a year, excavated, and extensively cleaned. Their shards were analysed using immunofluorescence microscopy with aptamers. Results show that eight aptamers (Clone1 and Kirby for egg residuals; seqU5 and BLG14 for milk residuals; HA for blood residuals; Gli4 for gluten residuals; Par1 for fish residuals; and D1 for collagen residuals) produced a successful/specific immunofluorescence microscopy result when they were hybridised to shards containing target protein residuals. Interestingly, on whole egg control samples, when the egg lysozyme-targeting aptamer Kirby was used, fluorescence intensity was 3.1 times greater compared to that observed with anti-ovalbumin antibodies.
Collapse
Affiliation(s)
- Janez Kosel
- Institute for the Protection of Cultural Heritage of Slovenia, Conservation Centre, Research Institute, Ljubljana, Slovenia.
| | - Polonca Ropret
- Institute for the Protection of Cultural Heritage of Slovenia, Conservation Centre, Research Institute, Ljubljana, Slovenia
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
41
|
Didarian R, Ozbek HK, Ozalp VC, Erel O, Yildirim-Tirgil N. Enhanced SELEX Platforms for Aptamer Selection with Improved Characteristics: A Review. Mol Biotechnol 2024:10.1007/s12033-024-01256-w. [PMID: 39152308 DOI: 10.1007/s12033-024-01256-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 06/26/2024] [Indexed: 08/19/2024]
Abstract
This review delves into the advancements in molecular recognition through enhanced SELEX (Systematic Evolution of Ligands by Exponential Enrichment) platforms and post-aptamer modifications. Aptamers, with their superior specificity and affinity compared to antibodies, are central to this discussion. Despite the advantages of the SELEX process-encompassing stages like ssDNA library preparation, incubation, separation, and PCR amplification-it faces challenges, such as nuclease susceptibility. To address these issues and propel aptamer technology forward, we examine next-generation SELEX platforms, including microfluidic-based SELEX, capillary electrophoresis SELEX, cell-based aptamer selection, counter-SELEX, in vivo SELEX, and high-throughput sequencing SELEX, highlighting their respective merits and innovations. Furthermore, this article underscores the significance of post-aptamer modifications, particularly chemical strategies that enhance aptamer stability, reduce renal filtration, and expand their target range, thereby broadening their utility in diagnostics, therapeutics, and nanotechnology. By synthesizing these advanced SELEX platforms and modifications, this review illuminates the dynamic progress in aptamer research and outlines the ongoing efforts to surmount existing challenges and enhance their clinical applicability, charting a path for future breakthroughs in this evolving field.
Collapse
Affiliation(s)
- Reza Didarian
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Ankara Yıldırım Beyazıt University, Ayvalı Mh. Takdir Cad.150 Sk. No:5, Etlik-Keçiören, Ankara, 06010, Türkiye
| | - Hatice K Ozbek
- Metallurgical and Materials Engineering Department, Faculty of Engineering and Natural Sciences, Ankara Yıldırım Beyazıt University, Ankara, 06010, Türkiye
| | - Veli C Ozalp
- Department of Medical Biology, School of Medicine, Atilim University, Ankara, 06830, Türkiye
| | - Ozcan Erel
- Department of Biochemistry, Faculty of Medicine, Yıldırım Beyazıt University, Ankara, 06010, Türkiye
| | - Nimet Yildirim-Tirgil
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Ankara Yıldırım Beyazıt University, Ayvalı Mh. Takdir Cad.150 Sk. No:5, Etlik-Keçiören, Ankara, 06010, Türkiye.
| |
Collapse
|
42
|
Yang T, Peng J, Zhang Z, Chen Y, Liu Z, Jiang L, Jin L, Han M, Su B, Li Y. Emerging therapeutic strategies targeting extracellular histones for critical and inflammatory diseases: an updated narrative review. Front Immunol 2024; 15:1438984. [PMID: 39206200 PMCID: PMC11349558 DOI: 10.3389/fimmu.2024.1438984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Extracellular histones are crucial damage-associated molecular patterns involved in the development and progression of multiple critical and inflammatory diseases, such as sepsis, pancreatitis, trauma, acute liver failure, acute respiratory distress syndrome, vasculitis and arthritis. During the past decade, the physiopathologic mechanisms of histone-mediated hyperinflammation, endothelial dysfunction, coagulation activation, neuroimmune injury and organ dysfunction in diseases have been systematically elucidated. Emerging preclinical evidence further shows that anti-histone strategies with either their neutralizers (heparin, heparinoids, nature plasma proteins, small anion molecules and nanomedicines, etc.) or extracorporeal blood purification techniques can significantly alleviate histone-induced deleterious effects, and thus improve the outcomes of histone-related critical and inflammatory animal models. However, a systemic evaluation of the efficacy and safety of these histone-targeting therapeutic strategies is currently lacking. In this review, we first update our latest understanding of the underlying molecular mechanisms of histone-induced hyperinflammation, endothelial dysfunction, coagulopathy, and organ dysfunction. Then, we summarize the latest advances in histone-targeting therapy strategies with heparin, anti-histone antibodies, histone-binding proteins or molecules, and histone-affinity hemoadsorption in pre-clinical studies. Finally, challenges and future perspectives for improving the clinical translation of histone-targeting therapeutic strategies are also discussed to promote better management of patients with histone-related diseases.
Collapse
Affiliation(s)
- Tinghang Yang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Jing Peng
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Zhuyun Zhang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Yu Chen
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, China
| | - Zhihui Liu
- Department of Rheumatology and Immunology, West China Hospital of Sichuan University, Chengdu, China
| | - Luojia Jiang
- Jiujiang City Key Laboratory of Cell Therapy, Department of Nephrology, Jiujiang No. 1 People’s Hospital, Jiujiang, China
| | - Lunqiang Jin
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Mei Han
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Baihai Su
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
- Med+ Biomaterial Institute of West China Hospital/West China School of Medicine, Sichuan University, Chengdu, China
- Med-X Center for Materials, Sichuan University, Chengdu, China
| | - Yupei Li
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Jannati S, Patnaik R, Banerjee Y. Beyond Anticoagulation: A Comprehensive Review of Non-Vitamin K Oral Anticoagulants (NOACs) in Inflammation and Protease-Activated Receptor Signaling. Int J Mol Sci 2024; 25:8727. [PMID: 39201414 PMCID: PMC11355043 DOI: 10.3390/ijms25168727] [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: 06/16/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 09/02/2024] Open
Abstract
Non-vitamin K oral anticoagulants (NOACs) have revolutionized anticoagulant therapy, offering improved safety and efficacy over traditional agents like warfarin. This review comprehensively examines the dual roles of NOACs-apixaban, rivaroxaban, edoxaban, and dabigatran-not only as anticoagulants, but also as modulators of inflammation via protease-activated receptor (PAR) signaling. We highlight the unique pharmacotherapeutic properties of each NOAC, supported by key clinical trials demonstrating their effectiveness in preventing thromboembolic events. Beyond their established anticoagulant roles, emerging research suggests that NOACs influence inflammation through PAR signaling pathways, implicating factors such as factor Xa (FXa) and thrombin in the modulation of inflammatory responses. This review synthesizes current evidence on the anti-inflammatory potential of NOACs, exploring their impact on inflammatory markers and conditions like atherosclerosis and diabetes. By delineating the mechanisms by which NOACs mediate anti-inflammatory effects, this work aims to expand their therapeutic utility, offering new perspectives for managing inflammatory diseases. Our findings underscore the broader clinical implications of NOACs, advocating for their consideration in therapeutic strategies aimed at addressing inflammation-related pathologies. This comprehensive synthesis not only enhances understanding of NOACs' multifaceted roles, but also paves the way for future research and clinical applications in inflammation and cardiovascular health.
Collapse
Affiliation(s)
- Shirin Jannati
- Yajnavalkaa Banerrji Research Group, College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (S.J.); (R.P.)
| | - Rajashree Patnaik
- Yajnavalkaa Banerrji Research Group, College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (S.J.); (R.P.)
| | - Yajnavalka Banerjee
- Yajnavalkaa Banerrji Research Group, College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai Health, Dubai P.O. Box 505055, United Arab Emirates; (S.J.); (R.P.)
- Centre for Medical Education, University of Dundee, Dundee DD1 4HN, UK
| |
Collapse
|
44
|
Wong K, Liu Y, Wong M, Liu J. Cornea-SELEX for aptamers targeting the surface of eyes and liposomal drug delivery. EXPLORATION (BEIJING, CHINA) 2024; 4:20230008. [PMID: 39175889 PMCID: PMC11335462 DOI: 10.1002/exp.20230008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 01/07/2024] [Indexed: 08/24/2024]
Abstract
Cornea is the major barrier to drug delivery to the eye, which results in low bioavailability and poor efficacy of topical eye treatment. In this work, we first select cornea-binding aptamers using tissue-SELEX on pig cornea. The top two abundant aptamers, Cornea-S1 and Cornea-S2, could bind to pig cornea, and their K d values to human corneal epithelial cells (HCECs) were 361 and 174 nм, respectively. Aptamer-functionalized liposomes loaded with cyclosporine A (CsA) were developed as a treatment for dry eye diseases. The K d of Cornea-S1- or Cornea-S2-functionalized liposomes reduces to 1.2 and 15.1 nм, respectively, due to polyvalent binding. In HCECs, Cornea-S1 or Cornea-S2 enhanced liposome uptake within 15 min and extended retention to 24 h. Aptamer CsA liposomes achieved similar anti-inflammatory and tight junction modulation effects with ten times less CsA than a free drug. In a rabbit dry eye disease model, Cornea-S1 CsA liposomes demonstrated equivalence in sustaining corneal integrity and tear break-up time when compared to commercial CsA eye drops while utilizing a lower dosage of CsA. The aptamers obtained from cornea-SELEX can serve as a general ligand for ocular drug delivery, suggesting a promising avenue for the treatment of various eye diseases and even other diseases.
Collapse
Affiliation(s)
- Ka‐Ying Wong
- Department of Chemistry, Waterloo Institute for NanotechnologyUniversity of WaterlooWaterlooOntarioCanada
- Centre for Eye and Vision Research (CEVR)17 W Hong Kong Science ParkHong KongHong Kong
| | - Yibo Liu
- Department of Chemistry, Waterloo Institute for NanotechnologyUniversity of WaterlooWaterlooOntarioCanada
- Centre for Eye and Vision Research (CEVR)17 W Hong Kong Science ParkHong KongHong Kong
| | - Man‐Sau Wong
- Centre for Eye and Vision Research (CEVR)17 W Hong Kong Science ParkHong KongHong Kong
- Department of Food Science and NutritionThe Hong Kong Polytechnic UniversityHung Hom, KowloonHong Kong
- Research Center for Chinese Medicine InnovationThe Hong Kong Polytechnic UniversityHung Hom, KowloonHong Kong SARP. R. China
| | - Juewen Liu
- Department of Chemistry, Waterloo Institute for NanotechnologyUniversity of WaterlooWaterlooOntarioCanada
- Centre for Eye and Vision Research (CEVR)17 W Hong Kong Science ParkHong KongHong Kong
| |
Collapse
|
45
|
Wang L, Lu X, Zhang Y, Wang Y, Ding Y, Liu M, Fu S, Ge W. Colorimetric aptasensor utilizing MOF-235 with exceptional peroxidase-like activity for the detection of oxytetracycline residues in raw milk. Food Chem 2024; 447:138966. [PMID: 38479142 DOI: 10.1016/j.foodchem.2024.138966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 02/23/2024] [Accepted: 03/05/2024] [Indexed: 04/10/2024]
Abstract
In this work, a simple, convenient and cost-effective colorimetric aptasensor was successfully constructed for the detection of antibiotic residues in raw milk based on the property that aptamer (Apt) synergistically enhances the catalase-like activity of MOF-235. Under optimised conditions, the proposed colorimetric aptasensor exhibited a wide detection range (15-1500 nM) with a low detection limit (6.92 nM). Furthermore, the proposed aptasensor demonstrated high selectivity, good resistance to interference and storage stability. The proposed aptasensor was validated by spiking recovery in camel milk, cow milk and goat milk with satisfactory recoveries, which demonstrated the great potential of the aptasensor for further application in real food samples, and also suggested that MOF-235 can be used as a potential universal platform to build a sensitive detection platform for other targets.
Collapse
Affiliation(s)
- Long Wang
- Northwest A&F Univ, Coll Food Sci & Engn, Shaanxi Engn Res Ctr Dairy Prod Qual Safety & Hlth, Yangling 712100, PR China
| | - Xia Lu
- Northwest A&F Univ, Coll Food Sci & Engn, Shaanxi Engn Res Ctr Dairy Prod Qual Safety & Hlth, Yangling 712100, PR China
| | - Yan Zhang
- Fuping County Inspection and Testing Center, Shaanxi Goat Milk Product Quality Supervision and Inspection Center, Fuping 711700, PR China
| | - Yuxin Wang
- Shaanxi Zhongjian Test Technology Co., Ltd, Xi'an, Shaanxi 71000, PR China
| | - Yi Ding
- Northwest A&F Univ, Coll Food Sci & Engn, Shaanxi Engn Res Ctr Dairy Prod Qual Safety & Hlth, Yangling 712100, PR China
| | - Mengjia Liu
- Northwest A&F Univ, Coll Food Sci & Engn, Shaanxi Engn Res Ctr Dairy Prod Qual Safety & Hlth, Yangling 712100, PR China
| | - Shangchen Fu
- Northwest A&F Univ, Coll Food Sci & Engn, Shaanxi Engn Res Ctr Dairy Prod Qual Safety & Hlth, Yangling 712100, PR China
| | - Wupeng Ge
- Northwest A&F Univ, Coll Food Sci & Engn, Shaanxi Engn Res Ctr Dairy Prod Qual Safety & Hlth, Yangling 712100, PR China.
| |
Collapse
|
46
|
Lu N, Li Z, Su D, Chen J, Zhao J, Gao Y, Liu Q, Liu G, Luo X, Luo R, Deng X, Zhu H, Luo Z. Design of novel chiral self-assembling peptides to explore the efficiency and mechanism of mRNA-FIPV vaccine delivery vehicles. Int J Pharm 2024; 660:124344. [PMID: 38885779 DOI: 10.1016/j.ijpharm.2024.124344] [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: 04/03/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
The enhancement of conventional liposome and lipid nanoparticle (LNP) methodologies in the formulation and deployment of messenger RNA (mRNA) vaccines necessitates further refinement to augment both their effectiveness and biosafety profiles. Additionally, researching these innovative delivery carrier materials represents both a prominent focus and a significant challenge in the current scientific landscape. Here we designed new chiral self-assembling peptides as the delivery carrier for RNA vaccines to study the underlying mechanisms in the feline infectious peritonitis virus (FIPV) model system. Firstly, we successfully transcribed mature enhanced green fluorescent protein (EGFP) mRNA and feline infectious peritonitis virus nucleocapsid (FIPV N) mRNA in vitro from optimized vectors. Subsequently, we developed chiral self-assembling peptide-1 (CSP-1) and chiral self-assembling peptide-2 (CSP-2) peptides, taking into account the physical and chemical characteristics of nucleic acid molecules as well as the principles of self-assembling peptides, with the aim of improving the delivery efficiency of mRNA molecule complexes. We determined the optimal coating ratio between CSP and mRNA by electrophoretic mobility shift assay. We found that the peptides and mRNA complexes can protect the mRNA from RNase A enzyme and efficiently deliver mRNA into cells for target antigen proteins expression. Animal experiments confirmed that CSP-1/mRNA complex can effectively trigger immune response mechanisms involving IFN-γ and T cell activation. It can also stimulate CD4+ and CD8+ T cell proliferation and induce serum antibody titers up to 10,000 times higher. And no pathological changes were observed by immunohistochemistry in liver, spleen, and kidney, indicating that CSP-1 may be a safe and promising delivery system for mRNA vaccines. Methodologically, this research represents a novel endeavor in the utilization of chiral self-assembling peptides within the realm of mRNA vaccines. This approach not only introduces fresh prospects for employing such nanomaterials in various mRNA vaccines but also expands the potential for developing small molecules, proteins, and antibodies. Furthermore, it paves the way for new clinical applications of existing pharmaceuticals.
Collapse
Affiliation(s)
- Na Lu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Zhaoxu Li
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China; Department of Materials Science and Engineering, University of California, Irvine, CA, United States
| | - Di Su
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Jialei Chen
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Jiawei Zhao
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Yu Gao
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Qichen Liu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Guicen Liu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Xinyi Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China; Department of Physiology and Biophysics, University of California, Irvine, CA, United States
| | - Ruyue Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China; Department of Medicine, Northwestern University Feinberg School of Medicine,Chicago, IL,United States
| | - Xiaoyan Deng
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Huifang Zhu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Zhongli Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
47
|
Fu X, Li J, Chen X, Chen H, Wang Z, Qiu F, Xie D, Huang J, Yue S, Cao C, Liang Y, Lu A, Liang C. Repurposing AS1411 for constructing ANM-PROTACs. Cell Chem Biol 2024; 31:1290-1304.e7. [PMID: 38657608 DOI: 10.1016/j.chembiol.2024.03.011] [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: 06/12/2023] [Revised: 11/29/2023] [Accepted: 03/29/2024] [Indexed: 04/26/2024]
Abstract
Proteolysis-targeting chimeras (PROTACs) are heterobifunctional molecules consisting of two ligands joined by a linker, enabling them to simultaneously bind with an E3 ligase and a protein of interest (POI) and trigger proteasomal degradation of the POI. Limitations of PROTAC include lack of potent E3 ligands, poor cell selectivity, and low permeability. AS1411 is an antitumor aptamer specifically recognizing a membrane-nucleus shuttling nucleolin (NCL). Here, we repurpose AS1411 as a ligand for an E3 ligase mouse double minute 2 homolog (MDM2) via anchoring the NCL-MDM2 complex. Then, we construct an AS1411-NCL-MDM2-based PROTAC (ANM-PROTAC) by conjugating AS1411 with large-molecular-weight ligands for "undruggable" oncogenic STAT3, c-Myc, p53-R175H, and AR-V7. We show that the ANM-PROTAC efficiently penetrates tumor cells, recruits MDM2 and degrades the POIs. The ANM-PROTAC achieves tumor-selective distribution and exhibits excellent antitumor activity with no systemic toxicity. This is a PROTAC with built-in tumor-targeting and cell-penetrating capacities.
Collapse
Affiliation(s)
- Xuekun Fu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Jin Li
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xinxin Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hongzhen Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhuqian Wang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Fang Qiu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Duoli Xie
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Jie Huang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Siran Yue
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Chunhao Cao
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Yiying Liang
- Shenzhen LingGene Biotech Co., Ltd, Shenzhen 518055, China
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510006, China; Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China.
| |
Collapse
|
48
|
Gong H, Zhang Y, Xue Y, Fang B, Li Y, Zhu X, Du Y, Peng P. NETosis-Inspired Cell Surface-Constrained Framework Nucleic Acids Traps (FNATs) for Cascaded Extracellular Recognition and Cellular Behavior Modulation. Angew Chem Int Ed Engl 2024; 63:e202319908. [PMID: 38693057 DOI: 10.1002/anie.202319908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/11/2024] [Accepted: 04/29/2024] [Indexed: 05/03/2024]
Abstract
Upon pathogenic stimulation, activated neutrophils release nuclear DNA into the extracellular environment, forming web-like DNA structures known as neutrophil extracellular traps (NETs), which capture and kill bacteria, fungi, and cancer cells. This phenomenon is commonly referred to as NETosis. Inspired by this, we introduce a cell surface-constrained web-like framework nucleic acids traps (FNATs) with programmable extracellular recognition capability and cellular behavior modulation. This approach facilitates dynamic key chemical signaling molecule recognition such as adenosine triphosphate (ATP), which is elevated in the extracellular microenvironment, and triggers FNA self-assembly. This, in turn, leads to in situ tightly interwoven FNAs formation on the cell surface, thereby inhibiting target cell migration. Furthermore, it activates a photosensitizer-capturing switch, chlorin e6 (Ce6), and induces cell self-destruction. This cascade platform provides new potential tools for visualizing dynamic extracellular activities and manipulating cellular behaviors using programmable in situ self-assembling DNA molecular devices.
Collapse
Affiliation(s)
- Hangsheng Gong
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Yihan Zhang
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Yuan Xue
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Bowen Fang
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Yuting Li
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Xudong Zhu
- School of Physical Sciences, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yi Du
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Pai Peng
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230032, China
| |
Collapse
|
49
|
Abdal Dayem A, Yan E, Do M, Kim Y, Lee Y, Cho SG, Kim DH. Engineering extracellular vesicles for ROS scavenging and tissue regeneration. NANO CONVERGENCE 2024; 11:24. [PMID: 38922501 PMCID: PMC11208369 DOI: 10.1186/s40580-024-00430-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024]
Abstract
Stem cell therapy holds promise for tissue regeneration, yet significant challenges persist. Emerging as a safer and potentially more effective alternative, extracellular vesicles (EVs) derived from stem cells exhibit remarkable abilities to activate critical signaling cascades, thereby facilitating tissue repair. EVs, nano-scale membrane vesicles, mediate intercellular communication by encapsulating a diverse cargo of proteins, lipids, and nucleic acids. Their therapeutic potential lies in delivering cargos, activating signaling pathways, and efficiently mitigating oxidative stress-an essential aspect of overcoming limitations in stem cell-based tissue repair. This review focuses on engineering and applying EVs in tissue regeneration, emphasizing their role in regulating reactive oxygen species (ROS) pathways. Additionally, we explore strategies to enhance EV therapeutic activity, including functionalization and incorporation of antioxidant defense proteins. Understanding these molecular mechanisms is crucial for optimizing EV-based regenerative therapies. Insights into EV and ROS signaling modulation pave the way for targeted and efficient regenerative therapies harnessing the potential of EVs.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ellie Yan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Minjae Do
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yoojung Kim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Yeongseo Lee
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
- R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120, Neungdong-ro, Gwangjin- gu, Seoul, 05029, Republic of Korea.
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, 21205, USA.
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Institute for NanoBiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
50
|
Kumar S, Mohan A, Sharma NR, Kumar A, Girdhar M, Malik T, Verma AK. Computational Frontiers in Aptamer-Based Nanomedicine for Precision Therapeutics: A Comprehensive Review. ACS OMEGA 2024; 9:26838-26862. [PMID: 38947800 PMCID: PMC11209897 DOI: 10.1021/acsomega.4c02466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/09/2024] [Accepted: 05/28/2024] [Indexed: 07/02/2024]
Abstract
In the rapidly evolving landscape of nanomedicine, aptamers have emerged as powerful molecular tools, demonstrating immense potential in targeted therapeutics, diagnostics, and drug delivery systems. This paper explores the computational features of aptamers in nanomedicine, highlighting their advantages over antibodies, including selectivity, low immunogenicity, and a simple production process. A comprehensive overview of the aptamer development process, specifically the Systematic Evolution of Ligands by Exponential Enrichment (SELEX) process, sheds light on the intricate methodologies behind aptamer selection. The historical evolution of aptamers and their diverse applications in nanomedicine are discussed, emphasizing their pivotal role in targeted drug delivery, precision medicine and therapeutics. Furthermore, we explore the integration of artificial intelligence (AI), machine learning (ML), Internet of Things (IoT), Internet of Medical Things (IoMT), and nanotechnology in aptameric development, illustrating how these cutting-edge technologies are revolutionizing the selection and optimization of aptamers for tailored biomedical applications. This paper also discusses challenges in computational methods for advancing aptamers, including reliable prediction models, extensive data analysis, and multiomics data incorporation. It also addresses ethical concerns and restrictions related to AI and IoT use in aptamer research. The paper examines progress in computer simulations for nanomedicine. By elucidating the importance of aptamers, understanding their superiority over antibodies, and exploring the historical context and challenges, this review serves as a valuable resource for researchers and practitioners aiming to harness the full potential of aptamers in the rapidly evolving field of nanomedicine.
Collapse
Affiliation(s)
- Shubham Kumar
- School
of Bioengineering and Biosciences, Lovely
Professional University, Phagwara, Punjab 144001, India
| | - Anand Mohan
- School
of Bioengineering and Biosciences, Lovely
Professional University, Phagwara, Punjab 144001, India
| | - Neeta Raj Sharma
- School
of Bioengineering and Biosciences, Lovely
Professional University, Phagwara, Punjab 144001, India
| | - Anil Kumar
- Gene
Regulation Laboratory, National Institute
of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Madhuri Girdhar
- Division
of Research and Development, Lovely Professional
University, Phagwara 144401, Punjab, India
| | - Tabarak Malik
- Department
of Biomedical Sciences, Institute of Health, Jimma University, MVJ4+R95 Jimma, Ethiopia
| | - Awadhesh Kumar Verma
- School
of Bioengineering and Biosciences, Lovely
Professional University, Phagwara, Punjab 144001, India
| |
Collapse
|