1
|
Gomari MM, Ghantabpour T, Pourgholam N, Rostami N, Hatfield SM, Namazifar F, Abkhiz S, Eslami SS, Ramezanpour M, Darestanifarahani M, Astsaturov I, Bencherif SA. Breaking barriers: Smart vaccine platforms for cancer immunomodulation. Cancer Commun (Lond) 2025. [PMID: 39901621 DOI: 10.1002/cac2.70002] [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: 07/23/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Despite significant advancements in cancer treatment, current therapies often fail to completely eradicate malignant cells. This shortfall underscores the urgent need to explore alternative approaches such as cancer vaccines. Leveraging the immune system's natural ability to target and kill cancer cells holds great therapeutic potential. However, the development of cancer vaccines is hindered by several challenges, including low stability, inadequate immune response activation, and the immunosuppressive tumor microenvironment, which limit their efficacy. Recent progress in various fields, such as click chemistry, nanotechnology, exosome engineering, and neoantigen design, offer innovative solutions to these challenges. These achievements have led to the emergence of smart vaccine platforms (SVPs), which integrate protective carriers for messenger ribonucleic acid (mRNA) with functionalization strategies to optimize targeted delivery. Click chemistry further enhances SVP performance by improving the encapsulation of mRNA antigens and facilitating their precise delivery to target cells. This review highlights the latest developments in SVP technologies for cancer therapy, exploring both their opportunities and challenges in advancing these transformative approaches.
Collapse
Affiliation(s)
- Mohammad Mahmoudi Gomari
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Taha Ghantabpour
- Department of Anatomy, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Nima Pourgholam
- School of Nursing and Midwifery, Iran University of Medical Science, Tehran, Iran
| | - Neda Rostami
- Department of Chemical Engineering, Arak University, Arak, Iran
| | - Stephen M Hatfield
- New England Inflammation and Tissue Protection Institute, Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | | | - Shadi Abkhiz
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Sadegh Eslami
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Molecular Proteomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Mahsa Ramezanpour
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Darestanifarahani
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey, USA
| | - Igor Astsaturov
- Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Sidi A Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- Polymers, Biopolymers Surfaces (PBS) Laboratory, National Center for Scientific Research (CNRS) Mixed Research Unit (UMR) 6270, University Rouen Normandie, Rouen, France
| |
Collapse
|
2
|
Theivendren P, Pavadai P, Veerachamy S, Palanisamy P, Kunjiappan S. Surface receptor-targeted protein-based nanocarriers for drug delivery: advances in cancer therapy. NANOTECHNOLOGY 2025; 36:122003. [PMID: 39847811 DOI: 10.1088/1361-6528/adad7a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 01/23/2025] [Indexed: 01/25/2025]
Abstract
Significant progress has been made in cancer therapy with protein-based nanocarriers targeted directly to surface receptors for drug delivery. The nanocarriers are a potentially effective solution for the potential drawbacks of traditional chemotherapy, such as lack of specificity, side effects, and development resistance. Peptides as nanocarriers have been designed based on their biocompatible, biodegradable, and versatile functions to deliver therapeutic agents into cancer cells, reduce systemic toxicity, and maximize therapy efficacy through utilizing targeted ligands such as antibodies, amino acids, vitamins, and other small molecules onto protein-based nanocarriers and thus ensuring that drugs selectively accumulate in the cancer cells instead of healthy organs/drug release at a target site without effects on normal cells, which inherently caused less systemic toxicity/off-target effect. Moreover, their intrinsic protein backbone naturally degradesin vivo, providing another level of safety over synthetic materials. Various issues like immunogenicity, mass production, and quality control must be addressed for widespread use. However, further studies are necessary to perfect protein engineering and improve drug loading, protein modification, and targeting. Thus, it can be concluded that protein-based nanocarriers targeted against the surface receptors would help achieve cancer management in a more focused manner, thus minimizing toxicity. The further development of these nanoparticles could bring a significant change in cancer treatment so that more personalized, targeted, and safe therapies would be available to all patients.
Collapse
Affiliation(s)
- Panneerselvam Theivendren
- Department of Pharmaceutical Chemistry & Analysis, School of Pharmaceutical Sciences, Vels Institute of Science, Technology & Advanced Studies, Pallavaram, Chennai 600117, India
| | - Parasuraman Pavadai
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S. Ramaiah University of Applied Sciences, M.S.R. Nagar, Bengaluru 560054, Karnataka, India
| | - Suganthan Veerachamy
- School of Electronics Engineering, Vellore Institute of Technology, Vellore 632014, Tamilnadu, India
| | - Ponnusamy Palanisamy
- School of Mechanical Engineering, Vellore Institute of Technology, Vellore 632014, Tamilnadu, India
| | - Selvaraj Kunjiappan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, Tamil Nadu, India
| |
Collapse
|
3
|
Mohan A, Roy I. Exploring the diagnostic landscape: Portable aptasensors in point-of-care testing. Anal Biochem 2025; 700:115788. [PMID: 39884526 DOI: 10.1016/j.ab.2025.115788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/31/2024] [Accepted: 01/25/2025] [Indexed: 02/01/2025]
Abstract
Aptamers, discovered in the 1990s, have marked a significant milestone in the fields of therapeutics and diagnostics. This review provides a comprehensive survey of aptamers, focusing on their diagnostic applications. It especially encapsulates a decade of aptamer, encompassing research, patents, and market trends. The unique properties and inherent stability of aptamers are discussed, highlighting their potential for various clinical applications. It goes on to introduce biosensor design, emphasizing the advantages of aptamers over antibodies as conventional molecular recognition interface. The operation and design of aptasensors are examined, with a focus on single- and dual-site binding configurations and their respective recognition modes. Paper-based sensors are highlighted as cost-effective, user-friendly alternatives that are gaining widespread adoption, particularly in point-of-care platforms.
Collapse
Affiliation(s)
- Anu Mohan
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab, 160062, India
| | - Ipsita Roy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab, 160062, India.
| |
Collapse
|
4
|
Pipis N, James BD, Allen JB. Multifunctional DNA-Collagen Biomaterials: Developmental Advances and Biomedical Applications. ACS Biomater Sci Eng 2025. [PMID: 39869382 DOI: 10.1021/acsbiomaterials.4c01475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The complexation of nucleic acids and collagen forms a platform biomaterial greater than the sum of its parts. This union of biomacromolecules merges the extracellular matrix functionality of collagen with the designable bioactivity of nucleic acids, enabling advances in regenerative medicine, tissue engineering, gene delivery, and targeted therapy. This review traces the historical foundations and critical applications of DNA-collagen complexes and highlights their capabilities, demonstrating them as biocompatible, bioactive, and tunable platform materials. These complexes form structures across length scales, including nanoparticles, microfibers, and hydrogels, a process controlled by the relative amount of each component and the type of nucleic acid and collagen. The broad distribution of different types of collagen within the body contributes to the extensive biological relevance of DNA-collagen complexes. Functional nucleic acids can form these complexes, such as siRNA, antisense oligonucleotides, DNA origami nanostructures, and, in particular, single-stranded DNA aptamers, often distinguished by their rapid self-assembly at room temperature and formation without external stimuli and modifications. The simple and seamless integration of nucleic acids within collagenous matrices enhances biomimicry and targeted bioactivity, and provides stability against enzymatic degradation, positioning DNA-collagen complexes as an advanced biomaterial system for many applications including angiogenesis, bone tissue regeneration, wound healing, and more.
Collapse
Affiliation(s)
- Nikolaos Pipis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Bryan D James
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, United States
| | - Josephine B Allen
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
- Department of Materials Science & Engineering, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
5
|
Ji D, Wang B, Lo KW, Tsang CM, Kwok CK. Pre-Defined Stem-Loop Structure Library for the Discovery of L-RNA Aptamers that Target RNA G-Quadruplexes. Angew Chem Int Ed Engl 2025; 64:e202417247. [PMID: 39462761 DOI: 10.1002/anie.202417247] [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: 09/08/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
L-RNA aptamers have been developed to target G-quadruplexes (G4s) and regulate G4-mediated gene expression. However, the aptamer selection process is laborious and challenging, and aptamer identification is subject to high failure rates. By analyzing the previously reported G4-binding L-RNA aptamers, we found that the stem-loop (SL) structure is favored by G4 binding. Herein, we present a robust and effective G4-SLSELEX-Seq platform specifically for G4 targets by introducing a pre-defined stem-loop structure library during the SELEX process. Using G4-SLSELEX-Seq, we identified an L-RNA aptamer, L-Apt1-12, for the Epstein-Barr nuclear antigen 1 (EBNA1) RNA G4 (rG4) in just three selection rounds. L-Apt1-12 maintained the stem-loop structure initially introduced, and possessed a unique G-triplex motif that is important for the strong binding affinity and specificity to EBNA1 rG4. L-Apt1-12 effectively downregulated endogenous EBNA1 protein expression in human cancer cells and showed selective toxicity towards Epstein-Barr virus (EBV)-positive cancer cells, highlighting its potential for targeted therapy against EBV-associated cancers. Furthermore, we demonstrated the robustness and generality of G4-SLSELEX-Seq by selecting L-RNA aptamers for the amyloid precursor protein (APP) rG4 and the hepatitis C virus subtype 1a (HCV-1a) rG4, obtaining high-affinity aptamers in three selection rounds. These findings demonstrated G4-SLSELEX-Seq as a robust and efficient platform for the selection of rG4-targeting L-RNA aptamers.
Collapse
Grants
- 32471343, 32222089 the National Natural Science Foundation of China (NSFC) Projects
- RFS2425-1S02, CityU 11100123, CityU 11100222, CityU 11100421 Research Grants Council (RGC) of the Hong Kong Special Administra-tive Region
- 9509003 Croucher Foundation
- SCRF/0037, SCRF/0040, SCRF/0070 State Key Laboratory in Marine Pollution
- 7030001, 6000827, 9678302 City University of Hong Kong projects
- MRP/036/21X The Innovation and Technology Fund (Midstream Research Programme for Universities
- AoE/M-401/20, 14101721, 08191046 Research Grant Council, Hong Kong
- 14116124, 14113620, 14114523 Research Grants Council-General Research Fund (GRF)
- 24114922 Early Career Scheme (ECS)
- 09203176 Health and Medical Research Fund
- FIA2020/A/01 Faculty Innovation Award
Collapse
Affiliation(s)
- Danyang Ji
- Department of Chemistry and State Key Laboratory of Marine Pollution, City University of Hong Kong Kowloon Tong, Hong Kong SAR, China
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Bo Wang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Translational Oncology, Sir YK Pao Centre for Cancer, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Translational Oncology, Sir YK Pao Centre for Cancer, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chi Man Tsang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Translational Oncology, Sir YK Pao Centre for Cancer, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chun Kit Kwok
- Department of Chemistry and State Key Laboratory of Marine Pollution, City University of Hong Kong Kowloon Tong, Hong Kong SAR, China
- Shenzhen Research Institute of City University of Hong Kong, Shenzhen, China
| |
Collapse
|
6
|
Collins J, Farnsworth NL. Active targeting of type 1 diabetes therapies to pancreatic beta cells using nanocarriers. Diabetologia 2025:10.1007/s00125-024-06356-5. [PMID: 39847085 DOI: 10.1007/s00125-024-06356-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/21/2024] [Indexed: 01/24/2025]
Abstract
Type 1 diabetes is an autoimmune disease characterised by the destruction of pancreatic beta cells, resulting in lifelong insulin dependence. Although exogenous insulin can maintain glycaemic control, this approach does not protect residual or replacement pancreatic beta cells from immune-mediated death. Current therapeutics designed to protect functional beta cell mass or promote beta cell proliferation and regeneration can have off-target effects, resulting in higher dose requirements and adverse side effects. Targeted drug delivery using nanocarriers has demonstrated potential for overcoming these limitations. The critical bottleneck limiting the development of beta cell-targeted therapies is a lack of highly specific beta cell markers. This review provides an overview of the use of nanocarriers for cell-targeted delivery and the current state of the field of beta cell targeting. Technologies such as systematic evolution of ligands by exponential enrichment (SELEX) aptamer selection, phage display screening, and omics datasets from human samples are highlighted as tools to identify novel beta cell-specific targets that can be combined with nanocarriers for targeted delivery of therapeutics. Ultimately, beta cell-targeted therapies using nanocarriers present a unique opportunity to develop tailored treatments for each stage of type 1 diabetes with the goal of providing individuals with treatment options that prevent further progression or reverse the course of the disease.
Collapse
Affiliation(s)
- Jillian Collins
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO, USA
| | - Nikki L Farnsworth
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO, USA.
| |
Collapse
|
7
|
Liu S, Rahman MR, Wu H, Qin W, Wang Y, Su G. Development and application of hydrogels in pathogenic bacteria detection in foods. J Mater Chem B 2025; 13:1229-1251. [PMID: 39690945 DOI: 10.1039/d4tb01341g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Hydrogels are 3D networks of water-swollen hydrophilic polymers. It possesses unique properties (e.g., carrying biorecognition elements and creating a micro-environment) that make it highly suitable for bacteria detection (e.g., expedited and effective bacteria detection) and mitigation of bacterial contamination in specific environments (e.g., food systems). This study first introduces the materials used to create hydrogels for bacteria detection and the mechanisms for detection. We also summarize different hydrogel-based detection methods that rely on external stimuli and biorecognition elements, such as enzymes, temperature, pH, antibodies, and oligonucleotides. Subsequently, a range of widely utilized bacterial detection technologies were discussed where recently hydrogels are being used. These modifications allow for precise, real-time diagnostics across varied food matrices, responding effectively to industry needs for sensitivity, scalability, and portability. After highlighting the utilization of hydrogels and their role in these detection techniques, we outline limitations and advancements in the methods for the detection of foodborne pathogenic bacteria, especially the potential application of hydrogels in the food industry.
Collapse
Affiliation(s)
- Shuxiang Liu
- College of Food Science, Sichuan Agricultural University, Ya'an 625014, China.
| | - Md Rashidur Rahman
- College of Food Science, Sichuan Agricultural University, Ya'an 625014, China.
| | - Hejun Wu
- College of Science, Sichuan Agricultural University, Ya'an, 625000, China.
| | - Wen Qin
- College of Food Science, Sichuan Agricultural University, Ya'an 625014, China.
| | - Yanying Wang
- College of Science, Sichuan Agricultural University, Ya'an, 625000, China.
| | - Gehong Su
- College of Science, Sichuan Agricultural University, Ya'an, 625000, China.
| |
Collapse
|
8
|
Liu J, Wang L, Peng Y, Long S, Zeng H, Deng M, Xiang W, Liu B, Hu X, Liu X, Xie J, Hou W, Tang J, Liu J. A novel therapeutic strategy utilizing EpCAM aptamer-conjugated gemcitabine for targeting bladder cancer and cancer stem cells. Biomater Sci 2025. [PMID: 39835931 DOI: 10.1039/d4bm01471e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Gemcitabine (GEM) is a first line chemotherapy drug for bladder cancer (BCa). GEM's lack of specificity has led to disadvantages, resulting in low efficiency, especially when combined with the targeted treatment of BCa stem cells (CSCs), which is considered the cause of BCa recurrence and progression. To enhance the anti-cancer effect and reduce the side effects of GEM targeting of BCa cells/CSCs, an aptamer drug conjugate (ApDC) targeted delivery system was used to improve the efficiency of GEM in BCa therapy using EpCAM aptamer-GEM conjugates based on the epithelial cell adhesion molecule (EpCAM), which is highly expressed on the cell membrane of BCa cells/CSCs. We designed and synthesized EpCAM aptamer gemcitabine conjugates (EpCAM-GEMs, one aptamer carried three GEMs). The targeting effect of EpCAM-GEMs was examined in a xenograft model using an in vivo imaging system. To evaluate the antitumor activity and mechanism of EpCAM-GEMs, Cell Counting Kit-8, apoptosis and colony formation assays; BCa CSC xenotransplantation; xenotransplantation of subcutaneous tumors; a lung metastasis model; an in situ model; and biosafety assessment were used in vitro and in vivo. EpCAM is highly expressed on the surface of BCa cells/CSCs. EpCAM-GEMs were automatically synthesized using a DNA synthesizer, were stable in serum, and selectively delivered GEM to kill BCa cells/CSCs. EpCAM-GEMs entered BCa cells via macropinocytosis, released GEM to inhibit DNA synthesis, and degraded all BCa cells under the action of a BCa cell intracellular phosphatase; however, they did not kill normal cells because of their low EpCAM expression. EpCAM-GEMs inhibited BCa growth and metastasis in three bladder tumor models, with good biosafety. These results demonstrated the targeted anti-tumor efficiency and good biosafety of EpCAM-GEMs in BCa treatment, which will provide a new therapeutic strategy in BCa biomarker targeted therapy.
Collapse
Affiliation(s)
- Jiahao Liu
- Department of Urology, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Long Wang
- Department of Urology, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Yongbo Peng
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, No. 1, Yixueyuan Road, Chongqing, 400016, China
| | - Shuyang Long
- School of Medicine, Hunan University of Chinese Medicine, Grade 2024, China
| | - Hongliang Zeng
- Institute of Chinese Materia Medica, Hunan Academy of Chinese Medicine, No. 8, Yuehua Road, Changsha 410013, China
| | - Minhua Deng
- Department of Urology, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, Guangdong, China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, No. 651, Dongfeng Road East, Guangzhou, 510060, Guangdong, China
| | - Wei Xiang
- Department of Urology, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Biao Liu
- Department of Urology, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Xing Hu
- Department of Urology, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Xuewen Liu
- Department of Oncology, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Changsha, 410013, Hunan, China
| | - Jianfei Xie
- Department of Nursing, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Changsha, 410013, Hunan, China
| | - Weibin Hou
- Department of Urology, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Jin Tang
- Department of Urology, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Jianye Liu
- Department of Urology, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
9
|
Dantsu Y, Zhang Y, Zhang W. Selection of a Fluorinated Aptamer Targeting the Viral RNA Frameshift Element with Different Chiralities. Biochemistry 2025; 64:448-457. [PMID: 39772548 DOI: 10.1021/acs.biochem.4c00606] [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: 01/11/2025]
Abstract
The development of RNA aptamers with high specificity and affinity for target molecules is a critical advancement in the field of therapeutic and diagnostic applications. This study presents the selection of a 2'-fluoro-modified mirror-image RNA aptamer through the in vitro SELEX process. Using a random RNA library, we performed iterative rounds of selection and amplification to enrich aptamers that bind specifically to the viral attenuator hairpin RNA containing the opposite chirality, which is an important part of the frameshift element. The unnatural chirality of the aptamer improved its enzymatic stability, and the incorporation of 2'-fluoro modifications was crucial in enhancing the binding affinity of the aptamers. After nine rounds of SELEX, the enriched RNA pool was sequenced and analyzed, revealing the dominant aptamer sequences. The selected 2'-fluoro-modified mirror-image RNA aptamer demonstrated a dissociation constant of approximately 1.6 μM, indicating moderate binding affinity with the target and exceptional stability against nuclease degradation. Our findings highlight the potential of 2'-fluoro-modified mirror-image RNA aptamers in enhancing the stability and utility of RNA-based therapeutics and diagnostics, paving the way for future applications in diverse biomedical fields.
Collapse
Affiliation(s)
- Yuliya Dantsu
- Department of Biochemistry and Molecular Biology, Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Ying Zhang
- Department of Biochemistry and Molecular Biology, Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Wen Zhang
- Department of Biochemistry and Molecular Biology, Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| |
Collapse
|
10
|
Lapin D, Sharma A, Wang P. Extracellular cold-inducible RNA-binding protein in CNS injury: molecular insights and therapeutic approaches. J Neuroinflammation 2025; 22:12. [PMID: 39838468 PMCID: PMC11752631 DOI: 10.1186/s12974-025-03340-7] [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: 11/13/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025] Open
Abstract
Central nervous system (CNS) injuries, such as ischemic stroke (IS), intracerebral hemorrhage (ICH) and traumatic brain injury (TBI), are a significant global burden. The complex pathophysiology of CNS injury is comprised of primary and secondary injury. Inflammatory secondary injury is incited by damage-associated molecular patterns (DAMPs) which signal a variety of resident CNS cells and infiltrating immune cells. Extracellular cold-inducible RNA-binding protein (eCIRP) is a DAMP which acts through multiple immune and non-immune cells to promote inflammation. Despite the well-established role of eCIRP in systemic and sterile inflammation, its role in CNS injury is less elucidated. Recent literature suggests that eCIRP is a pleiotropic inflammatory mediator in CNS injury. eCIRP is also being evaluated as a clinical biomarker to indicate prognosis in CNS injuries. This review provides a broad overview of CNS injury, with a focus on immune-mediated secondary injury and neuroinflammation. We then review what is known about eCIRP in CNS injury, and its known mechanisms in both CNS and non-CNS cells, identifying opportunities for further study. We also explore eCIRP's potential as a prognostic marker of CNS injury severity and outcome. Next, we provide an overview of eCIRP-targeting therapeutics and suggest strategies to develop these agents to ameliorate CNS injury. Finally, we emphasize exploring novel molecular mechanisms, aside from neuroinflammation, by which eCIRP acts as a critical mediator with significant potential as a therapeutic target and prognostic biomarker in CNS injury.
Collapse
Affiliation(s)
- Dmitriy Lapin
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
- Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
| | - Archna Sharma
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA.
- Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA.
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA.
- Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA.
| |
Collapse
|
11
|
Nguyen JVL, Meziadi A, Nassif C, Da Fonte D, Malic L, Tabrizian M. Combinatorial Nanoparticle-Bound ssDNA Oligonucleotide Library Synthesized by Split-and-Pool Synthesis. ACS APPLIED BIO MATERIALS 2025; 8:844-853. [PMID: 39829268 DOI: 10.1021/acsabm.4c01681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Synthetic ssDNA oligonucleotides hold great potential for various applications, including DNA aptamers, DNA digital data storage, DNA origami, and synthetic genomes. In these contexts, precise control over the synthesis of the ssDNA strands is essential for generating combinatorial sequences with user-defined parameters. Desired features for creating synthetic DNA oligonucleotides include easy manipulation of DNA strands, effective detection of unique DNA sequences, and a straightforward mechanism for strand elongation and termination. In this study, we present a split-and-pool method for generating synthetic DNA oligonucleotides on nanoparticles, enabling the creation of scalable combinatorial libraries. Our approach involves coupling DNA to nanoparticles, ligating double-digested fragments for orientation-specific synthesis, and attaching a final single-digested fragment to ensure strand termination. We assess the quality of our method by characterizing both the DNA and the nanoparticles used as solid supports, confirming that our method produces scalable, combinatorial nanoparticle-bound ssDNA libraries with controllable strand lengths.
Collapse
Affiliation(s)
- John V L Nguyen
- Department of Biomedical Engineering, McGill University, 3775 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Ahlem Meziadi
- Department of Biomedical Engineering, McGill University, 3775 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Christina Nassif
- Life Sciences Division, National Research Council of Canada, 75 de Mortagne Boulevard, Boucherville, Quebec J4B 6Y4, Canada
| | - Dillon Da Fonte
- Life Sciences Division, National Research Council of Canada, 75 de Mortagne Boulevard, Boucherville, Quebec J4B 6Y4, Canada
| | - Lidija Malic
- Department of Biomedical Engineering, McGill University, 3775 University Street, Montreal, Quebec H3A 2B4, Canada
- Life Sciences Division, National Research Council of Canada, 75 de Mortagne Boulevard, Boucherville, Quebec J4B 6Y4, Canada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, McGill University, 3775 University Street, Montreal, Quebec H3A 2B4, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, 2001 McGill College Avenue, Montreal, Quebec H3A 1G1, Canada
| |
Collapse
|
12
|
Ou L, Setegne MT, Elliot J, Shen F, Dassama LMK. Protein-Based Degraders: From Chemical Biology Tools to Neo-Therapeutics. Chem Rev 2025. [PMID: 39818743 DOI: 10.1021/acs.chemrev.4c00595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
The nascent field of targeted protein degradation (TPD) could revolutionize biomedicine due to the ability of degrader molecules to selectively modulate disease-relevant proteins. A key limitation to the broad application of TPD is its dependence on small-molecule ligands to target proteins of interest. This leaves unstructured proteins or those lacking defined cavities for small-molecule binding out of the scope of many TPD technologies. The use of proteins, peptides, and nucleic acids (otherwise known as "biologics") as the protein-targeting moieties in degraders addresses this limitation. In the following sections, we provide a comprehensive and critical review of studies that have used proteins and peptides to mediate the degradation and hence the functional control of otherwise challenging disease-relevant protein targets. We describe existing platforms for protein/peptide-based ligand identification and the drug delivery systems that might be exploited for the delivery of biologic-based degraders. Throughout the Review, we underscore the successes, challenges, and opportunities of using protein-based degraders as chemical biology tools to spur discoveries, elucidate mechanisms, and act as a new therapeutic modality.
Collapse
Affiliation(s)
- Lisha Ou
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan ChEM-H Institute, Stanford University, Stanford, California 94305, United States
| | - Mekedlawit T Setegne
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan ChEM-H Institute, Stanford University, Stanford, California 94305, United States
| | - Jeandele Elliot
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Fangfang Shen
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Laura M K Dassama
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan ChEM-H Institute, Stanford University, Stanford, California 94305, United States
- Department of Microbiology & Immunology, Stanford School of Medicine, Stanford, California 94305, United States
| |
Collapse
|
13
|
Wang J, Zhou Y, Sun L, Liu T, Qu H, Mao Y, Zheng L. Binding-Site Directed Selection and Large-Scale Click-Synthesis of a Coagulation Factor XIa-Inhibiting Circular DNA Aptamer. Chemistry 2025:e202404372. [PMID: 39822082 DOI: 10.1002/chem.202404372] [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: 11/27/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/19/2025]
Abstract
Factor XIa (FXIa) is a plasma protease that catalyzes the intrinsic pathway of blood coagulation, thus being regarded as a promising antithrombotic target. Circular DNA aptamers, with their dramatically enhanced biological and structural stability, hold great potential as new-generation DNA-based anticoagulants. However, the functional selection of circular aptamers and large-scale synthesis of them remains a substantial challenge. In this study, we explored a binding-site directed selection strategy to enrich circular DNA aptamers that can target the catalytic domain of FXIa and inhibit its function. An asymmetrical dumbbell structured DNA library with self-shielding primer-binding site was utilized for selection. This approach has resulted in the identification of a circular DNA aptamer, named FICAPT1, which demonstrates a high binding affinity (Kd=20 nM), notable stability (with a half-life of 16 h in human plasma) and outstanding anticoagulation activity (with significantly prolonged aPTT), showing great promise as a novel anticoagulant. Moreover, a refined version of FICAPT1, named as CC-FICAPT1, was quantitatively synthesized (~2 mg/mL) by taking advantage of the pre-designed structure and high cyclization efficiency of click chemistry. The approach presented in this study enables the efficient generation of functional circular DNA aptamers and allows for their synthesis in a satisfactory scale, making it adaptable for the production of various circular aptamers for therapeutic applications.
Collapse
Affiliation(s)
- Jie Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Yuefei Zhou
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Long Sun
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Tao Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Hao Qu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Yu Mao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Lei Zheng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| |
Collapse
|
14
|
Xu K, Feng H, Zhao R, Huang Y. Targeting Tetraspanins at Cell Interfaces: Functional Modulation and Exosome-Based Drug Delivery for Precise Disease Treatment. ChemMedChem 2025; 20:e202400664. [PMID: 39415492 DOI: 10.1002/cmdc.202400664] [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: 08/26/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/18/2024]
Abstract
Tetraspanins are key players in various physiological and pathological processes, including malignancy, immune response, fertilization, and infectious disease. Affinity ligands targeting the interactions between tetraspanins and partner proteins are promising for modulating downstream signaling pathways, thus emerging as attractive candidates for interfering related biological functions. Due to the involvement in vesicle biogenesis and cargo trafficking, tetraspanins are also regarded as exosome markers, and become molecular targets for drug loading and delivery. Given the rapid development in these areas, this minireview focuses on recent advances in design and engineering of affinity binders toward tetraspanins including CD63, CD81, and CD9. Their mechanism of actions in modulating protein interactions at cell interfaces and treatment of malignant diseases are discussed. Strategies for constructing exosome-based drug delivery platforms are also reviewed, with emphasis on the important roles of tetraspanins and the affinity ligands. Finally, challenges and future development of tetraspanin-targeting therapy and exosomal drug delivery platforms are also discussed.
Collapse
Affiliation(s)
- Kun Xu
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huixia Feng
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Rui Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanyan Huang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
15
|
Guo W, Ma Y, Mou Q, Shao X, Lyu M, Garcia V, Kong L, Lewis W, Yang Z, Lu S, Lu Y. Sialic acid aptamer and RNA in situ hybridization-mediated proximity ligation assay for spatial imaging of glycoRNAs in single cells. Nat Protoc 2025:10.1038/s41596-024-01103-x. [PMID: 39779896 DOI: 10.1038/s41596-024-01103-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 11/05/2024] [Indexed: 01/11/2025]
Abstract
Glycosylated RNAs (glycoRNAs) have recently emerged as a new class of molecules of substantial interest owing to their potential roles in cellular processes and diseases. However, studying glycoRNAs is challenging owing to the lack of effective research tools including, but not limited to, imaging techniques to study the spatial distribution of glycoRNAs. Recently, we reported the development of a glycoRNA imaging technique, called sialic acid aptamer and RNA in situ hybridization-mediated proximity ligation assay (ARPLA), to visualize sialic acid-containing glycoRNAs with high sensitivity and specificity. Here we describe the experimental design principles and detailed step-by-step procedures for ARPLA-assisted glycoRNA imaging across multiple cell types. The procedure includes details for target selection, oligo design and preparation, optimized steps for RNA in situ hybridization, glycan recognition, proximity ligation, rolling circle amplification and a guideline for image acquisition and analysis. With properly designed probe sets and cells prepared, ARPLA-based glycoRNA imaging can typically be completed within 1 d by users with expertise in biochemistry and fluorescence microscopy. The ARPLA approach enables researchers to explore the spatial distribution, trafficking and functional contributions of glycoRNAs in various cellular processes.
Collapse
Affiliation(s)
- Weijie Guo
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
- Interdisciplinary Life Sciences Graduate Programs, The University of Texas at Austin, Austin, TX, USA
| | - Yuan Ma
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA
| | - Quanbing Mou
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA
| | - Xiangli Shao
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA
| | - Mingkuan Lyu
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA
| | - Valeria Garcia
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
- Interdisciplinary Life Sciences Graduate Programs, The University of Texas at Austin, Austin, TX, USA
| | - Linggen Kong
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
- Interdisciplinary Life Sciences Graduate Programs, The University of Texas at Austin, Austin, TX, USA
| | - Whitney Lewis
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA
| | - Zhenglin Yang
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA
| | - Shuya Lu
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA
| | - Yi Lu
- Interdisciplinary Life Sciences Graduate Programs, The University of Texas at Austin, Austin, TX, USA.
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
16
|
Jeon BJ, Guareschi MM, Stewart JM, Wu E, Gopinath A, Arroyo-Currás N, Dauphin-Ducharme P, Plaxco KW, Lukeman PS, Rothemund PWK. Modular DNA origami-based electrochemical detection of DNA and proteins. Proc Natl Acad Sci U S A 2025; 122:e2311279121. [PMID: 39793064 PMCID: PMC11725875 DOI: 10.1073/pnas.2311279121] [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: 12/10/2023] [Accepted: 10/15/2024] [Indexed: 01/12/2025] Open
Abstract
The diversity and heterogeneity of biomarkers has made the development of general methods for single-step quantification of analytes difficult. For individual biomarkers, electrochemical methods that detect a conformational change in an affinity binder upon analyte binding have shown promise. However, because the conformational change must operate within a nanometer-scale working distance, an entirely new sensor, with a unique conformational change, must be developed for each analyte. Here, we demonstrate a modular electrochemical biosensor, built from DNA origami, which is easily adapted to diverse molecules by merely replacing its analyte binding domains. Instead of relying on a unique nanometer-scale movement of a single redox reporter, all sensor variants rely on the same 100-nm scale conformational change, which brings dozens of reporters close enough to a gold electrode surface that a signal can be measured via square-wave voltammetry, a standard electrochemical technique. To validate our sensor's mechanism, we used single-stranded DNA as an analyte, and optimized the number of redox reporters and various linker lengths. Adaptation of the sensor to streptavidin and Platelet-Derived Growth Factor-BB (PDGF-BB) analytes was achieved by simply adding biotin or anti-PDGF aptamers to appropriate DNA linkers. Geometrically optimized streptavidin sensors exhibited signal gain and limit of detection markedly better than comparable reagentless electrochemical sensors. After use, the same sensors could be regenerated under mild conditions: Performance was largely maintained over four cycles of DNA strand displacement and rehybridization. By leveraging the modularity of DNA nanostructures, our work provides a straightforward route to the single-step quantification of arbitrary nucleic acids and proteins.
Collapse
Affiliation(s)
- Byoung-jin Jeon
- Department of Bioengineering, California Institute of Technology, Pasadena, CA91125
| | - Matteo M. Guareschi
- Department of Bioengineering, California Institute of Technology, Pasadena, CA91125
| | | | - Emily Wu
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Ashwin Gopinath
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | | | | | - Kevin W. Plaxco
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA93106
| | | | - Paul W. K. Rothemund
- Department of Bioengineering, California Institute of Technology, Pasadena, CA91125
| |
Collapse
|
17
|
Ullah A, Khan M, Zhang Y, Shafiq M, Ullah M, Abbas A, Xianxiang X, Chen G, Diao Y. Advancing Therapeutic Strategies with Polymeric Drug Conjugates for Nucleic Acid Delivery and Treatment. Int J Nanomedicine 2025; 20:25-52. [PMID: 39802382 PMCID: PMC11717654 DOI: 10.2147/ijn.s429279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
The effective clinical translation of messenger RNA (mRNA), small interfering RNA (siRNA), and microRNA (miRNA) for therapeutic purposes hinges on the development of efficient delivery systems. Key challenges include their susceptibility to degradation, limited cellular uptake, and inefficient intracellular release. Polymeric drug conjugates (PDCs) offer a promising solution, combining the benefits of polymeric carriers and therapeutic agents for targeted delivery and treatment. This comprehensive review explores the clinical translation of nucleic acid therapeutics, focusing on polymeric drug conjugates. It investigates how these conjugates address delivery obstacles, enhance systemic circulation, reduce immunogenicity, and provide controlled release, improving safety profiles. The review delves into the conjugation strategies, preparation methods, and various classes of PDCs, as well as strategic design, highlighting their role in nucleic acid delivery. Applications of PDCs in treating diseases such as cancer, immune disorders, and fibrosis are also discussed. Despite significant advancements, challenges in clinical adoption persist. The review concludes with insights into future directions for this transformative technology, underscoring the potential of PDCs to advance nucleic acid-based therapies and combat infectious diseases significantly.
Collapse
Affiliation(s)
- Aftab Ullah
- School of Medicine, Huaqiao University, Quanzhou, Fujian, People’s Republic of China
| | - Marina Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Kohat, Pakistan
| | - Yibang Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Muhammad Shafiq
- Research Institute of Clinical Pharmacy, Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
| | - Mohsan Ullah
- School of Medicine, Huaqiao University, Quanzhou, Fujian, People’s Republic of China
| | - Azar Abbas
- Institute of Medicine, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Xu Xianxiang
- School of Medicine, Huaqiao University, Quanzhou, Fujian, People’s Republic of China
| | - Gang Chen
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, Shandong, People’s Republic of China
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao central Medical Group), Qingdao, Shandong, People’s Republic of China
| | - Yong Diao
- School of Medicine, Huaqiao University, Quanzhou, Fujian, People’s Republic of China
| |
Collapse
|
18
|
Huang Y, Lin G, Liu S, Chen M, Yang C, Song Y. Aptamer-based Immune Checkpoint Inhibition for Cancer Immunotherapy. Chembiochem 2025; 26:e202400599. [PMID: 39417693 DOI: 10.1002/cbic.202400599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/19/2024]
Abstract
Cancer has long been a significant threat to human life and health. The advent of immune checkpoint blockade strategies has reversed cancer-induced immune suppression, advanced the development of immunotherapy, and offered new hope in the fight against cancer. Aptamers, which possess the same specificity and affinity as antibodies, are advantageous due to their synthetic accessibility and ease of modification, providing novel insights for immune checkpoint research. In this review, we outline the key aptamers currently developed for immune checkpoints such as CTLA-4, PD-1, PD-L1 and Siglec-15. We explore their potential in therapeutic strategies, including functionalizing or engineering aptamers for covalent binding, valency control, and nanostructure assembly, as well as investigating molecular mechanisms such as glycosylated protein functions and cell-cell interactions. Finally, the future applications of aptamers in immunotherapy are discussed.
Collapse
Affiliation(s)
- Yihao Huang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
| | - Guihong Lin
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
| | - Sinong Liu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
| | - Mingying Chen
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
- Renji Hospital, School of medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yanling Song
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
| |
Collapse
|
19
|
Escobar L, Sun D, Dhiman M, Hunter CA. Sequence-selective pulldown of recognition-encoded melamine oligomers using covalent capture on a solid support. Chem Commun (Camb) 2025; 61:504-507. [PMID: 39641167 PMCID: PMC11622007 DOI: 10.1039/d4cc06018k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
The covalent capture of recognition-encoded melamine oligomers (REMO) with a target attached to a solid support was investigated. Sequence-selective pulldown of complementary oligomers was observed when the target was challenged with a randomised library of oligomers. The approach provides an affinity selection method for the discovery of functional REMO sequences.
Collapse
Affiliation(s)
- Luis Escobar
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - Daniel Sun
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - Mohit Dhiman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - Christopher A Hunter
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| |
Collapse
|
20
|
Han Y, Zhang R, Bao H, Yang M, Gao Y, Gao X, Wang R, Tan W, Ji D. Molecular Programming Design of Glyconucleic Acid Aptamer with High Stability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408168. [PMID: 39630080 PMCID: PMC11775523 DOI: 10.1002/advs.202408168] [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: 07/17/2024] [Revised: 11/15/2024] [Indexed: 01/30/2025]
Abstract
Functional nucleic acids (FNAs), possessing specific biological functions beyond their informational roles, have gained widespread attention in disease therapeutics. However, their clinical application is severely limited by their low serum stability in complex physiological environments. In this work, a precise molecular programming strategy is explored to prepare glyconucleic acid aptamers (GNAAs) with high serum stability. Four glyconucleic acid modules compatible with commercial solid-phase synthesis are designed and synthesized. Through precise molecular design, the accurate modification of four different carbohydrate ligands at specific sites of DNA aptamers is achieved. It is demonstrated that glycosylation modification can significantly increase DNA aptamers' serum stability while maintaining their structures and high affinity. The stabilization effect is superior to that of currently commonly used commercial chemical modifications. Moreover, it is confirmed that this approach displays insignificant effects on the DNA aptamers' tumor-targeting ability and metabolism in vivo. This method offers a simple, economical, and efficient strategy for precise glycosylation modification of nucleic acids. This allows to prepare glycosyl functional nucleic acids with high serum stability, which can expand the application scope of functional nucleic acids and promote the practical transformation of functional nucleic acids.
Collapse
Affiliation(s)
- Yongqi Han
- Institute of Molecular Medicine (IMM)Renji HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200240China
- College of Chemistry and Materials ScienceShanghai Normal UniversityShanghai200234China
| | - Rongjun Zhang
- Institute of Molecular Medicine (IMM)Renji HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200240China
| | - Hong‐Liang Bao
- Institute of Molecular Medicine (IMM)Renji HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200240China
| | - Mei Yang
- Institute of Molecular Medicine (IMM)Renji HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200240China
| | - Yuan Gao
- Institute of Molecular Medicine (IMM)Renji HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200240China
| | - Xiaobo Gao
- Department of Anatomy and PhysiologyShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Ruowen Wang
- Institute of Molecular Medicine (IMM)Renji HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200240China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM)Renji HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200240China
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)The Chinese Academy of SciencesHangzhouZhejiang310022China
- Molecular Science and Biomedicine Laboratory (MBL)State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Chemistry and Chemical EngineeringCollege of BiologyAptamer Engineering Center of Hunan ProvinceHunan University ChangshaHunan410082China
| | - Ding‐Kun Ji
- Institute of Molecular Medicine (IMM)Renji HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200240China
| |
Collapse
|
21
|
Virgilio A, Benigno D, Aliberti C, Bello I, Panza E, Smimmo M, Vellecco V, Esposito V, Galeone A. Probing the Effects of Chemical Modifications on Anticoagulant and Antiproliferative Activity of Thrombin Binding Aptamer. Int J Mol Sci 2024; 26:134. [PMID: 39795992 PMCID: PMC11719963 DOI: 10.3390/ijms26010134] [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: 10/24/2024] [Revised: 11/27/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
Thrombin binding aptamer (TBA) is one of the best-known G-quadruplex (G4)-forming aptamers that efficiently binds to thrombin, resulting in anticoagulant effects. TBA also possesses promising antiproliferative properties. As with most therapeutic oligonucleotides, chemical modifications are critical for therapeutic applications, particularly to improve thermodynamic stability, resistance in biological environment, and target affinity. To evaluate the effects of nucleobase and/or sugar moiety chemical modifications, five TBA analogues have been designed and synthesized considering that the chair-like G4 structure is crucial for biological activity. Their structural and biological properties have been investigated by Circular Dichroism (CD), Nuclear Magnetic Resonance (NMR), native polyacrylamide gel electrophoresis (PAGE) techniques, and PT and MTT assays. The analogue TBAB contains 8-bromo-2'-deoxyguanosine (B) in G-syn glycosidic positions, while TBAL and TBAM contain locked nucleic acid guanosine (L) or 2'-O-methylguanosine (M) in G-anti positions, respectively. Instead, both the two types of modifications have been introduced in TBABL and TBABM with the aim of obtaining synergistic effects. In fact, both derivatives include B in syn positions, exhibiting in turn L and M in the anti ones. The most appealing results have been obtained for TBABM, which revealed an interesting cytotoxic activity against breast and prostate cancer cell lines, while in the case of TBAB, extraordinary thermal stability (Tm approximately 30 °C higher than that of TBA) and an anticoagulant activity higher than original aptamer were observed, as expected. These data indicate TBAB as the best TBA anticoagulant analogue here investigated and TBABM as a promising antiproliferative derivative.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Veronica Esposito
- Department of Pharmacy, University of Naples Federico II, 80131 Napoli, Italy; (A.V.); (D.B.); (C.A.); (I.B.); (E.P.); (M.S.); (V.V.); (A.G.)
| | | |
Collapse
|
22
|
Le AT, Teclemichael E, Krylova SM, Krylov SN. Quantitative Characterization of Partitioning Stringency in SELEX. JACS AU 2024; 4:4910-4920. [PMID: 39735910 PMCID: PMC11673186 DOI: 10.1021/jacsau.4c00890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/31/2024]
Abstract
Maintaining stringent conditions in SELEX (Systematic Evolution of Ligands by EXponential enrichment) is crucial for obtaining high-affinity aptamers. However, excessive stringency greatly increases the risk of SELEX failure. Controlling stringency has remained a technical challenge, largely dependent on intuition, due to the absence of a clear, quantitative measure of stringency. This study was motivated by our insight that, while stringency is influenced by multiple factors, it can be quantified by its effect: increasing stringency reduces the quantity of binders normalized to that of nonbinders after partitioning. Based on this insight, we propose measuring stringency using the binder-to-nonbinder ratio (BNR), where a lower BNR indicates higher stringency. We derive an experimental method for determining BNR via quantitative PCR. Our theoretical analysis and SELEX experiments using two distinct proteins as selection targets underscore the importance of maintaining a BNR significantly greater than zero to avoid failure, a principle we call the SELEX nonfailure criterion. By employing inverse BNR to quantify stringency and applying this criterion, researchers can more rationally control SELEX progress. The quantitative stringency measure and nonfailure criterion can also be applied to other artificial evolution methods, provided that selected binders are quantifiable.
Collapse
Affiliation(s)
- An T.
H. Le
- Department
of Chemistry, York University, 4700 Keele Street, Toronto, Ontario M3J 1P3, Canada
- Centre
for Research on Biomolecular Interactions, York University, 4700
Keele Street, Toronto, Ontario M3J 1P3, Canada
| | - Eden Teclemichael
- Department
of Chemistry, York University, 4700 Keele Street, Toronto, Ontario M3J 1P3, Canada
- Centre
for Research on Biomolecular Interactions, York University, 4700
Keele Street, Toronto, Ontario M3J 1P3, Canada
| | - Svetlana M. Krylova
- Department
of Chemistry, York University, 4700 Keele Street, Toronto, Ontario M3J 1P3, Canada
- Centre
for Research on Biomolecular Interactions, York University, 4700
Keele Street, Toronto, Ontario M3J 1P3, Canada
| | - Sergey N. Krylov
- Department
of Chemistry, York University, 4700 Keele Street, Toronto, Ontario M3J 1P3, Canada
- Centre
for Research on Biomolecular Interactions, York University, 4700
Keele Street, Toronto, Ontario M3J 1P3, Canada
| |
Collapse
|
23
|
Al Mazid MF, Shkel O, Ryu E, Kim J, Shin KH, Kim YK, Lim HS, Lee JS. Aptamer and N-Degron Ensemble (AptaGron) as a Target Protein Degradation Strategy. ACS Chem Biol 2024; 19:2462-2468. [PMID: 39630150 DOI: 10.1021/acschembio.4c00536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Target protein degradation (TPD) is a promising strategy for catalytic downregulation of target proteins through various cellular proteolytic pathways. Despite numerous reports on novel TPD mechanisms, the discovery of target-specific ligands remains a major challenge. Unlike small-molecule ligands, aptamers offer significant advantages, owing to their SELEX-based systematic screening method. To fully utilize aptamers for TPD, we designed an aptamer and N-degron ensemble system (AptaGron) that circumvents the need for synthetic conjugations between aptamers and proteolysis-recruiting units. In our AptaGron system, a peptide nucleic acid containing an N-degron peptide and a sequence complementary to the aptamer was designed. Using this system, we successfully degraded three target proteins, tau, nucleolin, and eukaryotic initiation factor 4E (eIF4E), which lack specific small-molecule ligands. Our results highlight the potential of the AptaGron approach as a robust platform for targeted protein degradation.
Collapse
Affiliation(s)
- Mohammad Faysal Al Mazid
- Division of Bio-Medical Science and Technology, Korea Institute of Science and Technology (KIST) & Department of Biological Chemistry, KIST School UST, Seoul 02792, South Korea
- Department of Pharmacology, College of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, South Korea
| | - Olha Shkel
- Brain Science Institute, Korea Institute of Science and Technology (KIST) & Department of Biological Chemistry, KIST School UST, Seoul 02792, South Korea
- Department of Pharmacology, College of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, South Korea
| | - Eunteg Ryu
- Department of Pharmacology, College of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, South Korea
| | - Jiwon Kim
- Department of Pharmacology, College of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, South Korea
| | - Kyung Ho Shin
- Department of Pharmacology, College of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, South Korea
| | - Yun Kyung Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST) & Department of Biological Chemistry, KIST School UST, Seoul 02792, South Korea
| | - Hyun Suk Lim
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, South Korea
| | - Jun-Seok Lee
- Department of Pharmacology, College of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, South Korea
| |
Collapse
|
24
|
Ramírez-Cortés F, Ménová P. Hepatocyte targeting via the asialoglycoprotein receptor. RSC Med Chem 2024:d4md00652f. [PMID: 39628900 PMCID: PMC11609720 DOI: 10.1039/d4md00652f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/19/2024] [Indexed: 12/06/2024] Open
Abstract
This review highlights the potential of asialoglycoprotein receptor (ASGPR)-mediated targeting in advancing liver-specific treatments and underscores the ongoing progress in the field. First, we provide a comprehensive examination of the nature of ASGPR ligands, both natural and synthetic. Next, we explore various drug delivery strategies leveraging ASGPR, with a particular emphasis on the delivery of therapeutic nucleic acids such as small interfering RNAs (siRNAs) and antisense oligonucleotides (ASOs). An in-depth analysis of the current status of RNA interference (RNAi) and ASO-based therapeutics is included, detailing approved therapies and those in various stages of clinical development (phases 1 to 3). Afterwards, we give an overview of other ASGPR-targeted conjugates, such as those with peptide nucleic acids or aptamers. Finally, targeted protein degradation of extracellular proteins through ASGPR is briefly discussed.
Collapse
Affiliation(s)
| | - Petra Ménová
- University of Chemistry and Technology, Prague Technická 5 16628 Prague 6 Czech Republic
| |
Collapse
|
25
|
Rana D, Rouwkema J. Spatiotemporally programmed release of aptamer tethered dual angiogenic growth factors. Int J Biol Macromol 2024; 283:137632. [PMID: 39547607 DOI: 10.1016/j.ijbiomac.2024.137632] [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: 08/30/2024] [Revised: 10/30/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
In tissue extracellular matrix (ECM), multiple growth factors (GFs) are sequestered through affinity interactions and released as needed by proteases, establishing spatial morphogen gradients in a time-controlled manner to guide cell behavior. Inspired by these ECM characteristics, we developed an "intelligent" biomaterial platform that spatially controls the combined bioavailability of multiple angiogenic GFs, specifically vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF-BB). Utilizing aptamer affinity interactions and complementary sequences within a GelMA matrix, our platform achieves on-demand, triggered release of individual GFs which can be "programmed" in temporally-controlled, repeatable cycles. The platform features stable incorporation of dual aptamers specific for both GFs, functional aptamer-CS molecular recognition in a 3D microenvironment with long-term stability of at least 15 days at physiological temperature, and spatially localized sequestration of individual GFs. Additionally, the system allows differential amounts of GFs to be released from the same hydrogels at different time-points, mimicking dynamic GF presentation in a 3D matrix similar to the native ECM. This flexible control over individual GF release kinetics opens new possibilities for dynamic GF presentation, with adjustable release profiles to meet the spatiotemporal needs of growing engineered tissue.
Collapse
Affiliation(s)
- Deepti Rana
- Department of Biomechanical Engineering, Technical Medical Centre, University of Twente, 7522NB Enschede, the Netherlands
| | - Jeroen Rouwkema
- Department of Biomechanical Engineering, Technical Medical Centre, University of Twente, 7522NB Enschede, the Netherlands.
| |
Collapse
|
26
|
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
|
27
|
Nguyen MD, Osborne MT, Prevot GT, Churcher ZR, Johnson PE, Simine L, Dauphin-Ducharme P. Truncations and in silico docking to enhance the analytical response of aptamer-based biosensors. Biosens Bioelectron 2024; 265:116680. [PMID: 39213817 DOI: 10.1016/j.bios.2024.116680] [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: 07/25/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
Aptamers are short oligonucleotides capable of binding specifically to various targets (i.e., small molecules, proteins, and whole cells) which have been introduced in biosensors such as in the electrochemical aptamer-based (E-AB) sensing platform. E-AB sensors are comprised of a redox-reporter-modified aptamer attached to an electrode that undergoes, upon target addition, a binding-induced change in electron transfer rates. To date, E-AB sensors have faced a limitation in the translatability of aptamers into the sensing platform presumably because sequences obtained from Systematic Evolution of Ligands by Exponential Enrichment (SELEX) are typically long (>80 nucleotides) and that obtaining structural information remains time and resource consuming. In response, we explore the utility of aptamer base truncations and in silico docking to improve their translatability into E-AB sensors. Here, we first apply this to the glucose aptamer, which we characterize in solution using NMR methods to guide design and translate truncated variants in E-AB biosensors. We further investigated the applicability of the truncation and computational approaches to four other aptamer systems (vancomycin, cocaine, methotrexate and theophylline) from which we derived functional E-AB sensors. We foresee that our strategy will increase the success rate of translating aptamers into sensing platforms to afford low-cost measurements of molecules directly in undiluted complex matrices.
Collapse
Affiliation(s)
- Minh-Dat Nguyen
- Département de chimie, Université de Sherbrooke, Sherbrooke, Québec, J1K 2R1, Canada
| | - Meghan T Osborne
- Department of Chemistry, York University, Toronto, Ontario, M3J 1P3, Canada
| | - Guy Terence Prevot
- Département de chimie, Université de Sherbrooke, Sherbrooke, Québec, J1K 2R1, Canada
| | - Zachary R Churcher
- Department of Chemistry, York University, Toronto, Ontario, M3J 1P3, Canada
| | - Philip E Johnson
- Department of Chemistry, York University, Toronto, Ontario, M3J 1P3, Canada
| | - Lena Simine
- Department of Chemistry, McGill University, Montreal, Quebec, H3A 0B8, Canada
| | | |
Collapse
|
28
|
Zhou Z, Zhang R, Zhou A, Lv J, Chen S, Zou H, Zhang G, Lin T, Wang Z, Zhang Y, Weng S, Han X, Liu Z. Proteomics appending a complementary dimension to precision oncotherapy. Comput Struct Biotechnol J 2024; 23:1725-1739. [PMID: 38689716 PMCID: PMC11058087 DOI: 10.1016/j.csbj.2024.04.044] [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: 02/06/2024] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 05/02/2024] Open
Abstract
Recent advances in high-throughput proteomic profiling technologies have facilitated the precise quantification of numerous proteins across multiple specimens concurrently. Researchers have the opportunity to comprehensively analyze the molecular signatures in plentiful medical specimens or disease pattern cell lines. Along with advances in data analysis and integration, proteomics data could be efficiently consolidated and employed to recognize precise elementary molecular mechanisms and decode individual biomarkers, guiding the precision treatment of tumors. Herein, we review a broad array of proteomics technologies and the progress and methods for the integration of proteomics data and further discuss how to better merge proteomics in precision medicine and clinical settings.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Henan 450052, China
| | - Ruiqi Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Aoyang Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jinxiang Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Shuang Chen
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Haijiao Zou
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ting Lin
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhan Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Henan 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
29
|
Sandeep, Subba R, Mondal AC. Does COVID-19 Trigger the Risk for the Development of Parkinson's Disease? Therapeutic Potential of Vitamin C. Mol Neurobiol 2024; 61:9945-9960. [PMID: 37957424 DOI: 10.1007/s12035-023-03756-3] [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: 02/20/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), which was proclaimed a pandemic by the World Health Organization (WHO) in March 2020. There is mounting evidence that older patients with multimorbidity are more susceptible to COVID-19 complications than are younger, healthy people. Having neuroinvasive potential, SARS-CoV-2 infection may increase susceptibility toward the development of Parkinson's disease (PD), a progressive neurodegenerative disorder with extensive motor deficits. PD is characterized by the aggregation of α-synuclein in the form of Lewy bodies and the loss of dopaminergic neurons in the dorsal striatum and substantia nigra pars compacta (SNpc) of the nigrostriatal pathway in the brain. Increasing reports suggest that SARS-CoV-2 infection is linked with the worsening of motor and non-motor symptoms with high rates of hospitalization and mortality in PD patients. Common pathological changes in both diseases involve oxidative stress, mitochondrial dysfunction, neuroinflammation, and neurodegeneration. COVID-19 exacerbates the damage ensuing from the dysregulation of those processes, furthering neurological complications, and increasing the severity of PD symptomatology. Phytochemicals have antioxidant, anti-inflammatory, and anti-apoptotic properties. Vitamin C supplementation is found to ameliorate the common pathological changes in both diseases to some extent. This review aims to present the available evidence on the association between COVID-19 and PD, and discusses the therapeutic potential of vitamin C for its better management.
Collapse
Affiliation(s)
- Sandeep
- Laboratory of Cellular & Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Rhea Subba
- Laboratory of Cellular & Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Amal Chandra Mondal
- Laboratory of Cellular & Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
30
|
Albada B. Functionalized DNA secondary structures and nanostructures for specific protein modifications. Trends Biochem Sci 2024; 49:1124-1135. [PMID: 39443210 DOI: 10.1016/j.tibs.2024.09.003] [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: 06/27/2024] [Revised: 09/03/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024]
Abstract
The development of non-biological applications of DNA has not only resulted in delicately shaped DNA-based nano-objects with complex functions but also spawned their use for novel catalytic applications. From the multitude of applications of DNAzymes that operate on a relatively simple substrate, we have witnessed the emergence of multifunctional catalytically active DNA-based nanostructures for one of the most challenging tasks known to a chemist: the controlled and precise modification of a wild-type protein in its natural environment. By incorporating various elements associated with post-translational modification (PTM) writer enzymes into complex nanostructures, it is now possible to chemically modify a specific protein in cell lysates under the influence of an externally added trigger, clearly illustrating the promising future for this approach.
Collapse
Affiliation(s)
- Bauke Albada
- Laboratory of Organic Chemistry, Wageningen University and Research, Stippeneng 4, 6708, WE, Wageningen, The Netherlands.
| |
Collapse
|
31
|
Makela M, Tu D, Lin Z, Coté G, Lin PT. Chip-Scale Aptamer Sandwich Assay Using Optical Waveguide-Assisted Surface-Enhanced Raman Spectroscopy. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1927. [PMID: 39683314 DOI: 10.3390/nano14231927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/24/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024]
Abstract
Chip-scale optical waveguide-assisted surface-enhanced Raman spectroscopy (SERS) that used nanoparticles (NPs) was demonstrated. The Raman signals from Raman reporter (RR) molecules on NPs can be efficiently excited by the waveguide evanescent field when the molecules are in proximity to the waveguide surface. The Raman signal was enhanced by plasmon resonance due to the NPs close to the waveguide surface. The optical waveguide mode and the NP-induced field enhancement were calculated using a finite difference method (FDM). The sensing performance of the waveguide-assisted SERS device was experimentally characterized by measuring the Raman scattering from various RRs, including 4-mercaptobenzoic acid (4-MBA), 5,5'-dithio-bis-(2-nitrobenzoic acid) (DTNB), and malachite green isothiocyanate (MGITC). The observed Raman spectral features were identified and assigned to the complex vibrational modes associated with different reporters. A low detection limit of 1 nM was achieved. In addition, the device sensing method was applied to the detection of the biomarker cardiac troponin I (cTnI) using an aptamer sandwich assay immobilized on the device surface. Overall, the optical waveguides integrated with SERS show a miniaturized sensing platform for the detection of small molecules and large proteins, potentially enabling multiplexed detection for clinically relevant applications.
Collapse
Affiliation(s)
- Megan Makela
- Department of Materials Science and Engineering, Texas A&M University, College Station, TX 77843, USA
- Center for Remote Health Systems and Technologies, Texas A&M University, College Station, TX 77843, USA
| | - Dandan Tu
- Center for Remote Health Systems and Technologies, Texas A&M University, College Station, TX 77843, USA
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Zhihai Lin
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Gerard Coté
- Center for Remote Health Systems and Technologies, Texas A&M University, College Station, TX 77843, USA
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Pao Tai Lin
- Department of Materials Science and Engineering, Texas A&M University, College Station, TX 77843, USA
- Center for Remote Health Systems and Technologies, Texas A&M University, College Station, TX 77843, USA
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
32
|
Wang Z, Wang X, He Y, Wu H, Mao R, Wang H, Qiu L. Exploring Framework Nucleic Acids: A Perspective on Their Cellular Applications. JACS AU 2024; 4:4110-4128. [PMID: 39610738 PMCID: PMC11600171 DOI: 10.1021/jacsau.4c00776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/30/2024]
Abstract
Cells are fundamental units of life. The coordination of cellular functions and behaviors relies on a cascade of molecular networks. Technologies that enable exploration and manipulation of specific molecular events in living cells with high spatiotemporal precision would be critical for pathological study, disease diagnosis, and treatment. Framework nucleic acids (FNAs) represent a novel class of nucleic acid materials characterized by their monodisperse and rigid nanostructure. Leveraging their exceptional programmability, convenient modification property, and predictable atomic-level architecture, FNAs have attracted significant attention in diverse cellular applications such as cell recognition, imaging, manipulation, and therapeutic interventions. In this perspective, we will discuss the utilization of FNAs in living cell systems while critically assessing the opportunities and challenges presented in this burgeoning field.
Collapse
Affiliation(s)
- Zhaoyang Wang
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, China
| | - Xin Wang
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, 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
| | - Yao He
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, China
| | - Hui Wu
- 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
| | - Rui Mao
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, China
| | - Haiyuan Wang
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, China
| | - Liping Qiu
- Molecular
Science and Biomedicine Laboratory (MBL), State Key Laboratory of
Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Aptamer Engineering Center of Hunan
Province, Hunan University, Changsha, Hunan 410082, 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
| |
Collapse
|
33
|
Shin S, Jo H, Agura T, Jeong S, Ahn H, Pang S, Lee J, Park JH, Kim Y, Kang JS. Anti-Inflammatory Effects of Aptamin C in Pulmonary Fibrosis Induced by Bleomycin. Pharmaceuticals (Basel) 2024; 17:1577. [PMID: 39770419 PMCID: PMC11676684 DOI: 10.3390/ph17121577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/17/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Vitamin C is a well-known antioxidant with antiviral, anticancer, and anti-inflammatory properties. However, its therapeutic applications are limited by rapid oxidation due to heat and light sensitivity. Aptamin C, which employs aptamers to bind vitamin C, has demonstrated enhanced stability and efficacy. This study investigates the potential of Aptamin C to inhibit the progression of pulmonary fibrosis, a prominent inflammatory lung disease with no effective treatment. Methods: Mice bearing bleomycin-induced pulmonary fibrosis were administered vitamin C or Aptamin C, and their weight changes and survival rates were monitored. Inflammatory cell infiltration was assessed in the bronchoalveolar lavage fluid (BALF), and the degree of alveolar fibrosis was measured by H&E and Masson's trichrome staining. To elucidate the mechanism of action of Aptamin C, Western blot analysis was performed in HaCaT and lung tissues from bleomycin-induced pulmonary fibrosis mice. Results: The Aptamin C-treated group showed a notably higher survival rate at 50%, whereas all subjects in the vitamin C-treated group died. Histological examination of lung tissue showed that inflammation was significantly suppressed in the Aptamin C-supplemented group compared to the vitamin C-supplemented group, with a 10% greater reduction in cell infiltrations, along with noticeably less tissue damage. Additionally, it was observed that Aptamin C increased SVCT-1 expression in the HaCaT cells and the lung tissues. Conclusions: Taken together, Aptamin C not only increases the stability of vitamin C but also induces an increase in SVCT-1 expression, facilitating greater vitamin C absorption into cells and tissues, thereby inhibiting the progression of symptoms and associated inflammatory responses in pulmonary fibrosis.
Collapse
Affiliation(s)
- Seulgi Shin
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (S.S.); (H.J.); (T.A.); (S.J.); (H.A.); (S.P.); (Y.K.)
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 08826, Republic of Korea
- Department of Research and Development, N Therapeutics Co., Ltd., Seoul 08813, Republic of Korea
| | - Hyejung Jo
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (S.S.); (H.J.); (T.A.); (S.J.); (H.A.); (S.P.); (Y.K.)
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 08826, Republic of Korea
| | - Tomoyo Agura
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (S.S.); (H.J.); (T.A.); (S.J.); (H.A.); (S.P.); (Y.K.)
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 08826, Republic of Korea
| | - Seoyoun Jeong
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (S.S.); (H.J.); (T.A.); (S.J.); (H.A.); (S.P.); (Y.K.)
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 08826, Republic of Korea
| | - Hyovin Ahn
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (S.S.); (H.J.); (T.A.); (S.J.); (H.A.); (S.P.); (Y.K.)
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 08826, Republic of Korea
| | - Soyoung Pang
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (S.S.); (H.J.); (T.A.); (S.J.); (H.A.); (S.P.); (Y.K.)
| | - June Lee
- Nexmos, Inc., Yongin-si 168267, Republic of Korea; (J.L.); (J.-H.P.)
| | - Jeong-Ho Park
- Nexmos, Inc., Yongin-si 168267, Republic of Korea; (J.L.); (J.-H.P.)
| | - Yejin Kim
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (S.S.); (H.J.); (T.A.); (S.J.); (H.A.); (S.P.); (Y.K.)
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 08826, Republic of Korea
| | - Jae Seung Kang
- Laboratory of Vitamin C and Antioxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (S.S.); (H.J.); (T.A.); (S.J.); (H.A.); (S.P.); (Y.K.)
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 08826, Republic of Korea
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
34
|
Thomas BJ, Awan SZ, Joshi T, Daniels MA, Porciani D, Burke DH. Anti-EGFR aptamer exhibits direct anti-cancer effects in NSCLC cells harboring EGFR L858R mutations. NPJ Precis Oncol 2024; 8:271. [PMID: 39572699 PMCID: PMC11582725 DOI: 10.1038/s41698-024-00758-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) adenocarcinoma (LUAD) is a leading cause of death worldwide. Activating mutations in the tyrosine kinase domain of the oncogene epidermal growth factor receptor (EGFR) are responsible for ~10-50% of all LUAD cases. Although tyrosine kinase inhibitors (TKIs) have been effective in prolonging patient survival and quality of life, acquired resistance and disease progression are inevitable, presenting a clear unmet need for alternative or adjuvant therapeutics. Here we show that an anti-EGFR aptamer (EGFRapt) decreases viability and tumor growth of LUAD cell lines harboring the L858R ± T790M mutation in EGFR. Additionally, we elucidate the mechanism by which EGFRapt exerts these effects by monitoring cellular processes associated with kinase-dependent and kinase-independent mechanisms. Overall, these data establish that EGFRapt has direct anti-cancer activity in mutant EGFR positive LUAD via targetable mechanisms that are independent of existing approaches, and they provide a foundation for further development of nucleic acid-based therapies that target EGFR.
Collapse
Affiliation(s)
- Brian J Thomas
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, USA
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Sania Z Awan
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO, USA
| | - Trupti Joshi
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO, USA
- Department of Biomedical Informatics, Biostatistics and Medical Epidemiology (BBME), University of Missouri School of Medicine, Columbia, MO, USA
| | - Mark A Daniels
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, USA
- NextGen Precision Health, University of Missouri, Columbia, MO, USA
| | - David Porciani
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, USA.
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.
- Standard BioTools Inc./SomaLogic Inc., Boulder, CO, USA.
| | - Donald H Burke
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO, USA.
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.
- Department of Biochemistry, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
35
|
Goldbloom-Helzner L, Bains H, Loll EG, Henson T, Mizenko RR, Kumar P, Tan C, Farmer DL, Carney RP, Wang A. Assessing the conjugation efficiency of surface-modified extracellular vesicles using single nanovesicle analysis technologies. NANOSCALE 2024; 16:20903-20916. [PMID: 39310954 PMCID: PMC11748815 DOI: 10.1039/d4nr01603c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Extracellular vesicles (EVs) are cell-secreted nanoscale vesicles with important roles in cell-cell communication and drug delivery. Although EVs pose a promising alternative to cell-based therapy, targeted delivery in vivo is lacking. Their surface is often modified to endow them with active targeting molecules to enable specific cell uptake and tailor EV biodistribution. A dominant paradigm has been to evaluate the EV surface functionalization using bulk analysis assays, such as western blotting and bead-based flow cytometry. Yet, the heterogeneity of EVs is now recognized as a major bottleneck for their clinical translation. Here, we engineer the EV surface at the single-vesicle level. We applied orthogonal platforms with single vesicle resolution to determine and optimize the efficiency of conjugating the myelin-targeting aptamer LJM-3064 to single EVs (Apt-EVs). The aptamers were conjugated using either lipid insertion or covalent protein modification, followed by an assessment of single-EV integrity and stability. We observed unique aptamer conjugation to single EVs that depends on EV size. Our study underscores the importance of single vesicle analysis for engineering EVs and provides a novel single-EV-based framework for modifying EV surfaces.
Collapse
Affiliation(s)
- Leora Goldbloom-Helzner
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA, 95817, USA.
- Institute for Pediatric Regenerative Medicine, Shriners Children's, Sacramento, CA, 95817, USA
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, 95616, USA
| | - Harjn Bains
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, 95616, USA
| | - Emma G Loll
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA, 95817, USA.
- Institute for Pediatric Regenerative Medicine, Shriners Children's, Sacramento, CA, 95817, USA
| | - Tanner Henson
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA, 95817, USA.
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, 95616, USA
| | - Rachel R Mizenko
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, 95616, USA
| | - Priyadarsini Kumar
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA, 95817, USA.
- Institute for Pediatric Regenerative Medicine, Shriners Children's, Sacramento, CA, 95817, USA
| | - Cheemeng Tan
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, 95616, USA
| | - Diana L Farmer
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA, 95817, USA.
- Institute for Pediatric Regenerative Medicine, Shriners Children's, Sacramento, CA, 95817, USA
| | - Randy P Carney
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, 95616, USA
| | - Aijun Wang
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA, 95817, USA.
- Institute for Pediatric Regenerative Medicine, Shriners Children's, Sacramento, CA, 95817, USA
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, 95616, USA
| |
Collapse
|
36
|
Shan X, Cai Y, Zhu B, Zhou L, Sun X, Xu X, Yin Q, Wang D, Li Y. Rational strategies for improving the efficiency of design and discovery of nanomedicines. Nat Commun 2024; 15:9990. [PMID: 39557860 PMCID: PMC11574076 DOI: 10.1038/s41467-024-54265-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 11/06/2024] [Indexed: 11/20/2024] Open
Abstract
The rise of rational strategies in nanomedicine development, such as high-throughput methods and computer-aided techniques, has led to a shift in the design and discovery patterns of nanomedicines from a trial-and-error mode to a rational mode. This transition facilitates the enhancement of efficiency in the preclinical discovery pipeline of nanomaterials, particularly in improving the hit rate of nanomaterials and the optimization efficiency of promising candidates. Herein, we describe a directed evolution mode of nanomedicines driven by data to accelerate the discovery of nanomaterials with high delivery efficiency. Computer-aided design strategies are introduced in detail as one of the cutting-edge directions for the development of nanomedicines. Ultimately, we look forward to expanding the tools for the rational design and discovery of nanomaterials using multidisciplinary approaches. Rational design strategies may potentially boost the delivery efficiency of next-generation nanomedicines.
Collapse
Affiliation(s)
- Xiaoting Shan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Ying Cai
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong, 264000, China
| | - Binyu Zhu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Lingli Zhou
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xujie Sun
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Xiaoxuan Xu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Qi Yin
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Dangge Wang
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201260, China.
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China.
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, Shandong, 264000, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China.
| |
Collapse
|
37
|
Yim J, Park J, Kim G, Lee HH, Chung JS, Jo A, Koh M, Park J. Conditional PROTAC: Recent Strategies for Modulating Targeted Protein Degradation. ChemMedChem 2024; 19:e202400326. [PMID: 38993102 PMCID: PMC11581424 DOI: 10.1002/cmdc.202400326] [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: 04/30/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024]
Abstract
Proteolysis-targeting chimeras (PROTACs) have emerged as a promising technology for inducing targeted protein degradation by leveraging the intrinsic ubiquitin-proteasome system (UPS). While the potential druggability of PROTACs toward undruggable proteins has accelerated their rapid development and the wide-range of applications across diverse disease contexts, off-tissue effects and side-effects of PROTACs have recently received attentions to improve their efficacy. To address these issues, spatial or temporal target protein degradation by PROTACs has been spotlighted. In this review, we explore chemical strategies for modulating protein degradation in a cell type-specific (spatio-) and time-specific (temporal-) manner, thereby offering insights for expanding PROTAC applications to overcome the current limitations of target protein degradation strategy.
Collapse
Affiliation(s)
- Junhyeong Yim
- Department of ChemistryKangwon National UniversityChuncheon24341Republic of Korea
- Multidimensional Genomics Research CenterKangwon National UniversityChuncheon24341Republic of Korea
| | - Junyoung Park
- Department of ChemistryKangwon National UniversityChuncheon24341Republic of Korea
- Institute for Molecular Science and Fusion TechnologyKangwon National UniversityChuncheon24341Republic of Korea
| | - Gabin Kim
- Department of Chemistry and ChemistryInstitute for Functional MaterialsPusan National UniversityBusan46241Republic of Korea
| | - Hyung Ho Lee
- Department of UrologyUrological Cancer CenterResearch Institute and Hospital of National Cancer CenterGoyang10408Republic of Korea
| | - Jin Soo Chung
- Department of UrologyUrological Cancer CenterResearch Institute and Hospital of National Cancer CenterGoyang10408Republic of Korea
| | - Ala Jo
- Center for NanomedicineInstitute for Basic ScienceSeoul03722Republic of Korea
| | - Minseob Koh
- Department of Chemistry and ChemistryInstitute for Functional MaterialsPusan National UniversityBusan46241Republic of Korea
| | - Jongmin Park
- Department of ChemistryKangwon National UniversityChuncheon24341Republic of Korea
- Multidimensional Genomics Research CenterKangwon National UniversityChuncheon24341Republic of Korea
- Institute for Molecular Science and Fusion TechnologyKangwon National UniversityChuncheon24341Republic of Korea
| |
Collapse
|
38
|
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
|
39
|
Xue L, Yu X, Zhao L, Garrett A, Wu D, Liu HY. Targeted Delivery of AR-V7 siRNA with Bivalent PSMA Aptamers Effectively Suppresses the Growth of Enzalutamide-Resistant Prostate Cancer. Mol Pharm 2024; 21:5749-5760. [PMID: 39388218 DOI: 10.1021/acs.molpharmaceut.4c00743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Androgen deprivation therapy has been the primary treatment strategy for advanced prostate cancer (PCa). But most patients develop castration resistance over time. For FDA-approved second-generation androgen receptor (AR) antagonists, including enzalutamide (ENZ) and abiraterone (AA), patients who initially respond to them eventually develop resistance. The key mechanism for resistance to ENZ/AA involves AR splice variants (AR-Vs) and specifically AR-V7. Current AR antagonists cannot target AR-V7 due to its lack of the C-terminal ligand-binding domain (LBD) but keeping the AR N-terminal domain (NTD) which still can activate androgen-responsive genes. Therefore, targeting the AR NTD and AR-V7 is critically important to overcome ENZ resistance. Unfortunately, AR NTD has been considered an "undruggable" target due to the difficulty in defining its three-dimensional (3D) structure. In this context, siRNA is highly suitable to address this undruggable target. However, siRNA cannot freely diffuse into cells, and a carrier is needed. In this regard, nucleic acid-based aptamers are highly suitable for cell type-specific delivery of siRNA in vivo. In this study, we have developed a serum-stable bivalent prostate-specific membrane antigen (PSMA) aptamer-AR-V7 siRNA chimera (PAP). The results show that PAP can knock down both AR-full length and AR-V7 in PSMA-expressing castration-resistant cells. It can resensitize ENZ in cell lines and PCa xenografts. ENZ combined with PAP can significantly inhibit 22Rv1 xenograft growth in mice without experiencing castration. Owing to the low toxicity, PAP has potential to offer a new antiandrogen treatment for current ENZ-resistant PCa.
Collapse
MESH Headings
- Male
- Phenylthiohydantoin
- Humans
- Animals
- Benzamides
- Nitriles
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Mice
- RNA, Small Interfering/administration & dosage
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Glutamate Carboxypeptidase II/antagonists & inhibitors
- Glutamate Carboxypeptidase II/genetics
- Glutamate Carboxypeptidase II/metabolism
- Xenograft Model Antitumor Assays
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/pathology
- Aptamers, Nucleotide
- Antigens, Surface/genetics
- Antigens, Surface/metabolism
- Mice, Nude
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/pathology
- Androgen Receptor Antagonists/pharmacology
- Cell Proliferation/drug effects
Collapse
Affiliation(s)
- Lu Xue
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912, United States
- Department of Pediatrics Hematology, The First Hospital of Jilin University, Changchun 130021, China
| | - Xiaolin Yu
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912, United States
| | - Lijing Zhao
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912, United States
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun 130021, China
| | - Aria Garrett
- Dotquant, CoMotion at University of Washington, Seattle, Washington 98195, United States
| | - Daqing Wu
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912, United States
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, Georgia 30314, United States
| | - Hong Yan Liu
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912, United States
- Dotquant, CoMotion at University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
40
|
Wong F, He D, Krishnan A, Hong L, Wang AZ, Wang J, Hu Z, Omori S, Li A, Rao J, Yu Q, Jin W, Zhang T, Ilia K, Chen JX, Zheng S, King I, Li Y, Collins JJ. Deep generative design of RNA aptamers using structural predictions. NATURE COMPUTATIONAL SCIENCE 2024; 4:829-839. [PMID: 39506080 DOI: 10.1038/s43588-024-00720-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024]
Abstract
RNAs represent a class of programmable biomolecules capable of performing diverse biological functions. Recent studies have developed accurate RNA three-dimensional structure prediction methods, which may enable new RNAs to be designed in a structure-guided manner. Here, we develop a structure-to-sequence deep learning platform for the de novo generative design of RNA aptamers. We show that our approach can design RNA aptamers that are predicted to be structurally similar, yet sequence dissimilar, to known light-up aptamers that fluoresce in the presence of small molecules. We experimentally validate several generated RNA aptamers to have fluorescent activity, show that these aptamers can be optimized for activity in silico, and find that they exhibit a mechanism of fluorescence similar to that of known light-up aptamers. Our results demonstrate how structural predictions can guide the targeted and resource-efficient design of new RNA sequences.
Collapse
Affiliation(s)
- Felix Wong
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering & Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Integrated Biosciences, Redwood City, CA, USA
| | - Dongchen He
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
- Shanghai Artificial Intelligence Laboratory, Shanghai, China
| | - Aarti Krishnan
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering & Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Liang Hong
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Alexander Z Wang
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering & Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jiuming Wang
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhihang Hu
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Satotaka Omori
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Integrated Biosciences, Redwood City, CA, USA
| | - Alicia Li
- Integrated Biosciences, Redwood City, CA, USA
| | - Jiahua Rao
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, China
| | - Qinze Yu
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wengong Jin
- Eric and Wendy Schmidt Center, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, USA
| | - Tianqing Zhang
- Institute for Medical Engineering & Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Katherine Ilia
- Institute for Medical Engineering & Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jack X Chen
- Institute for Medical Engineering & Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shuangjia Zheng
- Global Institute of Future Technology, Shanghai Jiao Tong University, Shanghai, China
| | - Irwin King
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yu Li
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Institute for Medical Engineering & Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China.
- The CUHK Shenzhen Research Institute, Shenzhen, China.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| | - James J Collins
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Institute for Medical Engineering & Science and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| |
Collapse
|
41
|
Moeinfard T, Ghafar-Zadeh E, Magierowski S. CMOS Point-of-Care Diagnostics Technologies: Recent Advances and Future Prospects. MICROMACHINES 2024; 15:1320. [PMID: 39597132 PMCID: PMC11596111 DOI: 10.3390/mi15111320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024]
Abstract
This review provides a comprehensive overview of point-of-care (PoC) devices across several key diagnostic applications, including blood analysis, infectious disease detection, neural interfaces, and commercialized integrated circuits (ICs). In the blood analysis section, the focus is on biomarkers such as glucose, dopamine, and aptamers, and their respective detection techniques. The infectious disease section explores PoC technologies for detecting pathogens, RNA, and DNA, highlighting innovations in molecular diagnostics. The neural interface section reviews advancements in neural recording and stimulation for therapeutic applications. Finally, a survey of commercialized ICs from companies such as Abbott and Medtronic is presented, showcasing existing PoC devices already in widespread clinical use. This review emphasizes the role of complementary metal-oxide-semiconductor (CMOS) technology in enabling compact, efficient diagnostic systems and offers insights into the current and future landscape of PoC devices.
Collapse
Affiliation(s)
- Tania Moeinfard
- Department of Electrical Engineering and Computer Science, Lassonde School of Engineering, York University, Toronto, ON M3J 1P3, Canada; (T.M.); (S.M.)
- Biologically Inspired Sensors and Actuators (BioSA) Laboratory, York University, Toronto, ON M3J 1P3, Canada
- Electronic Machine Intelligence Lab, York University, Toronto, ON M3J 1P3, Canada
| | - Ebrahim Ghafar-Zadeh
- Department of Electrical Engineering and Computer Science, Lassonde School of Engineering, York University, Toronto, ON M3J 1P3, Canada; (T.M.); (S.M.)
- Biologically Inspired Sensors and Actuators (BioSA) Laboratory, York University, Toronto, ON M3J 1P3, Canada
| | - Sebastian Magierowski
- Department of Electrical Engineering and Computer Science, Lassonde School of Engineering, York University, Toronto, ON M3J 1P3, Canada; (T.M.); (S.M.)
- Electronic Machine Intelligence Lab, York University, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
42
|
Driscoll J, Gondaliya P, Ziemer A, Yan IK, Gupta Y, Patel T. In Silico Design of Novel EpCAM-Binding Aptamers for Targeted Delivery of RNA Therapeutics. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1727. [PMID: 39513807 PMCID: PMC11548041 DOI: 10.3390/nano14211727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/26/2024] [Accepted: 10/27/2024] [Indexed: 11/15/2024]
Abstract
Aptamers are short DNA or RNA sequences that adopt 3D structures and can bind to protein targets with high binding affinity and specificity. Aptamers exhibit excellent tissue penetration, are inexpensive to produce, and can be internalized by cells. Therefore, aptamers are attractive targeting ligands to direct the delivery of theranostic agents to the desired cells. Epithelial cell adhesion molecule (EpCAM) is a tumor-associated antigen that is aberrantly overexpressed on many epithelial-derived cancers, including on cholangiocarcinoma (CCA) cells. Its expression on treatment-resistant cancer stem cells, along with its abundance in the CCA tumor microenvironment, highlights the need to develop EpCAM-targeted therapies for CCA. Herein, an in silico approach was used to design and screen DNA aptamers capable of binding to the EpCAM monomer and homodimer. Two aptamers, PLD01 and PLD02, met the selection criteria and were validated in vitro. Both aptamers exhibited high affinity for EpCAM+ CCA cells, with negligible binding to EpCAM- leukemia cells. Modified versions of PLD01 and PLD02 were successfully incorporated into the membranes of milk-derived nanovesicles. PLD01-functionalized nanovesicles enabled EpCAM-targeted delivery of the therapeutic cargo to CCA cells. In summary, these EpCAM-targeting aptamers can be utilized to direct the delivery of theranostic agents to EpCAM-expressing cells.
Collapse
Affiliation(s)
- Julia Driscoll
- Department of Transplantation, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224, USA (I.K.Y.)
| | - Piyush Gondaliya
- Department of Transplantation, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224, USA (I.K.Y.)
| | - Abbye Ziemer
- Department of Transplantation, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224, USA (I.K.Y.)
| | - Irene K. Yan
- Department of Transplantation, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224, USA (I.K.Y.)
| | - Yash Gupta
- Division of Infectious Diseases, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Tushar Patel
- Department of Transplantation, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL 32224, USA (I.K.Y.)
| |
Collapse
|
43
|
Mottard K, Cokaiko J, Rogister B, Neirinckx V. Therapeutic targeting of the protein tyrosine kinase-7 in cancer: an overview. Oncologist 2024:oyae290. [PMID: 39468753 DOI: 10.1093/oncolo/oyae290] [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: 05/14/2024] [Accepted: 09/10/2024] [Indexed: 10/30/2024] Open
Abstract
The protein tyrosine kinase-7 (PTK7) is an evolutionarily conserved transmembrane receptor that has emerged as a potential therapeutic target for human tumors. PTK7 is a pseudokinase that is involved in the modulation of the Wnt signaling pathway through interactions with other receptors. These interactions result in targeted gene activation that regulates cell polarity, migration, and proliferation during embryogenesis. Aside of this role during development, PTK7 has been shown as overexpressed in numerous cancers including colon carcinoma, leukemia, neuroblastoma, hepatoma, and ovarian cancer. The activity of PTK7 and the direct correlation with poor prognosis have fostered preclinical investigations and phase I clinical trials, aiming at inhibiting PTK7 and inducing antitumoral effects. In this review, we provide an exhaustive overview of the diverse approaches that use PTK7 as a new molecular target for cancer therapy in different tumor types. We discuss current therapies and future strategies including chimeric antigen receptor-T cells, antibody-drug conjugates, aptamers, based on up-to-date literature and ongoing research progress.
Collapse
Affiliation(s)
- Kim Mottard
- Laboratory of Nervous System Diseases and Therapy, GIGA Institute, University of Liège, 4000 Liège, Belgium
| | - Julie Cokaiko
- Laboratory of Nervous System Diseases and Therapy, GIGA Institute, University of Liège, 4000 Liège, Belgium
| | - Bernard Rogister
- Laboratory of Nervous System Diseases and Therapy, GIGA Institute, University of Liège, 4000 Liège, Belgium
- Neurology Department, University Hospital, University of Liège, 4000 Liège, Belgium
| | - Virginie Neirinckx
- Laboratory of Nervous System Diseases and Therapy, GIGA Institute, University of Liège, 4000 Liège, Belgium
| |
Collapse
|
44
|
Kalaimani K, Balachandran S, Boopathy LK, Roy A, Jayachandran B, Sankaranarayanan S, Arumugam MK. Recent advancements in small interfering RNA based therapeutic approach on breast cancer. Eur J Pharmacol 2024; 981:176877. [PMID: 39128807 DOI: 10.1016/j.ejphar.2024.176877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 07/23/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Breast cancer (BC) is the most common and malignant tumor diagnosed in women, with 2.9 million cases in 2023 and the fifth highest cancer-causing mortality worldwide. Recent developments in targeted therapy options for BC have demonstrated the promising potential of small interfering RNA (siRNA)-based cancer therapeutic approaches. As BC continues to be a global burden, siRNA therapy emerges as a potential treatment strategy to regulate disease-related genes in other types of cancers, including BC. siRNAs are tiny RNA molecules that, by preventing their expression, can specifically silence genes linked to the development of cancer. In order to increase the stability and effectiveness of siRNA delivery to BC cells, minimize off-target effects, and improve treatment efficacy, advanced delivery technologies such as lipid nanoparticles and nanocarriers have been created. Additionally, combination therapies, such as siRNAs that target multiple pathways are used in conjunction with conventional chemotherapy agents, have shown synergistic effects in various preclinical studies, opening up new treatment options for breast cancer that are personalized and precision medicine-oriented. Targeting important genes linked to BC growth, metastasis, and chemo-resistance has been reported in BC research using siRNA-based therapies. This study reviews recent reports on therapeutic approaches to siRNA for advanced treatment of BC. Furthermore, this review evaluates the role and mechanisms of siRNA in BC and demonstrates the potential of exploiting siRNA as a novel target for BC therapy.
Collapse
Affiliation(s)
- Kathirvel Kalaimani
- Cancer Biology Lab, Centre for Molecular and Nanomedical Sciences, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamil Nadu, India
| | - Shana Balachandran
- Cancer Biology Lab, Centre for Molecular and Nanomedical Sciences, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamil Nadu, India
| | - Lokesh Kumar Boopathy
- Centre for Laboratory Animal Technology and Research, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamil Nadu, India
| | - Anitha Roy
- Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, 600077, Tamil Nadu, India
| | - Bhuvaneshwari Jayachandran
- Cancer Biology Lab, Centre for Molecular and Nanomedical Sciences, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamil Nadu, India
| | - Sangamithra Sankaranarayanan
- Cancer Biology Lab, Centre for Molecular and Nanomedical Sciences, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamil Nadu, India
| | - Madan Kumar Arumugam
- Cancer Biology Lab, Centre for Molecular and Nanomedical Sciences, Sathyabama Institute of Science and Technology, Chennai, 600119, Tamil Nadu, India.
| |
Collapse
|
45
|
Leitão MIPS, Morais TS. Tailored Metal-Based Catalysts: A New Platform for Targeted Anticancer Therapies. J Med Chem 2024; 67:16967-16990. [PMID: 39348603 DOI: 10.1021/acs.jmedchem.4c01680] [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: 10/02/2024]
Abstract
Innovative strategies for targeted anticancer therapies have gained significant momentum, with metal complexes emerging as tunable catalysts for more effective and safer treatments. Rational design and engineering of metal complexes enable the development of tailored molecular structures optimized for precision oncology. The strategic incorporation of metal complex catalysts within combinatorial therapies amplifies their anticancer properties. This perspective highlights the advancements in synthetic strategies and rational design since 2019, showing how tailored metal catalysts are optimized by designing structures to release or in situ synthesize active drugs, leveraging the target-specific characteristics to develop more precise cancer therapies. This review explores metal-based catalysts, including those conjugated with biomolecules, nanostructures, and metal-organic frameworks (MOFs), highlighting their catalytic activity in biological environments and their in vitro/in vivo performance. To sum up, the potential of metal complexes as catalysts to reshape the landscape of anticancer therapies and foster novel avenues for therapeutic advancement is emphasized.
Collapse
Affiliation(s)
- Maria Inês P S Leitão
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| | - Tânia S Morais
- Centro de Química Estrutural, Institute of Molecular Sciences and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisbon, Portugal
| |
Collapse
|
46
|
Ouyang Y, Zhang P, Willner I. DNA Tetrahedra as Functional Nanostructures: From Basic Principles to Applications. Angew Chem Int Ed Engl 2024; 63:e202411118. [PMID: 39037936 DOI: 10.1002/anie.202411118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 07/24/2024]
Abstract
Self-assembled supramolecular DNA tetrahedra composed of programmed sequence-engineered complementary base-paired strands represent elusive nanostructures having key contributions to the development and diverse applications of DNA nanotechnology. By appropriate engineering of the strands, DNA tetrahedra of tuneable sizes and chemical functionalities were designed. Programmed functionalities for diverse applications were integrated into tetrahedra structures including sequence-specific recognition strands (aptamers), catalytic DNAzymes, nanoparticles, proteins, or fluorophore. The article presents a comprehensive review addressing methods to assemble and characterize the DNA tetrahedra nanostructures, and diverse applications of DNA tetrahedra framework are discussed. Topics being addressed include the application of structurally functionalized DNA tetrahedra nanostructure for the assembly of diverse optical or electrochemical sensing platforms and functionalized intracellular sensing and imaging modules. In addition, the triggered reconfiguration of DNA tetrahedra nanostructures and dynamic networks and circuits emulating biological transformations are introduced. Moreover, the functionalization of DNA tetrahedra frameworks with nanoparticles provides building units for the assembly of optical devices and for the programmed crystallization of nanoparticle superlattices. Finally, diverse applications of DNA tetrahedra in the field of nanomedicine are addressed. These include the DNA tetrahedra-assisted permeation of nanocarriers into cells for imaging, controlled drug release, active chemodynamic/photodynamic treatment of target tissues, and regenerative medicine.
Collapse
Affiliation(s)
- Yu Ouyang
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Pu Zhang
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
- Current address: Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing, 400715, P.R. China
| | - Itamar Willner
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| |
Collapse
|
47
|
Sujith S, Naresh R, Srivisanth BU, Sajeevan A, Rajaramon S, David H, Solomon AP. Aptamers: precision tools for diagnosing and treating infectious diseases. Front Cell Infect Microbiol 2024; 14:1402932. [PMID: 39386170 PMCID: PMC11461471 DOI: 10.3389/fcimb.2024.1402932] [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: 03/18/2024] [Accepted: 09/03/2024] [Indexed: 10/12/2024] Open
Abstract
Infectious diseases represent a significant global health challenge, with bacteria, fungi, viruses, and parasitic protozoa being significant causative agents. The shared symptoms among diseases and the emergence of new pathogen variations make diagnosis and treatment complex. Conventional diagnostic methods are laborious and intricate, underscoring the need for rapid, accurate techniques. Aptamer-based technologies offer a promising solution, as they are cost-effective, sensitive, specific, and convenient for molecular disease diagnosis. Aptamers, which are single-stranded RNA or DNA sequences, serve as nucleotide equivalents of monoclonal antibodies, displaying high specificity and affinity for target molecules. They are structurally robust, allowing for long-term storage without substantial activity loss. Aptamers find applications in diverse fields such as drug screening, material science, and environmental monitoring. In biomedicine, they are extensively studied for biomarker detection, diagnostics, imaging, and targeted therapy. This comprehensive review focuses on the utility of aptamers in managing infectious diseases, particularly in the realms of diagnostics and therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | - Helma David
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| |
Collapse
|
48
|
Wang Z, Liu Z, Zhang W, Li Y, Feng Y, Lv S, Diao H, Luo Z, Yan P, He M, Li X. AptaDiff: de novo design and optimization of aptamers based on diffusion models. Brief Bioinform 2024; 25:bbae517. [PMID: 39431516 PMCID: PMC11491854 DOI: 10.1093/bib/bbae517] [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: 04/01/2024] [Revised: 07/05/2024] [Accepted: 10/05/2024] [Indexed: 10/22/2024] Open
Abstract
Aptamers are single-stranded nucleic acid ligands, featuring high affinity and specificity to target molecules. Traditionally they are identified from large DNA/RNA libraries using $in vitro$ methods, like Systematic Evolution of Ligands by Exponential Enrichment (SELEX). However, these libraries capture only a small fraction of theoretical sequence space, and various aptamer candidates are constrained by actual sequencing capabilities from the experiment. Addressing this, we proposed AptaDiff, the first in silico aptamer design and optimization method based on the diffusion model. Our Aptadiff can generate aptamers beyond the constraints of high-throughput sequencing data, leveraging motif-dependent latent embeddings from variational autoencoder, and can optimize aptamers by affinity-guided aptamer generation according to Bayesian optimization. Comparative evaluations revealed AptaDiff's superiority over existing aptamer generation methods in terms of quality and fidelity across four high-throughput screening data targeting distinct proteins. Moreover, surface plasmon resonance experiments were conducted to validate the binding affinity of aptamers generated through Bayesian optimization for two target proteins. The results unveiled a significant boost of $87.9\%$ and $60.2\%$ in RU values, along with a 3.6-fold and 2.4-fold decrease in KD values for the respective target proteins. Notably, the optimized aptamers demonstrated superior binding affinity compared to top experimental candidates selected through SELEX, underscoring the promising outcomes of our AptaDiff in accelerating the discovery of superior aptamers.
Collapse
Affiliation(s)
- Zhen Wang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018 Zhejiang, China
- College of Electrical and Information Engineering, Hunan University, Changsha, 410082 Hunan, China
| | - Ziqi Liu
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018 Zhejiang, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024 Zhejiang, China
| | - Wei Zhang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018 Zhejiang, China
| | - Yanjun Li
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, FL 32610, United States
| | - Yizhen Feng
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018 Zhejiang, China
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, 310014 Zhejiang, China
| | - Shaokang Lv
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018 Zhejiang, China
- Department of Chemical Biology, Zhejiang University of Technology, Huzhou, 313200 Zhejiang, China
| | - Han Diao
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018 Zhejiang, China
- Department of Chemical Biology, Zhejiang University of Technology, Huzhou, 313200 Zhejiang, China
| | - Zhaofeng Luo
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018 Zhejiang, China
| | - Pengju Yan
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018 Zhejiang, China
- ElasticMind Inc, Hangzhou, 310018 Zhejiang, China
| | - Min He
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018 Zhejiang, China
- College of Electrical and Information Engineering, Hunan University, Changsha, 410082 Hunan, China
| | - Xiaolin Li
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, 310018 Zhejiang, China
- ElasticMind Inc, Hangzhou, 310018 Zhejiang, China
| |
Collapse
|
49
|
Yu S, Shi T, Li C, Xie C, Wang F, Liu X. Programming DNA Nanoassemblies into Polyvalent Lysosomal Degraders for Potent Degradation of Pathogenic Membrane Proteins. NANO LETTERS 2024; 24:11573-11580. [PMID: 39225423 DOI: 10.1021/acs.nanolett.4c03102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Lysosome-targeting chimera (LYTAC) shows great promise for protein-based therapeutics by targeted degradation of disease-associated membrane or extracellular proteins, yet its efficiency is constrained by the limited binding affinity between LYTAC reagents and designated proteins. Here, we established a programmable and multivalent LYTAC system by tandem assembly of DNA into a high-affinity protein degrader, a heterodimer aptamer nanostructure targeting both pathogenic membrane protein and lysosome-targeting receptor (insulin-like growth factor 2 receptor, IGF2R) with adjustable spatial distribution or organization pattern. The DNA-based multivalent LYTACs showed enhanced efficacy in removing immune-checkpoint protein programmable death-ligand 1 (PD-L1) and vascular endothelial growth factor receptor 2 (VEGFR2) in tumor cell membrane that respectively motivated a significant increase in T cell activity and a potent effect on cancer cell growth inhibition. With high programmability and versatility, this multivalent LYTAC system holds considerable promise for realizing protein therapeutics with enhanced activity.
Collapse
Affiliation(s)
- Shuyi Yu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Tianhui Shi
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Chenbiao Li
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Chongyu Xie
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Fuan Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Xiaoqing Liu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
- Beijing Life Science Academy, Beijing 102209, China
| |
Collapse
|
50
|
Dume B, Licarete E, Banciu M. Advancing cancer treatments: The role of oligonucleotide-based therapies in driving progress. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102256. [PMID: 39045515 PMCID: PMC11264197 DOI: 10.1016/j.omtn.2024.102256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Although recent advancements in cancer immunology have resulted in the approval of numerous immunotherapies, minimal progress has been observed in addressing hard-to-treat cancers. In this context, therapeutic oligonucleotides, including interfering RNAs, antisense oligonucleotides, aptamers, and DNAzymes, have gained a central role in cancer therapeutic approaches due to their capacity to regulate gene expression and protein function with reduced toxicity compared with conventional chemotherapeutics. Nevertheless, systemic administration of naked oligonucleotides faces many extra- and intracellular challenges that can be overcome by using effective delivery systems. Thus, viral and non-viral carriers can improve oligonucleotide stability and intracellular uptake, enhance tumor accumulation, and increase the probability of endosomal escape while minimizing other adverse effects. Therefore, gaining more insight into fundamental mechanisms of actions of various oligonucleotides and the challenges posed by naked oligonucleotide administration, this article provides a comprehensive review of the recent progress on oligonucleotide delivery systems and an overview of completed and ongoing cancer clinical trials that can shape future oncological treatments.
Collapse
Affiliation(s)
- Bogdan Dume
- Doctoral School in Integrative Biology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| | - Emilia Licarete
- Department of Molecular Biology and Biotechnology, Centre of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Centre of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| |
Collapse
|