1
|
Xiang ZR, Fan SR, Ren J, Ruan T, Chen Y, Zhang YW, Wang YT, Yu ZZ, Wang CF, Sun XL, Hao XJ, Chen DZ. Utilizing artificial intelligence for precision exploration of N protein targeting phenanthridine sars-cov-2 inhibitors: A novel approach. Eur J Med Chem 2024; 279:116885. [PMID: 39307103 DOI: 10.1016/j.ejmech.2024.116885] [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: 03/04/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 10/28/2024]
Abstract
The persistent mutation of the novel coronavirus presents a continual threat of infections and associated illnesses. While considerable research efforts have concentrated on the functional proteins of SARS-CoV-2 in the development of anti-COVID-19 therapeutics, the structural proteins, particularly the N protein, have received comparatively less attention. This study focuses on the N protein, a critical structural component of the virus, and employs advanced deep learning models, including EMPIRE and DeepFrag, to optimize the structures of phenanthridine-based compounds. More than 10,000 small molecules, derived through deep learning, underwent high-throughput virtual screening, resulting in the synthesis of 44 compounds. Compound 38 showed a binding potential energy of -8.2 kcal/mol in molecular docking. Surface Plasmon Resonance (SPR) and Microscale Thermophoresis (MST) validation yielded dissociation constants of 353 nM and 726 nM, confirming strong binding to the N protein. Compound 38 demonstrated antiviral activity in vitro and exhibited anti-COVID-19 effects by interfering with the binding of N proteins to RNA. This research underscores the potential of targeting the SARS-CoV-2 N protein for therapeutic intervention and illustrates the efficacy of deep learning model in the design of lead compounds. The application of these deep learning models represents a promising approach for accelerating the discovery and development of antiviral agents.
Collapse
Affiliation(s)
- Zheng-Rui Xiang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Shi-Rui Fan
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Juan Ren
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Ting Ruan
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Yuan Chen
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Yun-Wu Zhang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China
| | - Yi-Ting Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Ze-Zhou Yu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Chao-Fan Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Research Unit of Chemical Biology of Natural Anti-Virus Products, Chinese Academy of Medical Sciences, Beijing, 100730, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Xiao-Long Sun
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China; Institute of International Rivers and Eco-Security, Yunnan University, Kunming, Yunnan, 650091, China
| | - Xiao-Jiang Hao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
| | - Duo-Zhi Chen
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
| |
Collapse
|
2
|
Pandey B, Wang Z, Jimenez A, Bhatia E, Jain R, Beach A, Maniar D, Hosten J, O'Farrell L, Vantucci C, Hur D, Noel R, Ringquist R, Smith C, Ochoa MA, Roy K. A Dual-Adjuvanted Parenteral-Intranasal Subunit Nanovaccine generates Robust Systemic and Mucosal Immunity Against SARS-CoV-2 in Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402792. [PMID: 39352717 PMCID: PMC11615772 DOI: 10.1002/advs.202402792] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 09/09/2024] [Indexed: 12/06/2024]
Abstract
Existing parenteral SARS-CoV-2 vaccines produce only limited mucosal responses, essential for reducing transmission and achieving sterilizing immunity. Appropriately designed mucosal boosters can overcome the shortcomings of parenteral vaccines and enhance pre-existing systemic immunity. Here, a new protein subunit nanovaccine is developed by utilizing dual-adjuvanted (RIG-I: PUUC RNA and TLR-9: CpG DNA) polysaccharide-amino acid-lipid nanoparticles (PAL-NPs) along with SARS-CoV-2 S1 trimer protein, that can be delivered both intramuscularly (IM) and intranasally (IN) to generate balanced mucosal-systemic SARS-CoV-2 immunity. Mice receiving IM-Prime PUUC+CpG PAL subunit nanovaccine, followed by an IN-Boost, developed high levels of IgA, IgG, and cellular immunity in the lungs and showed robust systemic humoral immunity. Interestingly, as a purely intranasal subunit vaccine (IN-Prime/IN-Boost), PUUC+CpG PAL-NPs induced stronger lung-specific T cell immunity than IM-Prime/IN-Boost, and a comparable IgA and neutralizing antibodies, although with a lower systemic antibody response, indicating that a fully mucosal delivery route for SARS-CoV-2 vaccination may also be feasible. The data suggest that PUUC+CpG PAL subunit nanovaccine is a promising candidate for generating SARS-CoV-2 specific mucosal immunity.
Collapse
MESH Headings
- Animals
- Mice
- Immunity, Mucosal/immunology
- Immunity, Mucosal/drug effects
- SARS-CoV-2/immunology
- Administration, Intranasal/methods
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- COVID-19/immunology
- COVID-19/prevention & control
- Nanoparticles/administration & dosage
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
- Antibodies, Viral/immunology
- Female
- Adjuvants, Vaccine/administration & dosage
- Spike Glycoprotein, Coronavirus/immunology
- Adjuvants, Immunologic/administration & dosage
- Antibodies, Neutralizing/immunology
- Mice, Inbred BALB C
- Nanovaccines
Collapse
Affiliation(s)
- Bhawana Pandey
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Zhengying Wang
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Angela Jimenez
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Eshant Bhatia
- Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Ritika Jain
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Alexander Beach
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Drishti Maniar
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Justin Hosten
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Laura O'Farrell
- Physiological Research LaboratoryGeorgia Institute of TechnologyAtlantaGAUSA
| | - Casey Vantucci
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - David Hur
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Richard Noel
- Physiological Research LaboratoryGeorgia Institute of TechnologyAtlantaGAUSA
| | - Rachel Ringquist
- The Parker H. Petit Institute for Bioengineering and BiosciencesSchool of Chemical & Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Clinton Smith
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Miguel A. Ochoa
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyAtlantaGAUSA
| | - Krishnendu Roy
- Wallace H. Coulter Department of Biomedical EngineeringThe Parker H. Petit Institute for Bioengineering and BiosciencesMarcus Center for Therapeutic Cell Characterization and ManufacturingGeorgia Institute of TechnologyAtlantaGAUSA
- Department of Biomedical EngineeringDepartment of Chemical and Biomolecular EngineeringSchool of EngineeringDepartment of Pathology, Microbiology and ImmunologySchool of MedicineVanderbilt UniversityNashvilleTNUSA
| |
Collapse
|
3
|
Yee YC, Mori T, Ito S, Taguchi T, Katayama Y. Impact of hydrophobic modification on biocompatibility of Alaska pollock gelatin microparticles. ANAL SCI 2024; 40:2053-2061. [PMID: 39120821 DOI: 10.1007/s44211-024-00643-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 07/28/2024] [Indexed: 08/10/2024]
Abstract
This study investigates the impact of hydrophobic modification on the immunogenicity, cytotoxicity, and inflammatory response of Alaska pollock gelatin (ApGltn) microparticles (MPs). Gelatin, known for its inherent biocompatibility, was modified with decyl group (C10) to explore potential alterations in its interaction with the immune system. Immunogenicity was evaluated through the measurement of material-specific IgM and IgG responses, indicating no significant increase post-modification. Cytotoxicity against Caco-2 cell lines and NF-κB-mediated LPS-induced inflammation were also assessed, revealing no exacerbation by the modified MPs. Furthermore, C10 modification with different types of linkage such as secondary amine and amide structure did not influence immune reactivity. These findings suggest that C10 modification maintains the non-immunogenicity and biocompatibility of gelatin MPs, supporting their potential use in biomedical applications.
Collapse
Affiliation(s)
- Ying Chuin Yee
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Takeshi Mori
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
- Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
| | - Shima Ito
- Degree Programs in Pure and Applied Sciences, Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
- Biomaterials Field, Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| | - Tetsushi Taguchi
- Degree Programs in Pure and Applied Sciences, Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
- Biomaterials Field, Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan.
| | - Yoshiki Katayama
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
- Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
- Center for Molecular Systems, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
- Centre for Advanced Medicine Open Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
- Department of Biomedical Engineering, Chung Yuan Christian University, 200 Chung Pei Rd., Chung Li, 32023, Taiwan, ROC.
| |
Collapse
|
4
|
Jogdeo CM, Siddhanta K, Das A, Ding L, Panja S, Kumari N, Oupický D. Beyond Lipids: Exploring Advances in Polymeric Gene Delivery in the Lipid Nanoparticles Era. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404608. [PMID: 38842816 PMCID: PMC11384239 DOI: 10.1002/adma.202404608] [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: 03/29/2024] [Revised: 05/23/2024] [Indexed: 06/07/2024]
Abstract
The recent success of gene therapy during the COVID-19 pandemic has underscored the importance of effective and safe delivery systems. Complementing lipid-based delivery systems, polymers present a promising alternative for gene delivery. Significant advances have been made in the recent past, with multiple clinical trials progressing beyond phase I and several companies actively working on polymeric delivery systems which provides assurance that polymeric carriers can soon achieve clinical translation. The massive advantage of structural tunability and vast chemical space of polymers is being actively leveraged to mitigate shortcomings of traditional polycationic polymers and improve the translatability of delivery systems. Tailored polymeric approaches for diverse nucleic acids and for specific subcellular targets are now being designed to improve therapeutic efficacy. This review describes the recent advances in polymer design for improved gene delivery by polyplexes and covalent polymer-nucleic acid conjugates. The review also offers a brief note on novel computational techniques for improved polymer design. The review concludes with an overview of the current state of polymeric gene therapies in the clinic as well as future directions on their translation to the clinic.
Collapse
Affiliation(s)
- Chinmay M Jogdeo
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kasturi Siddhanta
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ashish Das
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ling Ding
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Sudipta Panja
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Neha Kumari
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
5
|
Zhao T, Ren M, Shi J, Wang H, Bai J, Du W, Xiang B. Engineering the protein corona: Strategies, effects, and future directions in nanoparticle therapeutics. Biomed Pharmacother 2024; 175:116627. [PMID: 38653112 DOI: 10.1016/j.biopha.2024.116627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
Nanoparticles (NPs) serve as versatile delivery systems for anticancer, antibacterial, and antioxidant agents. The manipulation of protein-NP interactions within biological systems is crucial to the application of NPs in drug delivery and cancer nanotherapeutics. The protein corona (PC) that forms on the surface of NPs is the interface between biomacromolecules and NPs and significantly influences their pharmacokinetics and pharmacodynamics. Upon encountering proteins, NPs undergo surface alterations that facilitate their clearance from circulation by the mononuclear phagocytic system (MPS). PC behavior depends largely on the biological microenvironment and the physicochemical properties of the NPs. This review describes various strategies employed to engineer PC compositions on NP surfaces. The effects of NP characteristics such as size, shape, surface modification and protein precoating on PC performance were explored. In addition, this study addresses these challenges and guides the future directions of this evolving field.
Collapse
Affiliation(s)
- Tianyu Zhao
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Mingli Ren
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiajie Shi
- Department of Breast Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Haijiao Wang
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing Bai
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Wenli Du
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Bai Xiang
- Department of Pharmaceutics, Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
6
|
Wang J, Xu Y, Zhou Y, Zhang J, Jia J, Jiao P, Liu Y, Su G. Modulating the toxicity of engineered nanoparticles by controlling protein corona formation: Recent advances and future prospects. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 914:169590. [PMID: 38154635 DOI: 10.1016/j.scitotenv.2023.169590] [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: 09/28/2023] [Revised: 12/11/2023] [Accepted: 12/17/2023] [Indexed: 12/30/2023]
Abstract
With the rapid development and widespread application of engineered nanoparticles (ENPs), understanding the fundamental interactions between ENPs and biological systems is essential to assess and predict the fate of ENPs in vivo. When ENPs are exposed to complex physiological environments, biomolecules quickly and inevitably adsorb to ENPs to form a biomolecule corona, such as a protein corona (PC). The formed PC has a significant effect on the physicochemical properties of ENPs and gives them a brand new identity in the biological environment, which determines the subsequent ENP-cell/tissue/organ interactions. Controlling the formation of PCs is therefore of utmost importance to accurately predict and optimize the behavior of ENPs within living organisms, as well as ensure the safety of their applications. In this review, we provide an overview of the fundamental aspects of the PC, including the formation mechanism, composition, and frequently used characterization techniques. We comprehensively discuss the potential impact of the PC on ENP toxicity, including cytotoxicity, immune response, and so on. Additionally, we summarize recent advancements in manipulating PC formation on ENPs to achieve the desired biological outcomes. We further discuss the challenges and prospects, aiming to provide valuable insights for a better understanding and prediction of ENP behaviors in vivo, as well as the development of low-toxicity ENPs.
Collapse
Affiliation(s)
- Jiali Wang
- School of Pharmacy, Nantong University, Nantong 226019, China; School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
| | - Yuhang Xu
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Yun Zhou
- School of Pharmacy, Nantong University, Nantong 226019, China
| | - Jian Zhang
- Digestive Diseases Center, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 510001, China; Center for Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-sen University, 510001 Guangzhou, China
| | - Jianbo Jia
- Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Peifu Jiao
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan 250200, China
| | - Yin Liu
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China.
| | - Gaoxing Su
- School of Pharmacy, Nantong University, Nantong 226019, China.
| |
Collapse
|
7
|
Casper J, Schenk SH, Parhizkar E, Detampel P, Dehshahri A, Huwyler J. Polyethylenimine (PEI) in gene therapy: Current status and clinical applications. J Control Release 2023; 362:667-691. [PMID: 37666302 DOI: 10.1016/j.jconrel.2023.09.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/24/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
Polyethlyenimine (PEI) was introduced 1995 as a cationic polymer for nucleic acid delivery. PEI and its derivatives are extensively used in basic research and as reference formulations in the field of polymer-based gene delivery. Despite its widespread use, the number of clinical applications to date is limited. Thus, this review aims to consolidate the past applications of PEI in DNA delivery, elucidate the obstacles that hinder its transition to clinical use, and highlight potential prospects for novel iterations of PEI derivatives. The present review article is divided into three sections. The first section examines the mechanism of action employed by PEI, examining fundamental aspects of cellular delivery including uptake mechanisms, release from endosomes, and transport into the cell nucleus, along with potential strategies for enhancing these delivery phases. Moreover, an in-depth analysis is conducted concerning the mechanism underlying cellular toxicity, accompanied with approaches to overcome this major challenge. The second part is devoted to the in vivo performance of PEI and its application in various therapeutic indications. While systemic administration has proven to be challenging, alternative localized delivery routes hold promise, such as treatment of solid tumors, application as a vaccine, or serving as a therapeutic agent for pulmonary delivery. In the last section, the outcome of completed and ongoing clinical trials is summarized. Finally, an expert opinion is provided on the potential of PEI and its future applications. PEI-based formulations for nucleic acid delivery have a promising potential, it will be an important task for the years to come to introduce innovations that address PEI-associated shortcomings by introducing well-designed PEI formulations in combination with an appropriate route of administration.
Collapse
Affiliation(s)
- Jens Casper
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Susanne H Schenk
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Elahehnaz Parhizkar
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Pascal Detampel
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Ali Dehshahri
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| |
Collapse
|
8
|
Weiss AM, Lopez MA, Rawe BW, Manna S, Chen Q, Mulder EJ, Rowan SJ, Esser-Kahn AP. Understanding How Cationic Polymers' Properties Inform Toxic or Immunogenic Responses via Parametric Analysis. Macromolecules 2023; 56:7286-7299. [PMID: 37781211 PMCID: PMC10537447 DOI: 10.1021/acs.macromol.3c01223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/16/2023] [Indexed: 10/03/2023]
Abstract
Cationic polymers are widely used materials in diverse biotechnologies. Subtle variations in these polymers' properties can change them from exceptional delivery agents to toxic inflammatory hazards. Conventional screening strategies optimize for function in a specific application rather than observing how underlying polymer-cell interactions emerge from polymers' properties. An alternative approach is to map basic underlying responses, such as immunogenicity or toxicity, as a function of basic physicochemical parameters to inform the design of materials for a breadth of applications. To demonstrate the potential of this approach, we synthesized 107 polymers varied in charge, hydrophobicity, and molecular weight. We then screened this library for cytotoxic behavior and immunogenic responses to map how these physicochemical properties inform polymer-cell interactions. We identify three compositional regions of interest and use confocal microscopy to uncover the mechanisms behind the observed responses. Finally, immunogenic activity is confirmed in vivo. Highly cationic polymers disrupted the cellular plasma membrane to induce a toxic phenotype, while high molecular weight, hydrophobic polymers were uptaken by active transport to induce NLRP3 inflammasome activation, an immunogenic phenotype. Tertiary amine- and triethylene glycol-containing polymers did not invoke immunogenic or toxic responses. The framework described herein allows for the systematic characterization of new cationic materials with different physicochemical properties for applications ranging from drug and gene delivery to antimicrobial coatings and tissue scaffolds.
Collapse
Affiliation(s)
- Adam M. Weiss
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S Ellis Ave., Chicago, Illinois 60637, United States
- Department
of Chemistry, University of Chicago, 5735 S Ellis Ave., Chicago, Illinois 60637, United States
| | - Marcos A. Lopez
- Department
of Chemistry, University of Chicago, 5735 S Ellis Ave., Chicago, Illinois 60637, United States
| | - Benjamin W. Rawe
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S Ellis Ave., Chicago, Illinois 60637, United States
| | - Saikat Manna
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S Ellis Ave., Chicago, Illinois 60637, United States
| | - Qing Chen
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S Ellis Ave., Chicago, Illinois 60637, United States
| | - Elizabeth J. Mulder
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S Ellis Ave., Chicago, Illinois 60637, United States
| | - Stuart J. Rowan
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S Ellis Ave., Chicago, Illinois 60637, United States
- Department
of Chemistry, University of Chicago, 5735 S Ellis Ave., Chicago, Illinois 60637, United States
| | - Aaron P. Esser-Kahn
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S Ellis Ave., Chicago, Illinois 60637, United States
| |
Collapse
|
9
|
Liang H, Yan Y, Sun W, Ma X, Su Z, Liu Z, Chen Y, Yu B. Preparation of Melatonin-Loaded Nanoparticles with Targeting and Sustained Release Function and Their Application in Osteoarthritis. Int J Mol Sci 2023; 24:ijms24108740. [PMID: 37240086 DOI: 10.3390/ijms24108740] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/07/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
(1) The vicious cycle of innate immune response and reactive oxygen species (ROS) generation is an important pathological process of osteoarthritis (OA). Melatonin may be a new hope for the treatment of OA because of its antioxidant capacity. However, the mechanism of melatonin in the treatment of OA is still not completely clear, and the physiological characteristics of articular cartilage make melatonin unable to play a long-term role in OA. (2) The effects of melatonin on ROS and the innate immune response system in OA chondrocytes and the therapeutic effect in vivo were evaluated. Then, a melatonin-loaded nano-delivery system (MT@PLGA-COLBP) was prepared and characterized. Finally, the behavior of MT@PLGA-COLPB in cartilage and the therapeutic effect in OA mice were evaluated. (3) Melatonin can inhibit the activation of the innate immune system by inhibiting the TLR2/4-MyD88-NFκB signal pathway and scavenging ROS, thus improving cartilage matrix metabolism and delaying the progression of OA in vivo. MT@PLGA-COLBP can reach the interior of cartilage and complete the accumulation in OA knee joints. At the same time, it can reduce the number of intra-articular injections and improve the utilization rate of melatonin in vivo. (4) This work provides a new idea for the treatment of osteoarthritis, updates the mechanism of melatonin in the treatment of osteoarthritis, and highlights the application prospect of PLGA@MT-COLBP nanoparticles in preventing OA.
Collapse
Affiliation(s)
- Haifeng Liang
- Orthopedic and Traumatology Department, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yiran Yan
- Orthopedic and Traumatology Department, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Wei Sun
- Orthopedic and Traumatology Department, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Xiaogang Ma
- Orthopedic and Traumatology Department, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Zhiwen Su
- Orthopedic and Traumatology Department, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Zhongxun Liu
- Orthopedic and Traumatology Department, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yan Chen
- Ultrasound Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Bo Yu
- Orthopedic and Traumatology Department, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| |
Collapse
|
10
|
Pandey B, Wang Z, Jimenez A, Bhatia E, Jain R, Beach A, Maniar D, Hosten J, O'Farrell L, Vantucci C, Hur D, Noel R, Ringuist R, Smith C, Ochoa MA, Roy K. A multiadjuvant polysaccharide-amino acid-lipid (PAL) subunit nanovaccine generates robust systemic and lung-specific mucosal immune responses against SARS-CoV-2 in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.05.539395. [PMID: 37215018 PMCID: PMC10197586 DOI: 10.1101/2023.05.05.539395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Existing parenteral SARS-CoV-2 vaccines produce only limited mucosal responses, which are essential for reducing transmission and achieving sterilizing immunity. Appropriately designed mucosal boosters could overcome the shortcomings of parenteral vaccines and enhance pre- existing systemic immunity. Here we present a new protein subunit nanovaccine using multiadjuvanted (e.g. RIG-I: PUUC, TLR9: CpG) polysaccharide-amino acid-lipid nanoparticles (PAL-NPs) that can be delivered both intramuscularly (IM) and intranasally (IN) to generate balanced mucosal-systemic SARS-CoV-2 immunity. Mice receiving IM-Prime PUUC+CpG PAL- NPs, followed by an IN-Boost, developed high levels of IgA, IgG, and cellular immunity in the lung, and showed robust systemic humoral immunity. Interestingly, as a purely intranasal vaccine (IN-Prime/IN-Boost), PUUC+CpG PAL-NPs induced stronger lung-specific T cell immunity than IM-Prime/IN-Boost, and a comparable IgA and neutralizing antibodies, although with a lower systemic antibody response, indicating that a fully mucosal delivery route for SARS-CoV-2 vaccination may also be feasible. Our data suggest that PUUC+CpG PAL-NP subunit vaccine is a promising candidate for generating SARS-CoV-2 specific mucosal immunity.
Collapse
|
11
|
Gagliardi M, Chiarugi S, De Cesari C, Di Gregorio G, Diodati A, Baroncelli L, Cecchini M, Tonazzini I. Crosslinked Chitosan Nanoparticles with Muco-Adhesive Potential for Intranasal Delivery Applications. Int J Mol Sci 2023; 24:6590. [PMID: 37047562 PMCID: PMC10094788 DOI: 10.3390/ijms24076590] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/22/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Intranasal drug delivery is convenient and provides a high bioavailability but requires the use of mucoadhesive nanocarriers. Chitosan is a well-established polymer for mucoadhesive applications but can suffer from poor cytocompatibility and stability upon administration. In this work, we present a method to obtain stable and cytocompatible crosslinked chitosan nanoparticles. We used 2,6-pyridinedicarboxylic acid as a biocompatible crosslinker and compared the obtained particles with those prepared by ionotropic gelation using sodium tripolyphosphate. Nanoparticles were tested to evaluate the size and the surface charge, as well as their stability in storage conditions (4 °C), at the nasal cavity temperature (32 °C), and at the body temperature (37 °C). The crosslinked chitosan nanoparticles showed a size around 150 nm and a surface charge of 10.3 mV ± 0.9 mV, both compatible with the intranasal drug administration. Size and surface charge parameters did not significantly vary over time, indicating the good stability of these nanoparticles. We finally tested their cytocompatibility in vitro using SHSY5Y human neuroblastoma and RPMI 2650 human nasal epithelial cells, with positive results. In conclusion, the proposed synthetic system shows an interesting potential as a drug carrier for intranasal delivery.
Collapse
Affiliation(s)
- Mariacristina Gagliardi
- National Enterprise for nanoScience and nanoTechnology (NEST), Nanoscience Institute—National Research Council (CNR) and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Sara Chiarugi
- National Enterprise for nanoScience and nanoTechnology (NEST), Nanoscience Institute—National Research Council (CNR) and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Chiara De Cesari
- National Enterprise for nanoScience and nanoTechnology (NEST), Nanoscience Institute—National Research Council (CNR) and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Giulia Di Gregorio
- National Enterprise for nanoScience and nanoTechnology (NEST), Nanoscience Institute—National Research Council (CNR) and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Alessandra Diodati
- National Enterprise for nanoScience and nanoTechnology (NEST), Nanoscience Institute—National Research Council (CNR) and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Laura Baroncelli
- Institute of Neuroscience, National Research Council (CNR), Via Giuseppe Moruzzi 1, 56124 Pisa, Italy
- Department of Developmental Neuroscience, Scientific Institute Stella Maris Foundation, Viale del Tirreno 331, Calambrone, 56128 Pisa, Italy
| | - Marco Cecchini
- National Enterprise for nanoScience and nanoTechnology (NEST), Nanoscience Institute—National Research Council (CNR) and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Ilaria Tonazzini
- National Enterprise for nanoScience and nanoTechnology (NEST), Nanoscience Institute—National Research Council (CNR) and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
| |
Collapse
|
12
|
Bruno MC, Cristiano MC, Celia C, d'Avanzo N, Mancuso A, Paolino D, Wolfram J, Fresta M. Injectable Drug Delivery Systems for Osteoarthritis and Rheumatoid Arthritis. ACS NANO 2022; 16:19665-19690. [PMID: 36512378 DOI: 10.1021/acsnano.2c06393] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Joint diseases are one of the most common causes of morbidity and disability worldwide. The main diseases that affect joint cartilage are osteoarthritis and rheumatoid arthritis, which require chronic treatment focused on symptomatic relief. Conventional drugs administered through systemic or intra-articular routes have low accumulation and/or retention in articular cartilage, causing dose-limiting toxicities and reduced efficacy. Therefore, there is an urgent need to develop improved strategies for drug delivery, in particular, the use of micro- and nanotechnology-based methods. Encapsulation of therapeutic agents in delivery systems reduces drug efflux from the joint and protects against rapid cellular and enzymatic clearance following intra-articular injection. Consequently, the use of drug delivery systems decreases side effects and increases therapeutic efficacy due to enhanced drug retention in the intra-articular space. Additionally, the frequency of intra-articular administration is reduced, as delivery systems enable sustained drug release. This review summarizes various advanced drug delivery systems, such as nano- and microcarriers, developed for articular cartilage diseases.
Collapse
Affiliation(s)
- Maria Chiara Bruno
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Maria Chiara Cristiano
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Christian Celia
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, Chieti, I-66100, Italy
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307, Kaunas, Lithuania
| | - Nicola d'Avanzo
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, Chieti, I-66100, Italy
| | - Antonia Mancuso
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Joy Wolfram
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Massimo Fresta
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| |
Collapse
|
13
|
p38MAPK guards the integrity of endosomal compartments through regulating necrotic death. Sci Rep 2022; 12:16357. [PMID: 36175595 PMCID: PMC9523023 DOI: 10.1038/s41598-022-20786-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 09/19/2022] [Indexed: 11/08/2022] Open
Abstract
Pathogens trigger activation of sensors of the innate immune system that initiate molecular signaling enabling appropriate host defense programs. Although recognition of pathogen-specific moieties or PAMPs by specialized receptors of the immune system is well defined for a great number of pathogens, the mechanisms of sensing of pathogen-induced functional perturbations to the host cell remain poorly understood. Here we show that the disruption of endosomal compartments in macrophages by a bacterium or fully synthetic nanoparticles activates stress-response p38MAPK kinase, which triggers execution of cell death of a necrotic type. p38MAPK-mediated necrosis occurs in cells with a compound homozygous deletion of pyroptosis-inducing caspases-1 and -11, apoptotic caspase-8, and necroptosis-inducing receptor-interacting protein kinase-3 (RIPK3), indicating that all of these principal cell death mediators are dispensable for p38MAPK-induced necrosis in response to endosome rupture. p38MAPK-mediated necrosis is suppressed by the receptor-interacting protein kinase 1, RIPK1, and degradation of RIPK1 sensitizes macrophages to necrotic death. Since pathogen-induced cell death of necrotic types is implicated in host defense against infection, our results indicate that functional perturbations in host cells are sensed as a component of the innate immune system.
Collapse
|
14
|
Soh WWM, Teoh RYP, Zhu J, Xun Y, Wee CY, Ding J, Thian ES, Li J. Facile Construction of a Two-in-One Injectable Micelleplex-Loaded Thermogel System for the Prolonged Delivery of Plasmid DNA. Biomacromolecules 2022; 23:3477-3492. [PMID: 35878156 DOI: 10.1021/acs.biomac.2c00648] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Nanoparticle-hydrogel systems have recently emerged as a class of interesting hybrid materials with immense potential for several biomedical applications. Remarkably, the incorporation of nanoparticles into a hydrogel may yield synergistic benefits lacking in a singular system. However, most synthetic strategies require laborious steps to achieve the system, severely restricting the process of translational research. Herein, a facile strategy to access a two-in-one system comprising two distinct polyurethane (PU)-based micellar systems is demonstrated and applied as a novel sustained gene delivery platform, where the two PUs are synthesized similarly but with slightly different compositions. One PU forms cationic micelles that complex with plasmid DNA (pDNA), which are loaded into a thermogel formed by another PU micellar system for the prolonged release of pDNA micelleplexes. Specifically, a thermogelling multiblock PU copolymer (denoted as EPH) was synthesized via the step-growth polymerization of poly(ethylene glycol), poly(propylene glycol), and poly(3-hydroxybutyrate). By further introducing a cationic extender, 3-(dimethylamino)-1,2-propanediol, into the reaction feed, a series of cationic PUs (denoted as EPHD) with varying compositions were obtained. The EPHDs formed positively charged micelles in aqueous solutions, efficiently condensed pDNA into nano-sized micelleplexes (<200 nm) at optimized w/w ratios, and mediated transient green fluorescence protein expression in HEK293T cells at 48 h post-transfection. On the other hand, aqueous EPH solution (4 wt %) was injectable at 4 °C and rapidly gelled upon heating to 37 °C to form a stable hydrogel depot. EPHD/pDNA micelleplexes were easily loaded into EPH by mixing the solutions at 4 °C, before heating to 37 °C, leading to the resultant hydrogel system. The in vitro release study revealed that while free pDNA loaded in the thermogel was completely released in 2 weeks, the release of EPHD/pDNA micelleplexes was prolonged to at least 28 days, suggesting substantial micelleplex-hydrogel interactions. Intact, bioactive, and noncytotoxic EPHD/pDNA micelleplexes in the release media were proved by gel retardation, in vitro gene transfection, and CCK-8 cytotoxicity assay results, respectively. Collectively, this work presents a simple approach to achieving and optimizing a novel two-in-one nanoparticle-hydrogel system for the prolonged delivery of pDNA and may be promising for long-term gene delivery applications.
Collapse
Affiliation(s)
- Wilson Wee Mia Soh
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Rachel Yun Pei Teoh
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Jingling Zhu
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore.,NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Yanran Xun
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Chien Yi Wee
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Jun Ding
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Eng San Thian
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Jun Li
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore.,NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| |
Collapse
|
15
|
Xie B, Du K, Huang F, Lin Z, Wu L. Cationic Nanomaterials for Autoimmune Diseases Therapy. Front Pharmacol 2022; 12:762362. [PMID: 35126109 PMCID: PMC8813968 DOI: 10.3389/fphar.2021.762362] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 12/30/2021] [Indexed: 01/14/2023] Open
Abstract
Cationic nanomaterials are defined as nanoscale structures smaller than 100 nm bearing positive charges. They have been investigated to apply to many aspects including clinical diagnosis, gene delivery, drug delivery, and tissue engineering for years. Recently, a novel concept has been made to use cationic nanomaterials as cell-free nucleic acid scavengers and inhibits the inflammatory responses in autoimmune diseases. Here, we highlighted different types of cationic materials which have the potential for autoimmune disease treatment and reviewed the strategy for autoimmune diseases therapy based on cationic nanoparticles. This review will also demonstrate the challenges and possible solutions that are encountered during the development of cationic materials-based therapeutics for autoimmune diseases.
Collapse
Affiliation(s)
- Baozhao Xie
- Division of Rheumatology, Department of Internal Medicine, the 7th Affiliated Hospital, Guang Xi Medical University, Wuzhou, China
| | - Keqian Du
- Department of Rheumatology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Fujian Huang
- Center for Chemical Biology and Drug Discovery, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhiming Lin
- Department of Rheumatology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Linping Wu
- Center for Chemical Biology and Drug Discovery, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
16
|
Si Z, Zheng W, Prananty D, Li J, Koh CH, Kang ET, Pethe K, Chan-Park MB. Polymers as advanced antibacterial and antibiofilm agents for direct and combination therapies. Chem Sci 2022; 13:345-364. [PMID: 35126968 PMCID: PMC8729810 DOI: 10.1039/d1sc05835e] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/12/2021] [Indexed: 12/13/2022] Open
Abstract
The growing prevalence of antimicrobial drug resistance in pathogenic bacteria is a critical threat to global health. Conventional antibiotics still play a crucial role in treating bacterial infections, but the emergence and spread of antibiotic-resistant micro-organisms are rapidly eroding their usefulness. Cationic polymers, which target bacterial membranes, are thought to be the last frontier in antibacterial development. This class of molecules possesses several advantages including a low propensity for emergence of resistance and rapid bactericidal effect. This review surveys the structure-activity of advanced antimicrobial cationic polymers, including poly(α-amino acids), β-peptides, polycarbonates, star polymers and main-chain cationic polymers, with low toxicity and high selectivity to potentially become useful for real applications. Their uses as potentiating adjuvants to overcome bacterial membrane-related resistance mechanisms and as antibiofilm agents are also covered. The review is intended to provide valuable information for design and development of cationic polymers as antimicrobial and antibiofilm agents for translational applications.
Collapse
Affiliation(s)
- Zhangyong Si
- School of Chemical and Biomedical Engineering, Nanyang Technological University Singapore 637459 Singapore
| | - Wenbin Zheng
- School of Chemical and Biomedical Engineering, Nanyang Technological University Singapore 637459 Singapore
| | - Dicky Prananty
- School of Chemical and Biomedical Engineering, Nanyang Technological University Singapore 637459 Singapore
| | - Jianghua Li
- School of Chemical and Biomedical Engineering, Nanyang Technological University Singapore 637459 Singapore
| | - Chong Hui Koh
- School of Chemical and Biomedical Engineering, Nanyang Technological University Singapore 637459 Singapore
| | - En-Tang Kang
- Department of Chemical & Biomolecular Engineering, National University of Singapore 4 Engineering Drive 4, Kent Ridge Singapore 117585 Singapore
| | - Kevin Pethe
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore 636921 Singapore
- School of Biological Sciences, Nanyang Technological University Singapore 637551 Singapore
| | - Mary B Chan-Park
- School of Chemical and Biomedical Engineering, Nanyang Technological University Singapore 637459 Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore 636921 Singapore
- School of Physical & Mathematical Sciences, Nanyang Technological University Singapore 637371 Singapore
| |
Collapse
|
17
|
Drozdov AS, Nikitin PI, Rozenberg JM. Systematic Review of Cancer Targeting by Nanoparticles Revealed a Global Association between Accumulation in Tumors and Spleen. Int J Mol Sci 2021; 22:13011. [PMID: 34884816 PMCID: PMC8657629 DOI: 10.3390/ijms222313011] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 12/13/2022] Open
Abstract
Active targeting of nanoparticles toward tumors is one of the most rapidly developing topics in nanomedicine. Typically, this strategy involves the addition of cancer-targeting biomolecules to nanoparticles, and studies on this topic have mainly focused on the localization of such formulations in tumors. Here, the analysis of the factors determining efficient nanoparticle targeting and therapy, various parameters such as types of targeting molecules, nanoparticle type, size, zeta potential, dose, and the circulation time are given. In addition, the important aspects such as how active targeting of nanoparticles alters biodistribution and how non-specific organ uptake influences tumor accumulation of the targeted nanoformulations are discussed. The analysis reveals that an increase in tumor accumulation of targeted nanoparticles is accompanied by a decrease in their uptake by the spleen. There is no association between targeting-induced changes of nanoparticle concentrations in tumors and other organs. The correlation between uptake in tumors and depletion in the spleen is significant for mice with intact immune systems in contrast to nude mice. Noticeably, modulation of splenic and tumor accumulation depends on the targeting molecules and nanoparticle type. The median survival increases with the targeting-induced nanoparticle accumulation in tumors; moreover, combinatorial targeting of nanoparticle drugs demonstrates higher treatment efficiencies. Results of the comprehensive analysis show optimal strategies to enhance the efficiency of actively targeted nanoparticle-based medicines.
Collapse
Affiliation(s)
- Andrey S. Drozdov
- Laboratory of Nanobiotechnology, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia;
| | - Petr I. Nikitin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Julian M. Rozenberg
- Cell Signaling Regulation Laboratory, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
| |
Collapse
|
18
|
Chen YY, Jiang WP, Chen HL, Huang HC, Huang GJ, Chiang HM, Chang CC, Huang CL, Juang TY. Cytotoxicity and cell imaging of six types of carbon nanodots prepared through carbonization and hydrothermal processing of natural plant materials. RSC Adv 2021; 11:16661-16674. [PMID: 35479143 PMCID: PMC9031421 DOI: 10.1039/d1ra01318a] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/29/2021] [Indexed: 12/20/2022] Open
Abstract
In this study we prepared six types of carbon nanodots (CNDs) from natural plant materials – through carbonization of two species of bamboo (Bamboo-I, Bamboo-II) and one type of wood (Wood), and through hydrothermal processing of the stem and root of the herb Mahonia oiwakensis Hayata (MO) and of the agricultural waste of two species of pineapple root (PA, PB). The resulting CNDs were spherical with dimensions on the nanoscale (3–7 nm); furthermore, CND-Bamboo I, CND-Wood, CND-Bamboo II, CND-MO, CND-PA, and CND-PB displayed fluorescence quantum yields of 9.63, 12.34, 0.90, 10.86, 0.35, and 0.71%, respectively. X-ray diffraction revealed that the carbon nanostructures possessed somewhat ordered and disordered lattices, as evidenced by broad signals at values of 2θ between 20 and 30°. CND-Bamboo I, CND-Wood, and CND-Bamboo II were obtained in yields of 2–3%; CND-MO, CND-PA, and CND-PB were obtained in yields of 17.64, 9.36, and 22.47%, respectively. Cytotoxicity assays for mouse macrophage RAW264.7 cells treated with the six types of CNDs and a commercial sample of Ag nanoparticles (NPs) revealed that each of our CNDs provided a cell viability of 90% at 2000 μg mL−1, whereas it was only 20% after treatment with the Ag NPs at 62.5 μg mL−1. The six types of CNDs also displayed low cytotoxicity toward human keratinocyte HacaT cells, human MCF-7 breast cancer cells, and HT-29 colon adenocarcinoma cells when treated at 500 μg mL−1. Moreover, confocal microscopic cell imaging revealed that the fluorescent CND-Bamboo I particles were located on the MCF-7 cell membrane and inside the cells after treatment for 6 and 24 h, respectively. We have thoroughly investigated the photoluminescence properties and carbon nanostructures of these highly dispersed CNDs. Because of the facile green synthesis of these six types of CNDs and their sourcing from abundant natural plants, herbs, and agriculture waste, these materials provide a cost-effective method, with low cytotoxicity and stable fluorescence, for biolabeling and for developing cell nanocarriers. Green nanotechnology of six types of carbon nanodots (CNDs), and their sourcing from abundant natural plants, herbs, and agriculture waste, provides a cost-effective method, with low cytotoxicity and stable fluorescence, for biolabeling and for developing cell nanocarriers.![]()
Collapse
Affiliation(s)
- Yu-Yu Chen
- Department of Cosmeceutics, China Medical University Taichung Taiwan
| | - Wen-Ping Jiang
- Department of Occupational Therapy, Asia University Taichung Taiwan.,Department of Pharmacy, Chia Nan University of Pharmacy and Science Tainan Taiwan
| | - Huan-Luen Chen
- Department of Cosmeceutics, China Medical University Taichung Taiwan
| | - Hui-Chi Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University Taichung Taiwan.,Master Program for Food and Drug Safety, China Medical University Taichung Taiwan
| | - Guan-Jhong Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University Taichung Taiwan
| | - Hsiu-Mei Chiang
- Department of Cosmeceutics, China Medical University Taichung Taiwan
| | - Chang-Cheng Chang
- Aesthetic Medical Center, China Medical University Hospital Taichung Taiwan.,School of Medicine, China Medical University Taichung Taiwan
| | - Cheng-Liang Huang
- Department of Applied Chemistry, National Chiayi University Chiayi Taiwan
| | - Tzong-Yuan Juang
- Department of Cosmeceutics, China Medical University Taichung Taiwan
| |
Collapse
|
19
|
Caprifico AE, Foot PJS, Polycarpou E, Calabrese G. Overcoming the protein corona in chitosan-based nanoparticles. Drug Discov Today 2021; 26:1825-1840. [PMID: 33892141 DOI: 10.1016/j.drudis.2021.04.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/20/2021] [Accepted: 04/11/2021] [Indexed: 12/17/2022]
Abstract
Numerous properties of chitosan have led to its extensive use in the formulation of nanomaterials for drug delivery. However, the cationic surface of chitosan-based nanoparticles adsorbs proteins upon exposure to biological fluids, forming a phenomenon known as 'protein corona'. This causes several effects such as decreased bioavailability and limited in vivo clinical applications of chitosan nanoparticles. Understanding and overcoming the effects of protein adsorption on chitosan nanoparticles is key for drug delivery purposes. This review focuses on the strategies implemented to increase the stability of chitosan nanoparticles in the systemic circulation by averting the formation of protein corona and the limitations of PEGylation.
Collapse
Affiliation(s)
- Anna E Caprifico
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Penrhyn Road, Kingston upon Thames KT1 2EE, UK
| | - Peter J S Foot
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Penrhyn Road, Kingston upon Thames KT1 2EE, UK
| | - Elena Polycarpou
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Penrhyn Road, Kingston upon Thames KT1 2EE, UK
| | - Gianpiero Calabrese
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Penrhyn Road, Kingston upon Thames KT1 2EE, UK.
| |
Collapse
|
20
|
Busatto S, Iannotta D, Walker SA, Di Marzio L, Wolfram J. A Simple and Quick Method for Loading Proteins in Extracellular Vesicles. Pharmaceuticals (Basel) 2021; 14:356. [PMID: 33924377 PMCID: PMC8069621 DOI: 10.3390/ph14040356] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 02/08/2023] Open
Abstract
Extracellular vesicles (EVs) mediate intercellular transport of biomolecular cargo in the body, making them promising delivery vehicles for bioactive compounds. Genetic engineering of producer cells has enabled encapsulation of therapeutic proteins in EVs. However, genetic engineering approaches can be expensive, time-consuming, and incompatible with certain EV sources, such as human plasma and bovine milk. The goal of this study was to develop a quick, versatile, and simple method for loading proteins in EVs post-isolation. Proteins, including CRISPR associated protein 9 (Cas9), were bound to cationic lipids that were further complexed with MDA-MB-231 cell-derived EVs through passive incubation. Size-exclusion chromatography was used to remove components that were not complexed with EVs. The ability of EVs to mediate intracellular delivery of proteins was compared to conventional methods, such as electroporation and commercial protein transfection reagents. The results indicate that EVs retain native features following protein-loading and obtain similar levels of intracellular protein delivery as conventional methods, but display less toxicity. This method opens up opportunities for rapid exploration of EVs for protein delivery.
Collapse
Affiliation(s)
- Sara Busatto
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (D.I.); (S.A.W.)
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Dalila Iannotta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (D.I.); (S.A.W.)
- Department of Pharmacy, University of Chieti—Pescara “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Sierra A. Walker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (D.I.); (S.A.W.)
| | - Luisa Di Marzio
- Department of Pharmacy, University of Chieti—Pescara “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Joy Wolfram
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (D.I.); (S.A.W.)
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
21
|
Bai X, Wang J, Mu Q, Su G. In vivo Protein Corona Formation: Characterizations, Effects on Engineered Nanoparticles' Biobehaviors, and Applications. Front Bioeng Biotechnol 2021; 9:646708. [PMID: 33869157 PMCID: PMC8044820 DOI: 10.3389/fbioe.2021.646708] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/16/2021] [Indexed: 12/17/2022] Open
Abstract
Understanding the basic interactions between engineered nanoparticles (ENPs) and biological systems is essential for evaluating ENPs’ safety and developing better nanomedicine. Profound interactions between ENPs and biomolecules such as proteins are inevitable to occur when ENPs are administered or exposed to biological systems, for example, through intravenous injection, oral, or respiration. As a key component of these interactions, protein corona (PC) is immediately formed surrounding the outlayer of ENPs. PC formation is crucial because it gives ENPs a new biological identity by altering not only the physiochemical properties, but also the biobehaviors of ENPs. In the past two decades, most investigations about PC formation were carried out with in vitro systems which could not represent the true events occurring within in vivo systems. Most recently, studies of in vivo PC formation were reported, and it was found that the protein compositions and structures were very different from those formed in vitro. Herein, we provide an in-time review of the recent investigations of this in vivo PC formation of ENPs. In this review, commonly used characterization methods and compositions of in vivo PC are summarized firstly. Next, we highlight the impacts of the in vivo PC formation on absorption, blood circulation, biodistribution, metabolism, and toxicity of administered ENPs. We also introduce the applications of modulating in vivo PC formation in nanomedicine. We further discuss the challenges and future perspectives.
Collapse
Affiliation(s)
- Xue Bai
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiali Wang
- School of Pharmacy, Nantong University, Nantong, China
| | - Qingxin Mu
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States
| | - Gaoxing Su
- School of Pharmacy, Nantong University, Nantong, China
| |
Collapse
|
22
|
P-selectin targeting polysaccharide-based nanogels for miRNA delivery. Int J Pharm 2021; 597:120302. [DOI: 10.1016/j.ijpharm.2021.120302] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/20/2022]
|
23
|
Zhou X, Su Q, Zhao H, Cao X, Yang Y, Xue W. Metal-Phenolic Network-Encapsulated Nanovaccine with pH and Reduction Dual Responsiveness for Enhanced Cancer Immunotherapy. Mol Pharm 2020; 17:4603-4615. [PMID: 33175556 DOI: 10.1021/acs.molpharmaceut.0c00802] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer nanovaccines have been widely explored to enhance immunotherapy efficiency, in which the significant irritation of antigen-specific cytotoxic T cells (CTLs) is the critical point. In this study, we developed a pH and reduction dual-sensitive nanovaccine (PMSN@OVA-MPN) composed of two parts. The inner part was made up of polyethyleneimine (PEI)-modified mesoporous silica nanoparticles (MSNs) loaded with model antigen ovalbumin (OVA) and the outer part was made up of disulfide bond-involved metal-phenolic networks (MPNs) as a protective corona. In vitro release experiments proved that PMSN@OVA-MPN could intelligently release OVA in the presence of reductive glutathione, but not in neutral phosphate-buffered saline (PBS). Moreover, in vitro cell assays indicated that the nanovaccine promoted not only the OVA uptake efficiency by DC2.4 cells but also antigen lysosome escape due to the proton sponge effect of PEI. Furthermore, in vivo animal experiments indicated that PMSN@OVA-MPN induced a large tumor-specific cellular immune response so as to effectively inhibit the growth of an existing tumor. Finally, the immune memory effect caused by the nanovaccine afforded conspicuous prophylaxis efficacy in neonatal tumors. Hence, the multifunctional vaccine delivery system prepared in this work exhibits a great application potential in cancer immunotherapy and offers a platform for the development of nanovaccines.
Collapse
Affiliation(s)
- Xin Zhou
- College of Food Science and Engineering, Hainan University, Haikou 570228, Hainan, China
| | - Qianhong Su
- College of Food Science and Engineering, Hainan University, Haikou 570228, Hainan, China
| | - Hongwei Zhao
- School of Material Science and Engineering, Hunan University of Science and Technology, Xiangtan 411201, Hunan, China
| | - Xianying Cao
- College of Food Science and Engineering, Hainan University, Haikou 570228, Hainan, China
| | - Yong Yang
- College of Food Science and Engineering, Hainan University, Haikou 570228, Hainan, China
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| |
Collapse
|
24
|
He J, Xu S, Mixson AJ. The Multifaceted Histidine-Based Carriers for Nucleic Acid Delivery: Advances and Challenges. Pharmaceutics 2020; 12:E774. [PMID: 32823960 PMCID: PMC7465012 DOI: 10.3390/pharmaceutics12080774] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/17/2022] Open
Abstract
Histidines incorporated into carriers of nucleic acids may enhance the extracellular stability of the nanoparticle, yet aid in the intracellular disruption of the nanoparticle, enabling the release of the nucleic acid. Moreover, protonation of histidines in the endosomes may result in endosomal swelling with subsequent lysis. These properties of histidine are based on its five-member imidazole ring in which the two nitrogen atoms may form hydrogen bonds or act as a base in acidic environments. A wide variety of carriers have integrated histidines or histidine-rich domains, which include peptides, polyethylenimine, polysaccharides, platform delivery systems, viral phages, mesoporous silica particles, and liposomes. Histidine-rich carriers have played key roles in our understanding of the stability of nanocarriers and the escape of the nucleic acids from endosomes. These carriers show great promise and offer marked potential in delivering plasmids, siRNA, and mRNA to their intracellular targets.
Collapse
Affiliation(s)
| | | | - A. James Mixson
- Department of Pathology, University Maryland School of Medicine, 10 S. Pine St., University of Maryland, Baltimore, MD 21201, USA; (J.H.); (S.X.)
| |
Collapse
|