1
|
Sun X, Tian T, Lian Y, Cui Z. Current Advances in Viral Nanoparticles for Biomedicine. ACS NANO 2024; 18:33827-33863. [PMID: 39648920 DOI: 10.1021/acsnano.4c13146] [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: 12/10/2024]
Abstract
Viral nanoparticles (VNPs) have emerged as crucial tools in the field of biomedicine. Leveraging their biological and physicochemical properties, VNPs exhibit significant advantages in the prevention, diagnosis, and treatment of human diseases. Through techniques such as chemical bioconjugation, infusion, genetic engineering, and encapsulation, these VNPs have been endowed with multifunctional capabilities, including the display of functional peptides or proteins, encapsulation of therapeutic drugs or inorganic particles, integration with imaging agents, and conjugation with bioactive molecules. This review provides an in-depth analysis of VNPs in biomedicine, elucidating their diverse types, distinctive features, production methods, and complex design principles behind multifunctional VNPs. It highlights recent innovative research and various applications, covering their roles in imaging, drug delivery, therapeutics, gene delivery, vaccines, immunotherapy, and tissue regeneration. Additionally, the review provides an assessment of their safety and biocompatibility and discusses challenges and future opportunities in the field, underscoring the vast potential and evolving nature of VNP research.
Collapse
Affiliation(s)
- Xianxun Sun
- School of Life Sciences, Jianghan University, Wuhan 430056, China
| | - Tao Tian
- School of Life Sciences, Jianghan University, Wuhan 430056, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yindong Lian
- School of Life Sciences, Jianghan University, Wuhan 430056, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| |
Collapse
|
2
|
Jiang C, Fu J, Li F, Xia K, Li S, Chang L, Bock R, Zhang J. Plastid engineering with an efficient RNAi delivery system based on bacteriophage MS2 virus-like particles enhances plant resistance to cotton bollworm. MOLECULAR PLANT 2024; 17:987-989. [PMID: 38835168 DOI: 10.1016/j.molp.2024.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/26/2024] [Accepted: 05/31/2024] [Indexed: 06/06/2024]
Affiliation(s)
- Chunmei Jiang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei Hongshan Laboratory, Hubei University, Wuhan 430062, China
| | - Jinqiu Fu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei Hongshan Laboratory, Hubei University, Wuhan 430062, China
| | - Fujun Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei Hongshan Laboratory, Hubei University, Wuhan 430062, China
| | - Kai Xia
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei Hongshan Laboratory, Hubei University, Wuhan 430062, China
| | - Shengchun Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei Hongshan Laboratory, Hubei University, Wuhan 430062, China
| | - Ling Chang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei Hongshan Laboratory, Hubei University, Wuhan 430062, China.
| | - Ralph Bock
- Max-Planck-Institut für Molekulare Pflanzenphysiologie, Am Mühlenberg 1, 14476 Potsdam-Golm, Germany
| | - Jiang Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei Hongshan Laboratory, Hubei University, Wuhan 430062, China; Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China.
| |
Collapse
|
3
|
Travassos R, Martins SA, Fernandes A, Correia JDG, Melo R. Tailored Viral-like Particles as Drivers of Medical Breakthroughs. Int J Mol Sci 2024; 25:6699. [PMID: 38928403 PMCID: PMC11204272 DOI: 10.3390/ijms25126699] [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: 04/10/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Despite the recognized potential of nanoparticles, only a few formulations have progressed to clinical trials, and an even smaller number have been approved by the regulatory authorities and marketed. Virus-like particles (VLPs) have emerged as promising alternatives to conventional nanoparticles due to their safety, biocompatibility, immunogenicity, structural stability, scalability, and versatility. Furthermore, VLPs can be surface-functionalized with small molecules to improve circulation half-life and target specificity. Through the functionalization and coating of VLPs, it is possible to optimize the response properties to a given stimulus, such as heat, pH, an alternating magnetic field, or even enzymes. Surface functionalization can also modulate other properties, such as biocompatibility, stability, and specificity, deeming VLPs as potential vaccine candidates or delivery systems. This review aims to address the different types of surface functionalization of VLPs, highlighting the more recent cutting-edge technologies that have been explored for the design of tailored VLPs, their importance, and their consequent applicability in the medical field.
Collapse
Affiliation(s)
- Rafael Travassos
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139.7), 2695-066 Bobadela, Portugal; (R.T.); (S.A.M.); (A.F.)
| | - Sofia A. Martins
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139.7), 2695-066 Bobadela, Portugal; (R.T.); (S.A.M.); (A.F.)
| | - Ana Fernandes
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139.7), 2695-066 Bobadela, Portugal; (R.T.); (S.A.M.); (A.F.)
| | - João D. G. Correia
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139.7), 2695-066 Bobadela, Portugal; (R.T.); (S.A.M.); (A.F.)
- Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139.7), 2695-066 Bobadela, Portugal
| | - Rita Melo
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139.7), 2695-066 Bobadela, Portugal; (R.T.); (S.A.M.); (A.F.)
| |
Collapse
|
4
|
Naskalska A, Heddle JG. Virus-like particles derived from bacteriophage MS2 as antigen scaffolds and RNA protective shells. Nanomedicine (Lond) 2024; 19:1103-1115. [PMID: 38629576 PMCID: PMC11225317 DOI: 10.2217/nnm-2023-0362] [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: 12/20/2023] [Accepted: 03/06/2024] [Indexed: 07/03/2024] Open
Abstract
The versatile potential of bacteriophage MS2-derived virus-like particles (VLPs) in medical biotechnology has been extensively studied during the last 30 years. Since the first reports showing that MS2 VLPs can be produced at high yield and relatively easily engineered, numerous applications have been proposed. Particular effort has been spent in developing MS2 VLPs as protective capsules and delivery platforms for diverse molecules, such as chemical compounds, proteins and nucleic acids. Among these, two are particularly noteworthy: as scaffolds displaying heterologous epitopes for vaccine development and as capsids for encapsulation of foreign RNA. In this review, we summarize the progress in developing MS2 VLPs for these two areas.
Collapse
Affiliation(s)
- Antonina Naskalska
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, 30-392, Poland
| | - Jonathan Gardiner Heddle
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, 30-392, Poland
- Department of Biosciences, Durham University, Durham, DH1 3LE, UK
| |
Collapse
|
5
|
Davodabadi F, Mirinejad S, Malik S, Dhasmana A, Ulucan-Karnak F, Sargazi S, Sargazi S, Fathi-Karkan S, Rahdar A. Nanotherapeutic approaches for delivery of long non-coding RNAs: an updated review with emphasis on cancer. NANOSCALE 2024; 16:3881-3914. [PMID: 38353296 DOI: 10.1039/d3nr05656b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
The long noncoding RNAs (lncRNAs) comprise a wide range of RNA species whose length exceeds 200 nucleotides, which regulate the expression of genes and cellular functions in a wide range of organisms. Several diseases, including malignancy, have been associated with lncRNA dysregulation. Due to their functions in cancer development and progression, lncRNAs have emerged as promising biomarkers and therapeutic targets in cancer diagnosis and treatment. Several studies have investigated the anti-cancer properties of lncRNAs; however, only a few lncRNAs have been found to exhibit tumor suppressor properties. Furthermore, their length and poor stability make them difficult to synthesize. Thus, to overcome the instability of lncRNAs, poor specificity, and their off-target effects, researchers have constructed nanocarriers that encapsulate lncRNAs. Recently, translational medicine research has focused on delivering lncRNAs into tumor cells, including cancer cells, through nano-drug delivery systems in vivo. The developed nanocarriers can protect, target, and release lncRNAs under controlled conditions without appreciable adverse effects. To deliver lncRNAs to cancer cells, various nanocarriers, such as exosomes, microbubbles, polymer nanoparticles, 1,2-dioleyl-3-trimethylammoniumpropane chloride nanocarriers, and virus-like particles, have been successfully developed. Despite this, every nanocarrier has its own advantages and disadvantages when it comes to delivering nucleic acids effectively and safely. This article examines the current status of nanocarriers for lncRNA delivery in cancer therapy, focusing on their potential to enhance cancer treatment.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Shekoufeh Mirinejad
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi-834002, India.
| | - Archna Dhasmana
- Himalayan School of Biosciences, Swami Rama Himalayan University, Jolly Grant, Dehradun, Uttarakhand, 248140, India.
| | - Fulden Ulucan-Karnak
- Department of Medical Biochemistry, Institute of Health Sciences, Ege University, İzmir 35100, Turkey.
| | - Sara Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Sonia Fathi-Karkan
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166, Iran
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd 9414974877, Iran.
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol, P. O. Box. 98613-35856, Iran.
| |
Collapse
|
6
|
Almalki WH. An Up-to-date Review on Protein-based Nanocarriers in the Management of Cancer. Curr Drug Deliv 2024; 21:509-524. [PMID: 37165498 DOI: 10.2174/1567201820666230509101020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 02/19/2023] [Accepted: 02/27/2023] [Indexed: 05/12/2023]
Abstract
BACKGROUND A big health issue facing the world's population is cancer. An alarming increase in cancer patients was anticipated by worldwide demographic statistics, which showed that the number of patients with different malignancies was rapidly increasing. By 2025, probably 420 million cases were projected to be achieved. The most common cancers diagnosed are breast, colorectal, prostate, and lung. Conventional treatments, such as surgery, chemotherapy, and radiation therapy, have been practiced. OBJECTIVE In recent years, the area of cancer therapy has changed dramatically with expanded studies on the molecular-level detection and treatment of cancer. Recent advances in cancer research have seen significant advances in therapies such as chemotherapy and immunotherapy, although both have limitations in effectiveness and toxicity. METHODS The development of nanotechnology for anticancer drug delivery has developed several potentials as nanocarriers, which may boost the pharmacokinetic and pharmacodynamic effects of the drug product and substantially reduce the side effects. RESULTS The advancement in non-viral to viral-based protein-based nanocarriers for treating cancer has earned further recognition in this respect. Many scientific breakthroughs have relied on protein-based nanocarriers, and proteins are essential organic macromolecules for life. It allows targeted delivery of passive or active tumors using non-viral-based protein-based nanocarriers to viral-based protein nanocarriers. When targeting cancer cells, both animal and plant proteins may be used in a formulation process to create self-assembled viruses and platforms that can successfully eradicate metastatic cancer cells. CONCLUSION This review, therefore, explores in depth the applications of non-viral to viral proteinbased noncarriers with a specific focus on intracellular drug delivery and anti-cancer drug targeting ability.
Collapse
Affiliation(s)
- Waleed H Almalki
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Aal-qura University, Saudi Arabia
| |
Collapse
|
7
|
Nguyen HM, Watanabe S, Sharmin S, Kawaguchi T, Tan XE, Wannigama DL, Cui L. RNA and Single-Stranded DNA Phages: Unveiling the Promise from the Underexplored World of Viruses. Int J Mol Sci 2023; 24:17029. [PMID: 38069353 PMCID: PMC10707117 DOI: 10.3390/ijms242317029] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/26/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
RNA and single-stranded DNA (ssDNA) phages make up an understudied subset of bacteriophages that have been rapidly expanding in the last decade thanks to advancements in metaviromics. Since their discovery, applications of genetic engineering to ssDNA and RNA phages have revealed their immense potential for diverse applications in healthcare and biotechnology. In this review, we explore the past and present applications of this underexplored group of phages, particularly their current usage as therapeutic agents against multidrug-resistant bacteria. We also discuss engineering techniques such as recombinant expression, CRISPR/Cas-based genome editing, and synthetic rebooting of phage-like particles for their role in tailoring phages for disease treatment, imaging, biomaterial development, and delivery systems. Recent breakthroughs in RNA phage engineering techniques are especially highlighted. We conclude with a perspective on challenges and future prospects, emphasizing the untapped diversity of ssDNA and RNA phages and their potential to revolutionize biotechnology and medicine.
Collapse
Affiliation(s)
- Huong Minh Nguyen
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke 329-0498, Tochigi, Japan; (H.M.N.); (S.W.); (S.S.); (T.K.); (X.-E.T.)
| | - Shinya Watanabe
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke 329-0498, Tochigi, Japan; (H.M.N.); (S.W.); (S.S.); (T.K.); (X.-E.T.)
| | - Sultana Sharmin
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke 329-0498, Tochigi, Japan; (H.M.N.); (S.W.); (S.S.); (T.K.); (X.-E.T.)
| | - Tomofumi Kawaguchi
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke 329-0498, Tochigi, Japan; (H.M.N.); (S.W.); (S.S.); (T.K.); (X.-E.T.)
| | - Xin-Ee Tan
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke 329-0498, Tochigi, Japan; (H.M.N.); (S.W.); (S.S.); (T.K.); (X.-E.T.)
| | - Dhammika Leshan Wannigama
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata 990-2292, Yamagata, Japan;
| | - Longzhu Cui
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke 329-0498, Tochigi, Japan; (H.M.N.); (S.W.); (S.S.); (T.K.); (X.-E.T.)
| |
Collapse
|
8
|
Chen YL, Bao CJ, Duan JL, Xie Y, Lu WL. Overcoming biological barriers by virus-like drug particles for drug delivery. Adv Drug Deliv Rev 2023; 203:115134. [PMID: 37926218 DOI: 10.1016/j.addr.2023.115134] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Abstract
Virus-like particles (VLPs) have natural structural antigens similar to those found in viruses, making them valuable in vaccine immunization. Furthermore, VLPs have demonstrated significant potential in drug delivery, and emerged as promising vectors for transporting chemical drug, genetic drug, peptide/protein, and even nanoparticle drug. With virus-like permeability and strong retention, they can effectively target specific organs, tissues or cells, facilitating efficient intracellular drug release. Further modifications allow VLPs to transfer across various physiological barriers, thus acting the purpose of efficient drug delivery and accurate therapy. This article provides an overview of VLPs, covering their structural classifications, deliverable drugs, potential physiological barriers in drug delivery, strategies for overcoming these barriers, and future prospects.
Collapse
Affiliation(s)
- Yu-Ling Chen
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Chun-Jie Bao
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jia-Lun Duan
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ying Xie
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Wan-Liang Lu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, and School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
9
|
Al-Gazally ME, Khan R, Imran M, Ramírez-Coronel AA, Alshahrani SH, Altalbawy FMA, Turki Jalil A, Romero-Parra RM, Zabibah RS, Shahid Iqbal M, Karampoor S, Mirzaei R. The role and mechanism of action of microRNA-122 in cancer: Focusing on the liver. Int Immunopharmacol 2023; 123:110713. [PMID: 37523968 DOI: 10.1016/j.intimp.2023.110713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 07/08/2023] [Accepted: 07/24/2023] [Indexed: 08/02/2023]
Abstract
microRNA-122 (miR-122) is a highly conserved microRNA that is predominantly expressed in the liver and plays a critical role in the regulation of liver metabolism. Recent studies have shown that miR-122 is involved in the pathogenesis of various types of cancer, particularly liver cancer. In this sense, The current findings highlighted the potential role of miR-122 in regulating many vital processes in cancer pathophysiology, including apoptosis, signaling pathway, cell metabolism, immune system response, migration, and invasion. These results imply that miR-122, which has been extensively studied for its biological functions and potential therapeutic applications, acts as a tumor suppressor or oncogene in cancer development. We first provide an overview and summary of the physiological function and mode of action of miR-122 in liver cancer. We will examine the various signaling pathways and molecular mechanisms through which miR-122 exerts its effects on cancer cells, including the regulation of oncogenic and tumor suppressor genes, the modulation of cell proliferation and apoptosis, and the regulation of metastasis. Most importantly, we will also discuss the potential diagnostic and therapeutic applications of miR-122 in cancer, including the development of miRNA-based biomarkers for cancer diagnosis and prognosis, and the potential use of miR-122 as a therapeutic target for cancer treatment.
Collapse
Affiliation(s)
| | - Ramsha Khan
- MBBS, Nawaz Sharif Medical College, Gujrat, Pakistan
| | - Muhammad Imran
- MBBS, Multan Medical and Dental College, Multan, Pakistan
| | | | | | - Farag M A Altalbawy
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza 12613, Egypt; Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla 51001, Iraq
| | | | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Muhammad Shahid Iqbal
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam bin Abdulaziz University, 11942 Alkharj, Saudi Arabia
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
10
|
Mellid-Carballal R, Gutierrez-Gutierrez S, Rivas C, Garcia-Fuentes M. Viral protein-based nanoparticles (part 2): Pharmaceutical applications. Eur J Pharm Sci 2023; 189:106558. [PMID: 37567394 DOI: 10.1016/j.ejps.2023.106558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/10/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
Viral protein nanoparticles (ViP NPs) such as virus-like particles and virosomes are structures halfway between viruses and synthetic nanoparticles. The biological nature of ViP NPs endows them with the biocompatibility, biodegradability, and functional properties that many synthetic nanoparticles lack. At the same time, the absence of a viral genome avoids the safety concerns of viruses. Such characteristics of ViP NPs offer a myriad of opportunities for theirapplication at several points across disease development: from prophylaxis to diagnosis and treatment. ViP NPs present remarkable immunostimulant properties, and thus the vaccination field has benefited the most from these platforms capable of overcoming the limitations of both traditional and subunit vaccines. This was reflected in the marketing authorization of several VLP- and virosome-based vaccines. Besides, ViP NPs inherit the ability of viruses to deliver their cargo to target cells. Because of that, ViP NPs are promising candidates as vectors for drug and gene delivery, and for diagnostic applications. In this review, we analyze the pharmaceutical applications of ViP NPs, describing the products that are commercially available or under clinical evaluation, but also the advances that scientists are making toward the implementation of ViP NPs in other areas of major pharmaceutical interest.
Collapse
Affiliation(s)
- Rocio Mellid-Carballal
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain
| | - Sara Gutierrez-Gutierrez
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain
| | - Carmen Rivas
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Universidad de Santiago de Compostela, Spain; Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología (CNB)-CSIC, Spain
| | - Marcos Garcia-Fuentes
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Universidad de Santiago de Compostela, Spain.
| |
Collapse
|
11
|
Kim KR, Lee AS, Kim SM, Heo HR, Kim CS. Virus-like nanoparticles as a theranostic platform for cancer. Front Bioeng Biotechnol 2023; 10:1106767. [PMID: 36714624 PMCID: PMC9878189 DOI: 10.3389/fbioe.2022.1106767] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/31/2022] [Indexed: 01/15/2023] Open
Abstract
Virus-like nanoparticles (VLPs) are natural polymer-based nanomaterials that mimic viral structures through the hierarchical assembly of viral coat proteins, while lacking viral genomes. VLPs have received enormous attention in a wide range of nanotechnology-based medical diagnostics and therapies, including cancer therapy, imaging, and theranostics. VLPs are biocompatible and biodegradable and have a uniform structure and controllable assembly. They can encapsulate a wide range of therapeutic and diagnostic agents, and can be genetically or chemically modified. These properties have led to sophisticated multifunctional theranostic platforms. This article reviews the current progress in developing and applying engineered VLPs for molecular imaging, drug delivery, and multifunctional theranostics in cancer research.
Collapse
Affiliation(s)
- Kyeong Rok Kim
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea
| | - Ae Sol Lee
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea
| | - Su Min Kim
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea
| | - Hye Ryoung Heo
- Senotherapy-Based Metabolic Disease Control Research Center, Yeungnam University, Gyeongsan, South Korea,*Correspondence: Chang Sup Kim, ; Hye Ryoung Heo,
| | - Chang Sup Kim
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea,School of Chemistry and Biochemistry, Yeungnam University, Gyeongsan, South Korea,*Correspondence: Chang Sup Kim, ; Hye Ryoung Heo,
| |
Collapse
|
12
|
Samad AFA, Kamaroddin MF. Innovative approaches in transforming microRNAs into therapeutic tools. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 14:e1768. [PMID: 36437633 DOI: 10.1002/wrna.1768] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/29/2022]
Abstract
MicroRNA (miRNA) is regarded as a prominent genetic regulator, as it can fine-tune an entire biological pathway by targeting multiple target genes. This characteristic makes miRNAs promising therapeutic tools to reinstate cell functions that are disrupted as a consequence of diseases. Currently, miRNA replacement by miRNA mimics and miRNA inhibition by anti-miRNA oligonucleotides are the main approaches to utilizing miRNA molecules for therapeutic purposes. Nevertheless, miRNA-based therapeutics are hampered by major issues such as off-target effects, immunogenicity, and uncertain delivery platforms. Over the past few decades, several innovative approaches have been established to minimize off-target effects, reduce immunostimulation, and provide efficient transfer to the target cells in which these molecules exert their function. Recent achievements have led to the testing of miRNA-based drugs in clinical trials, and these molecules may become next-generation therapeutics for medical intervention. Despite the achievement of exciting milestones, the dosage of miRNA administration remains unclear, and ways to address this issue are proposed. Elucidating the current status of the main factors of therapeutic miRNA would allow further developments and innovations to achieve safe therapeutic tools. This article is categorized under: RNA in Disease and Development > RNA in Disease Regulatory RNAs/RNAi/Riboswitches > RNAi: Mechanisms of Action.
Collapse
Affiliation(s)
- Abdul Fatah A Samad
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
| | - Mohd Farizal Kamaroddin
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
| |
Collapse
|
13
|
Sun X, Cui Z. Microbiological Nanotechnology. Nanomedicine (Lond) 2023. [DOI: 10.1007/978-981-16-8984-0_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
14
|
Virus-Like Particles as Nanocarriers for Intracellular Delivery of Biomolecules and Compounds. Viruses 2022; 14:v14091905. [PMID: 36146711 PMCID: PMC9503347 DOI: 10.3390/v14091905] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Virus-like particles (VLPs) are nanostructures assemble from viral proteins. Besides widely used for vaccine development, VLPs have also been explored as nanocarriers for cargo delivery as they combine the key advantages of viral and non-viral vectors. While it protects cargo molecules from degradation, the VLP has good cell penetrating property to mediate cargo passing the cell membrane and released into cells, making the VLP an ideal tool for intracellular delivery of biomolecules and drugs. Great progresses have been achieved and multiple challenges are still on the way for broad applications of VLP as delivery vectors. Here we summarize current advances and applications in VLP as a delivery vector. Progresses on delivery of different types of biomolecules as well as drugs by VLPs are introduced, and the strategies for cargo packaging are highlighted which is one of the key steps for VLP mediated intracellular delivery. Production and applications of VLPs are also briefly reviewed, with a discussion on future challenges in this rapidly developing field.
Collapse
|
15
|
Edwardson TGW, Levasseur MD, Tetter S, Steinauer A, Hori M, Hilvert D. Protein Cages: From Fundamentals to Advanced Applications. Chem Rev 2022; 122:9145-9197. [PMID: 35394752 DOI: 10.1021/acs.chemrev.1c00877] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Proteins that self-assemble into polyhedral shell-like structures are useful molecular containers both in nature and in the laboratory. Here we review efforts to repurpose diverse protein cages, including viral capsids, ferritins, bacterial microcompartments, and designed capsules, as vaccines, drug delivery vehicles, targeted imaging agents, nanoreactors, templates for controlled materials synthesis, building blocks for higher-order architectures, and more. A deep understanding of the principles underlying the construction, function, and evolution of natural systems has been key to tailoring selective cargo encapsulation and interactions with both biological systems and synthetic materials through protein engineering and directed evolution. The ability to adapt and design increasingly sophisticated capsid structures and functions stands to benefit the fields of catalysis, materials science, and medicine.
Collapse
Affiliation(s)
| | | | - Stephan Tetter
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Angela Steinauer
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Mao Hori
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| | - Donald Hilvert
- Laboratory of Organic Chemistry, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
16
|
Microbiological Nanotechnology. Nanomedicine (Lond) 2022. [DOI: 10.1007/978-981-13-9374-7_16-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
17
|
Veeranarayanan S, Azam AH, Kiga K, Watanabe S, Cui L. Bacteriophages as Solid Tumor Theragnostic Agents. Int J Mol Sci 2021; 23:402. [PMID: 35008840 PMCID: PMC8745063 DOI: 10.3390/ijms23010402] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 12/16/2022] Open
Abstract
Cancer, especially the solid tumor sub-set, poses considerable challenges to modern medicine owing to the unique physiological characteristics and substantial variations in each tumor's microenvironmental niche fingerprints. Though there are many treatment methods available to treat solid tumors, still a considerable loss of life happens, due to the limitation of treatment options and the outcomes of ineffective treatments. Cancer cells evolve with chemo- or radiation-treatment strategies and later show adaptive behavior, leading to failed treatment. These challenges demand tailored and individually apt personalized treatment methods. Bacteriophages (or phages) and phage-based theragnostic vectors are gaining attention in the field of modern cancer medicine, beyond their bactericidal ability. With the invention of the latest techniques to fine-tune phages, such as in the field of genetic engineering, synthetic assembly methods, phage display, and chemical modifications, noteworthy progress in phage vector research for safe cancer application has been realized, including use in pre-clinical studies. Herein, we discuss the distinct fingerprints of solid tumor physiology and the potential for bacteriophage vectors to exploit specific tumor features for improvised tumor theragnostic applications.
Collapse
Affiliation(s)
| | | | | | | | - Longzhu Cui
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke-shi 3290498, Japan; (S.V.); (A.H.A.); (K.K.); (S.W.)
| |
Collapse
|
18
|
Zhang J, Li D, Zhang R, Peng R, Li J. Delivery of microRNA-21-sponge and pre-microRNA-122 by MS2 virus-like particles to therapeutically target hepatocellular carcinoma cells. Exp Biol Med (Maywood) 2021; 246:2463-2472. [PMID: 34644206 DOI: 10.1177/15353702211035689] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
MicroRNAs are related to the development of hepatocellular carcinoma and can serve as potential therapeutic targets. Therapeutic strategies increasing tumor-suppressive microRNAs and reducing oncogenic microRNAs have been developed. Herein, the effects of simultaneously altering two microRNAs using MS2 virus-like particles were studied. The sequences of microRNA-21-sponge and pre-microRNA-122 were connected and cloned into a virus-like particle expression vector. Virus-like particles containing microRNA-21-sponge and pre-microRNA-122 sequences were prepared and crosslinked with a cell-specific peptide targeting hepatocellular carcinoma cells. Delivery effects were studied using RT-qPCR and functional assays to investigate the level of target mRNAs, cell toxicity, and the effects of proliferation, invasion, and migration. Virus-like particles delivered miR-21-sponge into cells, with the Ct value reaching 10 at most. The linked pre-miR-122 was processed into mature miR-122. The mRNA targets of miR-21 were derepressed as predicted and upregulated 1.2-2.8-fold, and the expression of proteins was elevated correspondingly. Proliferation, migration, and invasion of HCC cells were inhibited by miR-21-sponge. Simultaneous delivery of miR-21-sponge and miR-122 further decreased proliferation, migration, and invasion by up to 34%, 63%, and 65%, respectively. And the combination promoted the apoptosis of HCC cells. In conclusion, delivering miR-21-sponge and miR-122 using virus-like particles modified by cell-specific peptides is an effective and convenient strategy to correct microRNA dysregulation in hepatocellular carcinoma cells and is a promising therapeutic strategy for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Jiawei Zhang
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China.,Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing 100730, P.R. China
| | - Dandan Li
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China.,Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing 100730, P.R. China
| | - Rui Zhang
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China.,Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing 100730, P.R. China
| | - Rongxue Peng
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China.,Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing 100730, P.R. China
| | - Jinming Li
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China.,Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, Beijing 100730, P.R. China
| |
Collapse
|
19
|
Optimizing the synthesis and purification of MS2 virus like particles. Sci Rep 2021; 11:19851. [PMID: 34615923 PMCID: PMC8494748 DOI: 10.1038/s41598-021-98706-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/14/2021] [Indexed: 02/08/2023] Open
Abstract
Introducing bacteriophage MS2 virus-like particles (VLPs) as gene and drug delivery tools increases the demand for optimizing their production and purification procedure. PEG precipitation method is used efficiently to purify VLPs, while the effects of pH and different electrolytes on the stability, size, and homogeneity of purified MS2 VLPs, and the encapsulated RNA sequences remained to be elucidated. In this regard, a vector, capable of producing VLP with an shRNA packed inside was prepared. The resulting VLPs in different buffers/solutions were assessed for their size, polydispersity index, and ability to protect the enclosed shRNA. We report that among Tris, HEPES, and PBS, with or without NaNO3, and also NaNO3 alone in different pH and ionic concentrations, the 100 mM NaNO3-Tris buffer with pH:8 can be used as a new and optimal MS2 VLP production buffer, capable of inhibiting the VLPs aggregation. These VLPs show a size range of 27-30 nm and suitable homogeneity with minimum 12-month stability at 4 °C. Moreover, the resulting MS2 VLPs were highly efficient and stable for at least 48 h in conditions similar to in vivo. These features of MS2 VLPs produced in the newly introduced buffer make them an appropriate candidate for therapeutic agents' delivery.
Collapse
|
20
|
Hejtmánková A, Váňová J, Španielová H. Cell-penetrating peptides in the intracellular delivery of viral nanoparticles. VITAMINS AND HORMONES 2021; 117:47-76. [PMID: 34420585 DOI: 10.1016/bs.vh.2021.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell-penetrating peptides (CPPs) are a promising tool for the intracellular delivery of cargo. Due to their ability to cross membranes while also cotransporting various cargoes, they offer great potential for biomedical applications. Several CPPs have been derived from viral proteins with natural roles in the viral replication cycle that require them to breach or fuse to cellular membranes. Additionally, the ability of viruses to cross membranes makes viruses and virus-based particles a convenient model for research on nanoparticle delivery and nanoparticle-mediated gene therapy. In this chapter, we aim to characterize CPPs derived from both structural and nonstructural viral proteins. Their function as enhancers of viral infection and transduction by viral nanoparticles as well as the main features of viral CPPs employed in intracellular cargo delivery are summarized to emphasize their potential use in nanomedicine.
Collapse
Affiliation(s)
- Alžběta Hejtmánková
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Jana Váňová
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Hana Španielová
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic; Institute of Organic Chemistry and Biochemistry of the CAS, Prague, Czech Republic.
| |
Collapse
|
21
|
Carrion CC, Nasrollahzadeh M, Sajjadi M, Jaleh B, Soufi GJ, Iravani S. Lignin, lipid, protein, hyaluronic acid, starch, cellulose, gum, pectin, alginate and chitosan-based nanomaterials for cancer nanotherapy: Challenges and opportunities. Int J Biol Macromol 2021; 178:193-228. [PMID: 33631269 DOI: 10.1016/j.ijbiomac.2021.02.123] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/07/2021] [Accepted: 02/16/2021] [Indexed: 12/11/2022]
Abstract
Although nanotechnology-driven drug delivery systems are relatively new, they are rapidly evolving since the nanomaterials are deployed as effective means of diagnosis and delivery of assorted therapeutic agents to targeted intracellular sites in a controlled release manner. Nanomedicine and nanoparticulate drug delivery systems are rapidly developing as they play crucial roles in the development of therapeutic strategies for various types of cancer and malignancy. Nevertheless, high costs, associated toxicity and production of complexities are some of the critical barriers for their applications. Green nanomedicines have continually been improved as one of the viable approaches towards tumor drug delivery, thus making a notable impact on which considerably affect cancer treatment. In this regard, the utilization of natural and renewable feedstocks as a starting point for the fabrication of nanosystems can considerably contribute to the development of green nanomedicines. Nanostructures and biopolymers derived from natural and biorenewable resources such as proteins, lipids, lignin, hyaluronic acid, starch, cellulose, gum, pectin, alginate, and chitosan play vital roles in the development of cancer nanotherapy, imaging and management. This review uncovers recent investigations on diverse nanoarchitectures fabricated from natural and renewable feedstocks for the controlled/sustained and targeted drug/gene delivery systems against cancers including an outlook on some of the scientific challenges and opportunities in this field. Various important natural biopolymers and nanomaterials for cancer nanotherapy are covered and the scientific challenges and opportunities in this field are reviewed.
Collapse
Affiliation(s)
- Carolina Carrillo Carrion
- Department of Organic Chemistry, University of Córdoba, Campus de Rabanales, Edificio Marie Curie, Ctra Nnal IV-A Km. 396, E-14014 Cordoba, Spain
| | | | - Mohaddeseh Sajjadi
- Department of Chemistry, Faculty of Science, University of Qom, Qom 37185-359, Iran
| | - Babak Jaleh
- Department of Physics, Bu-Ali Sina University, 65174 Hamedan, Iran
| | | | - Siavash Iravani
- Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
22
|
Dasgupta I, Chatterjee A. Recent Advances in miRNA Delivery Systems. Methods Protoc 2021; 4:mps4010010. [PMID: 33498244 PMCID: PMC7839010 DOI: 10.3390/mps4010010] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 12/27/2022] Open
Abstract
MicroRNAs (miRNAs) represent a family of short non-coding regulatory RNA molecules that are produced in a tissue and time-specific manner to orchestrate gene expression post-transcription. MiRNAs hybridize to target mRNA(s) to induce translation repression or mRNA degradation. Functional studies have demonstrated that miRNAs are engaged in virtually every physiological process and, consequently, miRNA dysregulations have been linked to multiple human pathologies. Thus, miRNA mimics and anti-miRNAs that restore miRNA expression or downregulate aberrantly expressed miRNAs, respectively, are highly sought-after therapeutic strategies for effective manipulation of miRNA levels. In this regard, carrier vehicles that facilitate proficient and safe delivery of miRNA-based therapeutics are fundamental to the clinical success of these pharmaceuticals. Here, we highlight the strengths and weaknesses of current state-of-the-art viral and non-viral miRNA delivery systems and provide perspective on how these tools can be exploited to improve the outcomes of miRNA-based therapeutics.
Collapse
Affiliation(s)
- Ishani Dasgupta
- Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA 01605, USA;
| | - Anushila Chatterjee
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Correspondence:
| |
Collapse
|
23
|
Cell-penetrating peptides in oncologic pharmacotherapy: A review. Pharmacol Res 2020; 162:105231. [PMID: 33027717 DOI: 10.1016/j.phrs.2020.105231] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/23/2020] [Accepted: 09/30/2020] [Indexed: 01/10/2023]
Abstract
Cancer is the second leading cause of death in the world and its treatment is extremely challenging, mainly due to its complexity. Cell-Penetrating Peptides (CPPs) are peptides that can transport into the cell a wide variety of biologically active conjugates (or cargoes), and are, therefore, promising in the treatment and in the diagnosis of several types of cancer. Some notable examples are TAT and Penetratin, capable of penetrating the central nervous system (CNS) and, therefore, acting in cancers of this system, such as Glioblastoma Multiforme (GBM). These above-mentioned peptides, conjugated with traditional chemotherapeutic such as Doxorubicin (DOX) and Paclitaxel (PTX), have also been shown to induce apoptosis of breast and liver cancer cells, as well as in lung cancer cells, respectively. In other cancers, such as esophageal cancer, the attachment of Magainin 2 (MG2) to Bombesin (MG2B), another CPP, led to pronounced anticancer effects. Other examples are CopA3, that selectively decreased the viability of gastric cancer cells, and the CPP p28. Furthermore, in preclinical tests, the anti-tumor efficacy of this peptide was evaluated on human breast cancer, prostate cancer, ovarian cancer, and melanoma cells in vitro, leading to high expression of p53 and promoting cell cycle arrest. Despite the numerous in vitro and in vivo studies with promising results, and the increasing number of clinical trials using CPPs, few treatments reach the expected clinical efficacy. Usually, their clinical application is limited by its poor aqueous solubility, immunogenicity issues and dose-limiting toxicity. This review describes the most recent advances and innovations in the use of CPPs in several types of cancer, highlighting their crucial importance for various purposes, from therapeutic to diagnosis. Further clinical trials with these peptides are warranted to examine its effects on various types of cancer.
Collapse
|
24
|
Younis MA, Khalil IA, Harashima H. Gene Therapy for Hepatocellular Carcinoma: Highlighting the Journey from Theory to Clinical Applications. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Mahmoud A. Younis
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences Hokkaido University Kita‐12, Nishi‐6, Kita‐ku Sapporo 060‐0812 Japan
- Faculty of Pharmacy Assiut University Assiut 71526 Egypt
| | - Ikramy A. Khalil
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences Hokkaido University Kita‐12, Nishi‐6, Kita‐ku Sapporo 060‐0812 Japan
- Faculty of Pharmacy Assiut University Assiut 71526 Egypt
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences Hokkaido University Kita‐12, Nishi‐6, Kita‐ku Sapporo 060‐0812 Japan
| |
Collapse
|
25
|
Chung YH, Cai H, Steinmetz NF. Viral nanoparticles for drug delivery, imaging, immunotherapy, and theranostic applications. Adv Drug Deliv Rev 2020; 156:214-235. [PMID: 32603813 PMCID: PMC7320870 DOI: 10.1016/j.addr.2020.06.024] [Citation(s) in RCA: 238] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 02/06/2023]
Abstract
Viral nanoparticles (VNPs) encompass a diverse array of naturally occurring nanomaterials derived from plant viruses, bacteriophages, and mammalian viruses. The application and development of VNPs and their genome-free versions, the virus-like particles (VLPs), for nanomedicine is a rapidly growing. VLPs can encapsulate a wide range of active ingredients as well as be genetically or chemically conjugated to targeting ligands to achieve tissue specificity. VLPs are manufactured through scalable fermentation or molecular farming, and the materials are biocompatible and biodegradable. These properties have led to a wide range of applications, including cancer therapies, immunotherapies, vaccines, antimicrobial therapies, cardiovascular therapies, gene therapies, as well as imaging and theranostics. The use of VLPs as drug delivery agents is evolving, and sufficient research must continuously be undertaken to translate these therapies to the clinic. This review highlights some of the novel research efforts currently underway in the VNP drug delivery field in achieving this greater goal.
Collapse
Affiliation(s)
- Young Hun Chung
- Department of Bioengineering, University of California-San Diego, La Jolla, CA 92093, United States
| | - Hui Cai
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92093, United States
| | - Nicole F Steinmetz
- Department of Bioengineering, University of California-San Diego, La Jolla, CA 92093, United States; Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92093, United States; Department of Radiology, University of California-San Diego, La Jolla, CA 92093, United States; Moores Cancer Center, University of California-San Diego, La Jolla, CA 92093, United States; Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla, CA 92093, United States.
| |
Collapse
|
26
|
The Underlying Mechanisms of Noncoding RNAs in the Chemoresistance of Hepatocellular Carcinoma. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 21:13-27. [PMID: 32505000 PMCID: PMC7270498 DOI: 10.1016/j.omtn.2020.05.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/15/2020] [Accepted: 05/11/2020] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal human malignancies. Chemotherapeutic agents, such as sorafenib and lenvatinib, can improve the outcomes of HCC patients. Nevertheless, chemoresistance has become a major hurdle in the effective treatment of HCC. Noncoding RNAs (ncRNAs), including mircoRNAs (miRNAs), long ncRNAs (lncRNAs), and circular RNAs (circRNAs), have been demonstrated to participate in the onset and progression of HCC. Moreover, multiple lines of evidence have indicated that ncRNAs also play a pivotal role in HCC drug resistance. ncRNAs can regulate drug efflux and metabolism, glucose metabolism, cellular death pathways, and malignant characteristics in HCC. A deeper understanding of the molecular mechanisms responsible for ncRNA-mediated drug resistance in HCC will provide new opportunities for improving the treatment of HCC. In this review, we summarize recent findings on the molecular mechanisms by which ncRNAs regulate HCC chemoresistance, as well as their potential clinical implications in overcoming HCC chemoresistance.
Collapse
|
27
|
Demchuk AM, Patel TR. The biomedical and bioengineering potential of protein nanocompartments. Biotechnol Adv 2020; 41:107547. [PMID: 32294494 DOI: 10.1016/j.biotechadv.2020.107547] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 03/21/2020] [Accepted: 04/03/2020] [Indexed: 12/18/2022]
Abstract
Protein nanocompartments (PNCs) are self-assembling biological nanocages that can be harnessed as platforms for a wide range of nanobiotechnology applications. The most widely studied examples of PNCs include virus-like particles, bacterial microcompartments, encapsulin nanocompartments, enzyme-derived nanocages (such as lumazine synthase and the E2 component of the pyruvate dehydrogenase complex), ferritins and ferritin homologues, small heat shock proteins, and vault ribonucleoproteins. Structural PNC shell proteins are stable, biocompatible, and tolerant of both interior and exterior chemical or genetic functionalization for use as vaccines, therapeutic delivery vehicles, medical imaging aids, bioreactors, biological control agents, emulsion stabilizers, or scaffolds for biomimetic materials synthesis. This review provides an overview of the recent biomedical and bioengineering advances achieved with PNCs with a particular focus on recombinant PNC derivatives.
Collapse
Affiliation(s)
- Aubrey M Demchuk
- Department of Neuroscience, University of Lethbridge, 4401 University Drive West, Lethbridge, AB, Canada.
| | - Trushar R Patel
- Alberta RNA Research and Training Institute, Department of Chemistry and Biochemistry, University of Lethbridge, 4401 University Drive West, Lethbridge, AB, Canada; Department of Microbiology, Immunology and Infectious Diseases, Cumming, School of Medicine, University of Calgary, 2500 University Dr. N.W., Calgary, AB T2N 1N4, Canada; Li Ka Shing Institute of Virology and Discovery Lab, Faculty of Medicine & Dentistry, University of Alberta, 6-010 Katz Center for Health Research, Edmonton, AB T6G 2E1, Canada.
| |
Collapse
|
28
|
Taylor RE, Zahid M. Cell Penetrating Peptides, Novel Vectors for Gene Therapy. Pharmaceutics 2020; 12:E225. [PMID: 32138146 PMCID: PMC7150854 DOI: 10.3390/pharmaceutics12030225] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/27/2020] [Accepted: 03/01/2020] [Indexed: 12/31/2022] Open
Abstract
Cell penetrating peptides (CPPs), also known as protein transduction domains (PTDs), first identified ~25 years ago, are small, 6-30 amino acid long, synthetic, or naturally occurring peptides, able to carry variety of cargoes across the cellular membranes in an intact, functional form. Since their initial description and characterization, the field of cell penetrating peptides as vectors has exploded. The cargoes they can deliver range from other small peptides, full-length proteins, nucleic acids including RNA and DNA, liposomes, nanoparticles, and viral particles as well as radioisotopes and other fluorescent probes for imaging purposes. In this review, we will focus briefly on their history, classification system, and mechanism of transduction followed by a summary of the existing literature on use of CPPs as gene delivery vectors either in the form of modified viruses, plasmid DNA, small interfering RNA, oligonucleotides, full-length genes, DNA origami or peptide nucleic acids.
Collapse
Affiliation(s)
- Rebecca E. Taylor
- Mechanical Engineering, Biomedical Engineering and Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Maliha Zahid
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| |
Collapse
|
29
|
Withers SB, Dewhurst T, Hammond C, Topham CH. MiRNAs as Novel Adipokines: Obesity-Related Circulating MiRNAs Influence Chemosensitivity in Cancer Patients. Noncoding RNA 2020; 6:ncrna6010005. [PMID: 31979312 PMCID: PMC7151601 DOI: 10.3390/ncrna6010005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/08/2020] [Accepted: 01/11/2020] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue is an endocrine organ, capable of regulating distant physiological processes in other tissues via the release of adipokines into the bloodstream. Recently, circulating adipose-derived microRNAs (miRNAs) have been proposed as a novel class of adipokine, due to their capacity to regulate gene expression in tissues other than fat. Circulating levels of adipokines are known to be altered in obese individuals compared with typical weight individuals and are linked to poorer health outcomes. For example, obese individuals are known to be more prone to the development of some cancers, and less likely to achieve event-free survival following chemotherapy. The purpose of this review was twofold; first to identify circulating miRNAs which are reproducibly altered in obesity, and secondly to identify mechanisms by which these obesity-linked miRNAs might influence the sensitivity of tumors to treatment. We identified 8 candidate circulating miRNAs with altered levels in obese individuals (6 increased, 2 decreased). A second literature review was then performed to investigate if these candidates might have a role in mediating resistance to cancer treatment. All of the circulating miRNAs identified were capable of mediating responses to cancer treatment at the cellular level, and so this review provides novel insights which can be used by future studies which aim to improve obese patient outcomes.
Collapse
Affiliation(s)
- Sarah B. Withers
- Biomedical Research Centre, School of Science, Engineering and Environment, Peel Building, University of Salford, Salford M5 4WT, UK; (S.B.W.); (T.D.); (C.H.)
- Salford Royal Foundation Trust, Clinical Sciences Building, Stott Lane, Salford M6 8HD, UK
| | - Toni Dewhurst
- Biomedical Research Centre, School of Science, Engineering and Environment, Peel Building, University of Salford, Salford M5 4WT, UK; (S.B.W.); (T.D.); (C.H.)
| | - Chloe Hammond
- Biomedical Research Centre, School of Science, Engineering and Environment, Peel Building, University of Salford, Salford M5 4WT, UK; (S.B.W.); (T.D.); (C.H.)
| | - Caroline H. Topham
- Biomedical Research Centre, School of Science, Engineering and Environment, Peel Building, University of Salford, Salford M5 4WT, UK; (S.B.W.); (T.D.); (C.H.)
- Correspondence: ; Tel.: +44-(0)-161-295-4292
| |
Collapse
|
30
|
Wang Z, Zhi K, Ding Z, Sun Y, Li S, Li M, Pu K, Zou J. Emergence in protein derived nanomedicine as anticancer therapeutics: More than a tour de force. Semin Cancer Biol 2020; 69:77-90. [PMID: 31962173 DOI: 10.1016/j.semcancer.2019.11.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/14/2019] [Accepted: 11/30/2019] [Indexed: 12/12/2022]
Abstract
Cancer has thwarted as a major health problem affecting the global population. With an alarming increase in the patient population suffering from diverse varieties of cancers, the global demographic data predicts sharp escalation in the number of cancer patients. This can be expected to reach 420 million cases by 2025. Among the diverse types of cancers, the most frequently diagnosed cancers are the breast, colorectal, prostate and lung cancer. From years, conventional treatment approaches like surgery, chemotherapy and radiation therapy have been practiced. In the past few years, increasing research on molecular level diagnosis and treatment of cancers have significantly changed the realm of cancer treatment. Lately, uses of advanced chemotherapy and immunotherapy like treatments have gained significant progress in the cancer therapy, but these approaches have several limitations on their safety and toxicity. This has generated lot of momentum for the evolution of new drug delivery approaches for the effective delivery of anticancer therapeutics, which may improve the pharmacokinetic and pharmacodynamic effect of the drugs along with significant reduction in the side effects. In this regard, the protein-based nano-medicines have gained wider attention in the management of cancer. Proteins are organic macromolecules essential, for life and have quite well explored in developing the nano-carriers. Furthermore, it provides passive or active tumour cell targeted delivery, by using protein based nanovesicles or virus like structures, antibody drug conjugates, viral particles, etc. Moreover, by utilizing various formulation strategies, both the animal and plant derived proteins can be converted to produce self-assembled virus like nano-metric structures with high efficiency in targeting the metastatic cancer cells. Therefore, the present review extensively discusses the applications of protein-based nano-medicine with special emphasis on intracellular delivery/drug targeting ability for anticancer drugs.
Collapse
Affiliation(s)
- Zhenchang Wang
- Department of Spleen, Stomach and Liver Diseases, Guangxi International Zhuang Medical Hospital, Guangxi, Nanning, 530201, China
| | - Kangkang Zhi
- Vascular Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Zhongyang Ding
- General Surgery, Wuxi Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Jiangsu, Nanjing, 214023, China
| | - Yi Sun
- Oncology Department, Guizhou Provincial People's Hospital, Guizhou, Guiyang, 550002, China
| | - Shuang Li
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Jiamusi University, Heilongjiang, Jiamu, 154003, China
| | - Manyuan Li
- Laboratory Department, Jinzhou Maternal and Infant Hospital, Liaoning, Jinzhou, 121000, China
| | - Kefeng Pu
- Suzhou Institute of Nanotechnology and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu, 215123, China
| | - Jun Zou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China.
| |
Collapse
|
31
|
Abstract
ssRNA phages belonging to the family Leviviridae are among the tiniest viruses, infecting various Gram-negative bacteria by adsorption to their pilus structures. Due to their simplicity, they have been intensively studied as models for understanding various problems in molecular biology and virology. Several of the studied ssRNA characteristics, such as coat protein–RNA interactions and the ability to readily form virus-like particles in recombinant expression systems, have fueled many practical applications such as RNA labeling and tracking systems and vaccine development. In this chapter, we review the life cycle, structure and applications of these small yet fascinating viruses.
Collapse
|
32
|
Sandra F, Khaliq NU, Sunna A, Care A. Developing Protein-Based Nanoparticles as Versatile Delivery Systems for Cancer Therapy and Imaging. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E1329. [PMID: 31527483 PMCID: PMC6781024 DOI: 10.3390/nano9091329] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/04/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023]
Abstract
In recent years, it has become apparent that cancer nanomedicine's reliance on synthetic nanoparticles as drug delivery systems has resulted in limited clinical outcomes. This is mostly due to a poor understanding of their "bio-nano" interactions. Protein-based nanoparticles (PNPs) are rapidly emerging as versatile vehicles for the delivery of therapeutic and diagnostic agents, offering a potential alternative to synthetic nanoparticles. PNPs are abundant in nature, genetically and chemically modifiable, monodisperse, biocompatible, and biodegradable. To harness their full clinical potential, it is important for PNPs to be accurately designed and engineered. In this review, we outline the recent advancements and applications of PNPs in cancer nanomedicine. We also discuss the future directions for PNP research and what challenges must be overcome to ensure their translation into the clinic.
Collapse
Affiliation(s)
- Febrina Sandra
- Department of Molecular Sciences, Macquarie University, Sydney 2109, Australia.
| | - Nisar Ul Khaliq
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, Korea.
| | - Anwar Sunna
- Department of Molecular Sciences, Macquarie University, Sydney 2109, Australia.
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Macquarie University, Sydney, NSW 2109, Australia.
| | - Andrew Care
- Department of Molecular Sciences, Macquarie University, Sydney 2109, Australia.
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Macquarie University, Sydney, NSW 2109, Australia.
| |
Collapse
|
33
|
Reghupaty SC, Sarkar D. Current Status of Gene Therapy in Hepatocellular Carcinoma. Cancers (Basel) 2019; 11:cancers11091265. [PMID: 31466358 PMCID: PMC6770843 DOI: 10.3390/cancers11091265] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/21/2019] [Accepted: 08/27/2019] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer and the second leading cause of cancer related deaths world-wide. Liver transplantation, surgical resection, trans-arterial chemoembolization, and radio frequency ablation are effective strategies to treat early stage HCC. Unfortunately, HCC is usually diagnosed at an advanced stage and there are not many treatment options for late stage HCC. First-line therapy for late stage HCC includes sorafenib and lenvatinib. However, these treatments provide only an approximate three month increase in survival. Besides, they cannot specifically target cancer cells that lead to a wide array of side effects. Patients on these drugs develop resistance within a few months and have to rely on second-line therapy that includes regorafenib, pembrolizumab, nivolumab, and cabometyx. These disadvantages make gene therapy approach to treat HCC an attractive option. The two important questions that researchers have been trying to answer in the last 2-3 decades are what genes should be targeted and what delivery systems should be used. The objective of this review is to analyze the changing landscape of HCC gene therapy, with a focus on these two questions.
Collapse
Affiliation(s)
- Saranya Chidambaranathan Reghupaty
- Department of Human and Molecular Genetics, Massey Cancer Center, VCU Institute of Molecular Medicine (VIMM), Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Massey Cancer Center, VCU Institute of Molecular Medicine (VIMM), Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
34
|
The Utilization of Cell-Penetrating Peptides in the Intracellular Delivery of Viral Nanoparticles. MATERIALS 2019; 12:ma12172671. [PMID: 31443361 PMCID: PMC6747576 DOI: 10.3390/ma12172671] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/16/2019] [Accepted: 08/21/2019] [Indexed: 01/04/2023]
Abstract
Viral particles (VPs) have evolved so as to efficiently enter target cells and to deliver their genetic material. The current state of knowledge allows us to use VPs in the field of biomedicine as nanoparticles that are safe, easy to manipulate, inherently biocompatible, biodegradable, and capable of transporting various cargoes into specific cells. Despite the fact that these virus-based nanoparticles constitute the most common vectors used in clinical practice, the need remains for further improvement in this area. The aim of this review is to discuss the potential for enhancing the efficiency and versatility of VPs via their functionalization with cell-penetrating peptides (CPPs), short peptides that are able to translocate across cellular membranes and to transport various substances with them. The review provides and describes various examples of and means of exploitation of CPPs in order to enhance the delivery of VPs into permissive cells and/or to allow them to enter a broad range of cell types. Moreover, it is possible that CPPs are capable of changing the immunogenic properties of VPs, which could lead to an improvement in their clinical application. The review also discusses strategies aimed at the modification of VPs by CPPs so as to create a useful cargo delivery tool.
Collapse
|
35
|
Casanova I, Unzueta U, Arroyo-Solera I, Céspedes MV, Villaverde A, Mangues R, Vazquez E. Protein-driven nanomedicines in oncotherapy. Curr Opin Pharmacol 2019; 47:1-7. [PMID: 30685732 DOI: 10.1016/j.coph.2018.12.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/20/2018] [Indexed: 12/18/2022]
Abstract
Proteins are organic macromolecules essential in life but exploited, mainly in recombinant versions, as drugs or vaccine components, among other uses in industry or biomedicine. In oncology, individual proteins or supramolecular complexes have been tailored as small molecular weight drug carriers for passive or active tumor cell-targeted delivery, through the de novo design of appropriate drug stabilizing vehicles, or by generating constructs with different extents of mimesis of natural cell-targeted entities, such as viruses. In most of these approaches, a convenient nanoscale size is achieved through the oligomeric organization of the protein component in the drug conjugate. Among the different taken strategies, highly cytotoxic proteins such as microbial or plant toxins have been conveniently engineered to self-assemble as self-delivered virus-like, nanometric structures, chemically homogeneous that target metastatic cancer stem cells for the destruction of metastasis in absence of any partner vehicle.
Collapse
Affiliation(s)
- Isolda Casanova
- Biomedical Research Institute Sant Pau (IIB-Sant Pau) and Josep Carreras Research Institute, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain
| | - Ugutz Unzueta
- Biomedical Research Institute Sant Pau (IIB-Sant Pau) and Josep Carreras Research Institute, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
| | - Irene Arroyo-Solera
- Biomedical Research Institute Sant Pau (IIB-Sant Pau) and Josep Carreras Research Institute, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain
| | - Maria Virtudes Céspedes
- Biomedical Research Institute Sant Pau (IIB-Sant Pau) and Josep Carreras Research Institute, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain
| | - Antonio Villaverde
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain; Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain.
| | - Ramon Mangues
- Biomedical Research Institute Sant Pau (IIB-Sant Pau) and Josep Carreras Research Institute, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain.
| | - Esther Vazquez
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Bellaterra, 08193, Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain; Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain
| |
Collapse
|
36
|
miR-122 and hepatocellular carcinoma: from molecular biology to therapeutics. EBioMedicine 2018; 37:17-18. [PMID: 30337246 PMCID: PMC6284505 DOI: 10.1016/j.ebiom.2018.10.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 10/10/2018] [Indexed: 12/12/2022] Open
|
37
|
Donaldson B, Lateef Z, Walker GF, Young SL, Ward VK. Virus-like particle vaccines: immunology and formulation for clinical translation. Expert Rev Vaccines 2018; 17:833-849. [PMID: 30173619 PMCID: PMC7103734 DOI: 10.1080/14760584.2018.1516552] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Virus-like particle (VLP) vaccines face significant challenges in their translation from laboratory models, to routine clinical administration. While some VLP vaccines thrive and are readily adopted into the vaccination schedule, others are restrained by regulatory obstacles, proprietary limitations, or finding their niche amongst the crowded vaccine market. Often the necessity to supplant an existing vaccination regimen possesses an immediate obstacle for the development of a VLP vaccine, despite any preclinical advantages identified over the competition. Novelty, adaptability and formulation compatibility may prove invaluable in helping place VLP vaccines at the forefront of vaccination technology. AREAS COVERED The purpose of this review is to outline the diversity of VLP vaccines, VLP-specific immune responses, and to explore how modern formulation and delivery techniques can enhance the clinical relevance and overall success of VLP vaccines. EXPERT COMMENTARY The role of formation science, with an emphasis on the diversity of immune responses induced by VLP, is underrepresented amongst clinical trials for VLP vaccines. Harnessing such diversity, particularly through the use of combinations of select excipients and adjuvants, will be paramount in the development of VLP vaccines.
Collapse
Affiliation(s)
- Braeden Donaldson
- a Department of Microbiology and Immunology , School of Biomedical Sciences, University of Otago , Dunedin , New Zealand.,b Department of Pathology , Dunedin School of Medicine, University of Otago , Dunedin , New Zealand
| | - Zabeen Lateef
- c Department of Pharmacology and Toxicology , School of Biomedical Sciences, University of Otago , Dunedin , New Zealand
| | - Greg F Walker
- d School of Pharmacy , University of Otago , Dunedin , New Zealand
| | - Sarah L Young
- b Department of Pathology , Dunedin School of Medicine, University of Otago , Dunedin , New Zealand
| | - Vernon K Ward
- a Department of Microbiology and Immunology , School of Biomedical Sciences, University of Otago , Dunedin , New Zealand
| |
Collapse
|
38
|
Diaz D, Care A, Sunna A. Bioengineering Strategies for Protein-Based Nanoparticles. Genes (Basel) 2018; 9:E370. [PMID: 30041491 PMCID: PMC6071185 DOI: 10.3390/genes9070370] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/16/2018] [Accepted: 07/17/2018] [Indexed: 12/16/2022] Open
Abstract
In recent years, the practical application of protein-based nanoparticles (PNPs) has expanded rapidly into areas like drug delivery, vaccine development, and biocatalysis. PNPs possess unique features that make them attractive as potential platforms for a variety of nanobiotechnological applications. They self-assemble from multiple protein subunits into hollow monodisperse structures; they are highly stable, biocompatible, and biodegradable; and their external components and encapsulation properties can be readily manipulated by chemical or genetic strategies. Moreover, their complex and perfect symmetry have motivated researchers to mimic their properties in order to create de novo protein assemblies. This review focuses on recent advances in the bioengineering and bioconjugation of PNPs and the implementation of synthetic biology concepts to exploit and enhance PNP's intrinsic properties and to impart them with novel functionalities.
Collapse
Affiliation(s)
- Dennis Diaz
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
| | - Andrew Care
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Macquarie University, Sydney, NSW 2109, Australia.
| | - Anwar Sunna
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Macquarie University, Sydney, NSW 2109, Australia.
- Biomolecular Discovery and Design Research Centre, Macquarie University, Sydney, NSW 2109, Australia.
| |
Collapse
|
39
|
Zheng Q, Lin Z, Xu J, Lu Y, Meng Q, Wang C, Yang Y, Xin X, Li X, Pu H, Gui X, Li T, Xiong W, Lu D. Long noncoding RNA MEG3 suppresses liver cancer cells growth through inhibiting β-catenin by activating PKM2 and inactivating PTEN. Cell Death Dis 2018; 9:253. [PMID: 29449541 PMCID: PMC5833746 DOI: 10.1038/s41419-018-0305-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 12/13/2022]
Abstract
Maternally expressed gene 3 (MEG3) encodes an lncRNA which is suggested to function as a tumor suppressor and has been showed to involve in a variety of cancers. Herein, our findings demonstrate that MEG3 inhibits the malignant progression of liver cancer cells in vitro and in vivo. Mechanistically, MEG3 promotes the expression and maturition of miR122 which targets PKM2. Therefore, MEG3 decreases the expression and nuclear location of PKM2 dependent on miR122. Furthermore, MEG3 also inhibits CyclinD1 and C-Myc via PKM2 in liver cancer cells. On the other hand, MEG3 promotes β-catenin degradation through ubiquitin-proteasome system dependent on PTEN. Strikingly, MEG3 inhibits β-catenin activity through PKM2 reduction and PTEN increase. Significantly, we also found that excessive β-catenin abrogated the effect of MEG3 in liver cancer. In conclusion, our study for the first time demonstrates that MEG3 acts as a tumor suppressor by negatively regulating the activity of the PKM2 and β-catenin signaling pathway in hepatocarcinogenesis and could provide potential therapeutic targets for the treatment of liver cancer.
Collapse
Affiliation(s)
- Qidi Zheng
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Zhuojia Lin
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Jie Xu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Yanan Lu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Qiuyu Meng
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Chen Wang
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Yuxin Yang
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Xiaoru Xin
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Xiaonan Li
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Hu Pu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Xin Gui
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Tianming Li
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China
| | - Wujun Xiong
- Department of Hepatology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
| | - Dongdong Lu
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, 200092, Shanghai, China.
| |
Collapse
|
40
|
Li Y, Bai H, Wang H, Shen Y, Tang G, Ping Y. Reactive oxygen species (ROS)-responsive nanomedicine for RNAi-based cancer therapy. NANOSCALE 2017; 10:203-214. [PMID: 29210417 DOI: 10.1039/c7nr06689a] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Although much effort has been dedicated to the development of efficient siRNA delivery for cancer therapy, delivery nanomaterials that can particularly respond to reactive oxygen species (ROS), which are overproduced in the tissue and mitochondria of cancer cells, are still rare for the clinical translation of RNA interference (RNAi)-based therapy. To this end, we developed a ROS-responsive boronic vehicle with a lipid envelope for systemic vascular endothelial growth factor (VEGF) siRNA delivery so as to improve RNAi cancer therapy. We found that the efficiency of siRNA delivery largely relied on the ROS responsiveness of the carrier we have developed to mediate timely siRNA release, the PEG-functionalized lipid layer to shield the surface charge of polyplexes as well as the ability of the phenylboronic moiety to stabilize siRNA. The unique carrier nanostructure provides the efficient systemic transportation of siRNA to the tumor site for effective knockdown of the VEGF, which resulted in a significant antiangiogenesis effect and the effective inhibition of tumor growth in vivo. The current study defines a new systemic delivery strategy for siRNA by cooperatively integrating multifunctional lipid coatings with the ROS-responsive boronic polymer, which may potentially benefit RNAi-based therapy in the dawning era of precision nanomedicine for cancer therapy.
Collapse
Affiliation(s)
- Yang Li
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou 310028, P. R. China.
| | | | | | | | | | | |
Collapse
|
41
|
One-plasmid double-expression His-tag system for rapid production and easy purification of MS2 phage-like particles. Sci Rep 2017; 7:17501. [PMID: 29235545 PMCID: PMC5727534 DOI: 10.1038/s41598-017-17951-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 11/30/2017] [Indexed: 12/18/2022] Open
Abstract
MS2 phage-like particles (MS2 PLP) are artificially constructed pseudo-viral particles derived from bacteriophage MS2. They are able to carry a specific single stranded RNA (ssRNA) sequence of choice inside their capsid, thus protecting it against the effects of ubiquitous nucleases. Such particles are able to mimic ssRNA viruses and, thus, may serve as the process control for molecular detection and quantification of such agents in several kinds of matrices, vaccines and vaccine candidates, drug delivery systems, and systems for the display of immunologically active peptides or nanomachines. Currently, there are several different in vivo plasmid-driven packaging systems for production of MS2 PLP. In order to combine all the advantages of the available systems and to upgrade and simplify the production and purification of MS2 PLP, a one-plasmid double-expression His-tag system was designed. The described system utilizes a unique fusion insertional mutation enabling purification of particles using His-tag affinity. Using this new production system, highly pure MS2 PLP can be quickly produced and purified by a fast performance liquid chromatography (FPLC) approach. The system can be easily adapted to produce other MS2 PLP with different properties.
Collapse
|
42
|
Collado Camps E, Brock R. An opportunistic route to success: Towards a change of paradigm to fully exploit the potential of cell-penetrating peptides. Bioorg Med Chem 2017; 26:2780-2787. [PMID: 29157727 DOI: 10.1016/j.bmc.2017.11.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/27/2017] [Accepted: 11/02/2017] [Indexed: 01/06/2023]
Abstract
About 25years ago it was demonstrated that certain peptides possess the ability to cross the plasma membrane. This led to the development of cell-penetrating peptides (CPPs) as vectors to mediate the cellular entry of (macro-)molecules that do not show cell entry by themselves. Nonetheless, in spite of an early bloom of promising pre-clinical studies, not a single CPP-based drug has been approved, yet. It is a paradigm in CPP research that the peptides are taken up by virtually all cells. In exploratory research and early preclinical development, this assumption guides the choice of the therapeutic target. However, while this indiscriminatory uptake may be the case for tissue culture experiments, in an organism this is clearly not the case. Biodistribution analyses demonstrate that CPPs only target a very limited number of cells and many tissues are hardly reached at all. Here, we review biodistribution analyses of CPPs and CPP-based drug delivery systems. Based on this analysis we propose a paradigm change towards a more opportunistic approach in CPP research. The application of CPPs should focus on those pathophysiologies for which the relevant target cells have been shown to be reached in vivo.
Collapse
Affiliation(s)
- Estel Collado Camps
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Roland Brock
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands.
| |
Collapse
|
43
|
Liu L, Cai X, Liu E, Tian X, Tian C. MicroRNA-18a promotes proliferation and metastasis in hepatocellular carcinoma via targeting KLF4. Oncotarget 2017; 8:68263-68269. [PMID: 28978114 PMCID: PMC5620254 DOI: 10.18632/oncotarget.19293] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 06/19/2017] [Indexed: 01/02/2023] Open
Abstract
MicroRNAs (miRNAs) are short, non-coding and endogenous RNAs that played as important roles in the proliferation and metastasis of tumors. In this study, we determined the role of miR-18a in the regulation of HCC cell motility. We showed that miR-18a expression was upregulated in human HCC tissues and cell lines. Moreover, Elevated expression of miR-18a promoted the HCC cell proliferation and migration. KLF4 was identified as a direct target of miR-18a in HCC cells. Furthermore, overexpression of KLF4 attenuated the effects of miR-18a on the regulation of HCC cell motility. The expression of KLF4 was negatively associated with the expression of miR-18a expression in HCC tissues. We also showed that the cell cycle inhibitor p21 was aberrantly downregulated in HCC cells, whereas this inhibition was reversed by miR-18a inhibitor. These data indicated that miR-18a may play a positive role in hepatocellular carcinoma by promoting the proliferation and migration of HCC cells through targeting KLF4 as well as downstream p21.
Collapse
Affiliation(s)
- Li Liu
- Department of Medicine & Appliance, Yunyan District Market Supervision and Administration Bureau, Guizhou 550001, China
| | - Xun Cai
- Department of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Enqiang Liu
- Department of Oncology, Qianjiang Central Hospital of Chongqing Municipality, Chongqing 409000, China
| | - Xia Tian
- Department of Nuclear Medicine, Guizhou Provincial People’s Hospital, Guizhou 550000, China
| | - Chuan Tian
- Department of Nuclear Medicine, Guizhou Provincial People’s Hospital, Guizhou 550000, China
| |
Collapse
|
44
|
Yang J, Yuan Y, Yang X, Hong Z, Yang L. Decreased expression of microRNA-122 is associated with an unfavorable prognosis in childhood acute myeloid leukemia and function analysis indicates a therapeutic potential. Pathol Res Pract 2017; 213:1166-1172. [PMID: 28822593 DOI: 10.1016/j.prp.2017.06.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 05/15/2017] [Accepted: 06/25/2017] [Indexed: 12/15/2022]
Abstract
MicroRNA (miR)-122 functions as a tumor suppressor in various human cancers. However, its involvement in childhood acute myeloid leukemia (AML) remains unknown. In this study, quantitative real-time PCR assay demonstrated that miR-122 expression in bone marrow specimens from AML children were significantly lower than that in non-malignant controls (P<0.001). Statistically, AML children with low miR-122 expression more frequently had large white blood cell count (P=0.022), French-American-British classification subtype M7 (P<0.001), unfavorable cytogenetics (P=0.002) and day 7 response to the treatment (P=0.036), short relapse-free (P=0.001) and overall (P=0.008) survivals than those with high expression. Multivariate analysis also determined that miR-122 expression was an independent prognostic factor for both relapse-free and overall survivals. Functionally, the enforced expression of miR-122 in AML cell lines efficiently suppressed cell proliferation and reduced the ratio of S-phase cells in vitro (all P<0.05). In conclusion, the abnormal expression of miR-122 may be a marker of the aggressive progression in childhood AML. Importantly, its downregulation may serve as a prognostic factor to predict poor outcome. Our study also reveal that miR-122 may function as a tumor suppressor in childhood AML, highlighting a new therapeutic strategy for this malignancy.
Collapse
Affiliation(s)
- Juan Yang
- Department of Pediatrics, Huai'an Hospital Affiliated to Xuzhou Medical College and Huai'an Second People's Hospital, 62 Huaihai Road South, Huai'an 223002, China
| | - Yufang Yuan
- Department of Pediatrics, Huai'an Hospital Affiliated to Xuzhou Medical College and Huai'an Second People's Hospital, 62 Huaihai Road South, Huai'an 223002, China
| | - Xiaochun Yang
- Department of Pediatrics, Huai'an Hospital Affiliated to Xuzhou Medical College and Huai'an Second People's Hospital, 62 Huaihai Road South, Huai'an 223002, China
| | - Ze Hong
- Department of Pediatrics, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing Road West, Huai'an, Jiangsu 223300, China
| | - Lijuan Yang
- Department of Pediatrics, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing Road West, Huai'an, Jiangsu 223300, China.
| |
Collapse
|
45
|
Sun Y, Sun Y, Zhao R. Establishment of MicroRNA delivery system by PP7 bacteriophage-like particles carrying cell-penetrating peptide. J Biosci Bioeng 2017; 124:242-249. [PMID: 28442387 DOI: 10.1016/j.jbiosc.2017.03.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 03/15/2017] [Accepted: 03/21/2017] [Indexed: 02/08/2023]
Abstract
MicroRNAs have great therapeutic potential in cancer and other diseases. However, their instability and low in vivo delivery efficiency limits their application. Recombinant PP7 bacteriophage-based virus-like particles (VLPs) could protect microRNAs against rapid degradation by RNase by packaging specific exogenous pre-microRNAs using the pac site. Insertion of a cell-penetrating peptide (CPP) into the AB-loop of VLPs could significantly improve the delivery efficiency of microRNAs into mammalian cells. Unlike other microRNA delivery methods (viral or non-viral vectors), recombinant PP7 VLPs carrying a CPP and microRNA could be efficiently expressed in Escherichia coli using the one-plasmid double expression system. Here we showed that PP7 VLPs carrying a CPP penetrated hepatoma SK-HEP-1 cells and delivered the pre-microRNA-23b, which was processed into a mature product within 24 h; a concentration of 10 nM was sufficient for the inhibition of hepatoma cell migration via the downregulation of liver-intestine cadherin expression. Furthermore, PP7 VLPs carrying a CPP and a pre-microRNA were not infectious, replicative, or cytotoxic. Therefore, recombinant PP7 VLPs can be used for simultaneous and targeted delivery of both microRNAs and peptides because of their ability to package specific exogenous RNA using the pac site and to display peptides.
Collapse
Affiliation(s)
- Yanli Sun
- Institute of Nanomedicine Technology, Department of Laboratory Medicine, Institutional Key Laboratory of Clinical Laboratory Diagnostics, 12th 5-Year Project of Shandong Province, Key Discipline of Clinical Laboratory Medicine of Shandong Province, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang 261053, China.
| | - Yanhua Sun
- Department of Hematology, Weifang People's Hospital, Weifang 261000, China
| | - Ronglan Zhao
- Institute of Nanomedicine Technology, Department of Laboratory Medicine, Institutional Key Laboratory of Clinical Laboratory Diagnostics, 12th 5-Year Project of Shandong Province, Key Discipline of Clinical Laboratory Medicine of Shandong Province, Affiliated Hospital of Weifang Medical University, Weifang Medical University, Weifang 261053, China
| |
Collapse
|
46
|
Lin G, Zhang K, Zhang D, Han Y, Xie J, Li J. Fast preparation of a long chimeric armored RNA as controls for external quality assessment for molecular detection of Zika virus. Clin Chim Acta 2017; 466:138-144. [PMID: 28111270 DOI: 10.1016/j.cca.2017.01.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 01/18/2017] [Accepted: 01/19/2017] [Indexed: 01/31/2023]
Abstract
BACKGROUND The emergence of Zika virus demands accurate laboratory diagnostics. Nucleic acid testing is currently the definitive method for diagnosis of Zika infection. In 2016, an external quality assurance (EQA) for assessing the quality of molecular testing of Zika virus was carried out in China. METHODS A single armored RNA encapsulating a 4942-nucleotides (nt) long specific RNA sequence of Zika virus was prepared and used as positive samples. A pre-tested EQA panel, consisting of 4 negative and 6 positive samples with different concentrations of armored RNA, was distributed to 38 laboratories that perform molecular detection of Zika virus. RESULTS A total of 39 data sets (1 laboratory used two test kits in parallel), produced by using commercial (n=38) or laboratory developed (n=1) quantitative reverse-transcriptase PCR (qRT-PCR) kits, were received. Of these, 35 (89.7%) had correct results for all 10 samples, and 4 (10.3%) reported at least 1 error (11 in total). The testing errors were all false-negatives, highlighting the need of improvements in detecting sensitivity. CONCLUSIONS The EQA reveals that the majority of participating laboratories are proficient in molecular testing of Zika virus.
Collapse
Affiliation(s)
- Guigao Lin
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing, China.
| | - Kuo Zhang
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing, China.
| | - Dong Zhang
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing, China.
| | - Yanxi Han
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing, China.
| | - Jiehong Xie
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing, China.
| | - Jinming Li
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, PR China; Beijing Engineering Research Center of Laboratory Medicine, Beijing, China.
| |
Collapse
|