1
|
Hheidari A, Mohammadi J, Ghodousi M, Mahmoodi M, Ebrahimi S, Pishbin E, Rahdar A. Metal-based nanoparticle in cancer treatment: lessons learned and challenges. Front Bioeng Biotechnol 2024; 12:1436297. [PMID: 39055339 PMCID: PMC11269265 DOI: 10.3389/fbioe.2024.1436297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024] Open
Abstract
Cancer, being one of the deadliest diseases, poses significant challenges despite the existence of traditional treatment approaches. This has led to a growing demand for innovative pharmaceutical agents that specifically target cancer cells for effective treatment. In recent years, the use of metal nanoparticles (NPs) as a promising alternative to conventional therapies has gained prominence in cancer research. Metal NPs exhibit unique properties that hold tremendous potential for various applications in cancer treatment. Studies have demonstrated that certain metals possess inherent or acquired anticancer capabilities through their surfaces. These properties make metal NPs an attractive focus for therapeutic development. In this review, we will investigate the applicability of several distinct classes of metal NPs for tumor targeting in cancer treatment. These classes may include gold, silver, iron oxide, and other metals with unique properties that can be exploited for therapeutic purposes. Additionally, we will provide a comprehensive summary of the risk factors associated with the therapeutic application of metal NPs. Understanding and addressing these factors will be crucial for successful clinical translation and to mitigate any potential challenges or failures in the translation of metal NP-based therapies. By exploring the therapeutic potential of metal NPs and identifying the associated risk factors, this review aims to contribute to the advancement of cancer treatment strategies. The anticipated outcome of this review is to provide valuable insights and pave the way for the advancement of effective and targeted therapies utilizing metal NPs specifically for cancer patients.
Collapse
Affiliation(s)
- Ali Hheidari
- Department of Mechanical Engineering, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Javad Mohammadi
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Maryam Ghodousi
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, United States
| | - Mohammadreza Mahmoodi
- Bio-microfluidics Lab, Department of Electrical Engineering and Information Technology, Iranian Research Organization for Science and Technology, Tehran, Iran
| | - Sina Ebrahimi
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Esmail Pishbin
- Bio-microfluidics Lab, Department of Electrical Engineering and Information Technology, Iranian Research Organization for Science and Technology, Tehran, Iran
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol, Iran
| |
Collapse
|
2
|
Lee SK, Han MS, Tung CH. In vivo senescence imaging nanoprobe targets the associated reactive oxygen species. NANOSCALE 2024; 16:1371-1383. [PMID: 38131616 DOI: 10.1039/d3nr04083f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Cellular senescence, a cell-cycle arrest state upon stress or damage, can adversely impact aging and cancers. We have designed a novel near infrared fluorogenic nanoprobe, named D3, which can only be turned on by highly elevated levels of reactive oxygen species (ROS), critical players for the induction and maintenance of senescence, for real-time senescence sensing and imaging. In contrast to glowing senescent cells, non-senescent cells whose ROS levels are too low to activate the D3 signal remain optically silent. Upon systemic injection into senescent tumor-bearing mice, the D3 nanoprobe quickly accumulates in tumors, and its fluorescence signal is turned on specifically by senescence-associated ROS in the senescent tumors. The fluorescence signal at senescent tumors was 3-fold higher than that of non-senescent tumors. This groundbreaking design introduces a novel activation mechanism and a powerful imaging nanoprobe to identify and assess cellular senescence in living organisms.
Collapse
Affiliation(s)
- Seung Koo Lee
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, 413 East 69th Street, Box 290, New York, NY 10021, USA.
| | - Myung Shin Han
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, 413 East 69th Street, Box 290, New York, NY 10021, USA.
| | - Ching-Hsuan Tung
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, 413 East 69th Street, Box 290, New York, NY 10021, USA.
| |
Collapse
|
3
|
Vanharen M, Girard D. Impact of gold nanoparticles (AuNPs) on eosinophils isolated from male and female individuals. Immunobiology 2023; 228:152762. [PMID: 38006680 DOI: 10.1016/j.imbio.2023.152762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 11/06/2023] [Accepted: 11/19/2023] [Indexed: 11/27/2023]
Abstract
It is well established that some differences exist between the male and female immune systems. Despites this, a sex-based analysis is not frequently performed in most scientific published reports. Knowing that inflammation is a common undesired effect observed resulting from nanoparticle (NP) exposure, we investigate here how in vitro treatment of gold NPs with a primary size of 20 and 70 nm (AuNP20 and AuNP70, respectively) will alter the biology of human eosinophils isolated from men and women blood. We found that treatment of AuNP70, but not AuNP20, significantly delay apoptosis only in eosinophils isolated from women. AuNPs were found to decrease eosinophil phagocytosis, however, significance was only observed in AuNP20-induced eosinophils isolated from women. The production of IL-8 was significantly increased in response to both AuNPs but only in eosinophils isolated from men and the production of IL-1β was increased in AuNPs-induced eosinophils, although significance was observed only in AuNP70-induced eosinophils isolated from women. We conclude that future studies investigating the toxicity of AuNPs (or other NPs) should include a sex-based analysis, especially if the tested NPs have potential medical applications knowing the increased interest in the development of personalized precision medicine.
Collapse
Affiliation(s)
- Marion Vanharen
- Laboratoire de recherche en inflammation et physiologie des granulocytes, Université du Québec, INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec, Canada
| | - Denis Girard
- Laboratoire de recherche en inflammation et physiologie des granulocytes, Université du Québec, INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec, Canada.
| |
Collapse
|
4
|
Kim SJ, Ko WK, Han GH, Lee D, Cho MJ, Sheen SH, Sohn S. Axon guidance gene-targeted siRNA delivery system improves neural stem cell transplantation therapy after spinal cord injury. Biomater Res 2023; 27:101. [PMID: 37840145 PMCID: PMC10577901 DOI: 10.1186/s40824-023-00434-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND Neural stem cells (NSCs) derived from the embryonic spinal cord are excellent candidates for the cellular regeneration of lost neural cells after spinal cord injury (SCI). Semaphorin 3 A (Sema3A) is well known as being implicated in the major axon guidance of the growth cone as a repulsive function during the development of the central nervous system, yet its function in NSC transplantation therapy for SCI has not been investigated. Here, we report for the first time that embryonic spinal cord-derived NSCs significantly express Sema3A in the SCI environment, potentially facilitating inhibition of cell proliferation after transplantation. METHODS siRNA-Sema3A was conjugated with poly-l-lysin-coated gold nanoparticles (AuNPs) through a charge interaction process. NSCs were isolated from embryonic spinal cords of rats. Then, the cells were embedded into a dual-degradable hydrogel with the siRNA- Sema3A loaded-AuNPs and transplanted after complete SCI in rats. RESULTS The knockdown of Sema3A by delivering siRNA nanoparticles via dual-degradable hydrogels led to a significant increase in cell survival and neuronal differentiation of the transplanted NSCs after SCI. Of note, the knockdown of Sema3A increased the synaptic connectivity of transplanted NSC in the injured spinal cord. Moreover, extracellular matrix molecule and functional recovery were significantly improved in Sema3A-inhibited rats compared to those in rats with only NSCs transplanted. CONCLUSIONS These findings demonstrate the important role of Sema3A in NSC transplantation therapy, which may be considered as a future cell transplantation therapy for SCI cases.
Collapse
Affiliation(s)
- Seong Jun Kim
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, 59, Yatap-ro, Bundang- gu, Seongnam-si, 13496, Gyeonggi-do, Republic of Korea
- Department of Biomedical Science, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Wan-Kyu Ko
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, 59, Yatap-ro, Bundang- gu, Seongnam-si, 13496, Gyeonggi-do, Republic of Korea
- Department of Biomedical Science, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Gong Ho Han
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, 59, Yatap-ro, Bundang- gu, Seongnam-si, 13496, Gyeonggi-do, Republic of Korea
- Department of Biomedical Science, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Daye Lee
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, 59, Yatap-ro, Bundang- gu, Seongnam-si, 13496, Gyeonggi-do, Republic of Korea
- Department of Biomedical Science, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Min Jai Cho
- Department of Neurosurgery, Chungbuk National University, 776, 1Sunhawn-ro, Seowon-gu, Cheongju-si, 28644, Republic of Korea
| | - Seung Hun Sheen
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, 59, Yatap-ro, Bundang- gu, Seongnam-si, 13496, Gyeonggi-do, Republic of Korea
| | - Seil Sohn
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, 59, Yatap-ro, Bundang- gu, Seongnam-si, 13496, Gyeonggi-do, Republic of Korea.
- Department of Biomedical Science, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
5
|
Lee A, Gosnell N, Milinkovic D, Taladriz-Blanco P, Rothen-Rutishauser B, Petri-Fink A. Layer-by-Layer siRNA Particle Assemblies for Localized Delivery of siRNA to Epithelial Cells through Surface-Mediated Particle Uptake. ACS APPLIED BIO MATERIALS 2023; 6:83-92. [PMID: 36598879 PMCID: PMC9847476 DOI: 10.1021/acsabm.2c00668] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/08/2022] [Indexed: 01/05/2023]
Abstract
Localized delivery of small interfering RNA (siRNA) is a promising approach for spatial control of cell responses at biomaterial interfaces. Layer-by-layer (LbL) assembly of siRNA with cationic polyelectrolytes has been used in film and nanoparticle vectors for transfection. Herein, we combine the ability of particles to efficiently deliver siRNA with the ability of film polyelectrolyte multilayers to act locally. LbL particles were prepared with alternating layers of poly(l-arginine) and siRNA and capped with hyaluronic acid. Negatively charged LbL particles were subsequently assembled on the poly(l-lysine)-functionalized substrate to form a LbL particle-decorated surface. Cells grown in contact with the particle-decorated surface were able to survive, internalize particles, and undergo gene silencing. This work shows that particle-decorated surfaces can be engineered by using electrostatic interactions and used to deliver therapeutic payloads for cell-instructive biointerfaces.
Collapse
Affiliation(s)
- Aaron Lee
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Natalia Gosnell
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Daela Milinkovic
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Patricia Taladriz-Blanco
- International
Iberian Nanotechnology Laboratory, Water
Quality Group, Avenue
Mestre Jose Veiga s/n, 4715-330 Braga, Portugal
| | | | - Alke Petri-Fink
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
- Department
of Chemistry, University of Fribourg, Chemin du Musee 9, 1700 Fribourg, Switzerland
| |
Collapse
|
6
|
Kanu GA, Parambath JBM, Abu Odeh RO, Mohamed AA. Gold Nanoparticle-Mediated Gene Therapy. Cancers (Basel) 2022; 14:5366. [PMID: 36358785 PMCID: PMC9653658 DOI: 10.3390/cancers14215366] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 07/30/2023] Open
Abstract
Gold nanoparticles (AuNPs) have gained increasing attention as novel drug-delivery nanostructures for the treatment of cancers, infections, inflammations, and other diseases and disorders. They are versatile in design, synthesis, modification, and functionalization. This has many advantages in terms of gene editing and gene silencing, and their application in genetic illnesses. The development of several techniques such as CRISPR/Cas9, TALEN, and ZFNs has raised hopes for the treatment of genetic abnormalities, although more focused experimentation is still needed. AuNPs, however, have been much more effective in trending research on this subject. In this review, we highlight recently well-developed advancements that are relevant to cutting-edge gene therapies, namely gene editing and gene silencing in diseases caused by a single gene in humans by taking an edge of the unique properties of the AuNPs, which will be an important outlook for future research.
Collapse
Affiliation(s)
- Gayathri A. Kanu
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Javad B. M. Parambath
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Center for Advanced Materials Research, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Raed O. Abu Odeh
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Ahmed A. Mohamed
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Center for Advanced Materials Research, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
7
|
Claßen R, Pouokam E, Wickleder M, Diener M, Mattern A. Atropine-functionalized gold nanoparticles binding to muscarinic receptors after passage across the intestinal epithelium. ROYAL SOCIETY OPEN SCIENCE 2022; 9:220244. [PMID: 36249335 PMCID: PMC9533000 DOI: 10.1098/rsos.220244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 09/09/2022] [Indexed: 06/16/2023]
Abstract
Gold nanoparticles have a high potential to be a treatment of diseases by their specific drug delivery properties and multivalent receptor stimulation. For the present project, spherical gold nanoparticles were synthesized and functionalized with the muscarinic receptor antagonist atropine (Au-MUDA-AT NPs). The diameter of the gold core could precisely be controlled by using different synthetic methods and reducing agents resulting in functionalized gold nanoparticles with diameters ranging from 8 to 16 nm. The ability to interact with intestinal muscarinic receptors is size-dependent. When using intestinal chloride secretion induced by the stable acetylcholine derivative, carbachol, as read-out, the strongest inhibition, i.e. the most efficient blockade of muscarinic receptors, was observed with 13 nm sized Au-MUDA-AT NPs. Functional experiments indicate that Au-MUDA-AT NPs with a diameter of 14 nm are able to pass the intestinal mucosa in a time-dependent manner after administration to the intestinal lumen. For example, luminally administered Au-MUDA-AT NPs inhibited contractions of the small intestinal longitudinal muscle layer induced by electrical stimulation of myenteric neurons. A similar inhibition of basolateral epithelial receptors was observed after luminal administration of Au-MUDA-AT NPs when using carbachol-induced chloride secretion across the intestinal epithelium as a test system. Thus, Au-MUDA-AT NPs might be a therapeutic tool for the modulation of intestinal secretion and motility after oral application in the future.
Collapse
Affiliation(s)
- Rebecca Claßen
- Institute for Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Frankfurter Strasse 100, 35392 Giessen, Germany
| | - Ervice Pouokam
- Institute for Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Frankfurter Strasse 100, 35392 Giessen, Germany
| | - Matthias Wickleder
- Institute of Inorganic Chemistry, University of Cologne, Greinstrasse 6, 50939 Cologne, Germany
| | - Martin Diener
- Institute for Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Frankfurter Strasse 100, 35392 Giessen, Germany
| | - Annabelle Mattern
- Institute of Inorganic Chemistry, University of Cologne, Greinstrasse 6, 50939 Cologne, Germany
| |
Collapse
|
8
|
Nanoparticle-based delivery strategies of multifaceted immunomodulatory RNA for cancer immunotherapy. J Control Release 2022; 343:564-583. [DOI: 10.1016/j.jconrel.2022.01.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/25/2022] [Accepted: 01/29/2022] [Indexed: 12/18/2022]
|
9
|
Yan Y, Liu XY, Lu A, Wang XY, Jiang LX, Wang JC. Non-viral vectors for RNA delivery. J Control Release 2022; 342:241-279. [PMID: 35016918 PMCID: PMC8743282 DOI: 10.1016/j.jconrel.2022.01.008] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/13/2022]
Abstract
RNA-based therapy is a promising and potential strategy for disease treatment by introducing exogenous nucleic acids such as messenger RNA (mRNA), small interfering RNA (siRNA), microRNA (miRNA) or antisense oligonucleotides (ASO) to modulate gene expression in specific cells. It is exciting that mRNA encoding the spike protein of COVID-19 (coronavirus disease 2019) delivered by lipid nanoparticles (LNPs) exhibits the efficient protection of lungs infection against the virus. In this review, we introduce the biological barriers to RNA delivery in vivo and discuss recent advances in non-viral delivery systems, such as lipid-based nanoparticles, polymeric nanoparticles, N-acetylgalactosamine (GalNAc)-siRNA conjugate, and biomimetic nanovectors, which can protect RNAs against degradation by ribonucleases, accumulate in specific tissue, facilitate cell internalization, and allow for the controlled release of the encapsulated therapeutics.
Collapse
Affiliation(s)
- Yi Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiao-Yu Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - An Lu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiang-Yu Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Lin-Xia Jiang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jian-Cheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China..
| |
Collapse
|
10
|
Mahalanobish S, Kundu M, Ghosh S, Das J, Sil PC. Fabrication of phenyl boronic acid modified pH-responsive zinc oxide nanoparticles as targeted delivery of chrysin on human A549 cells. Toxicol Rep 2022; 9:961-969. [PMID: 35875254 PMCID: PMC9301599 DOI: 10.1016/j.toxrep.2022.04.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/08/2022] [Accepted: 04/17/2022] [Indexed: 01/22/2023] Open
Abstract
Recently, different natural bioactive compounds have been used as anticancer agents for their various therapeutic benefits and non-toxic nature to other organs. However, they have various restrictions in preclinical and clinical studies due to their non-targeting nature and insufficient bioavailability. As a result, a zinc oxide nanoparticle (ZnO) based drug delivery medium was constructed which has good bio-compatibility and bio-degradability. It also displays cancer cell-specific drug delivery in a targeted and controlled way. In the present study, phenylboronic acid (PBA) tagged ZnO nanoparticles (ZnO-PBA) was fabricated and in the next step, chrysin (a natural bio-active molecule) was loaded to it to form the nanoconjugate (ZnO-PBA-Chry). Different characterization techniques were used to confirm the successful fabrication of ZnO-PBA-Chry. PBA-tagging to the nanoparticle helps in targeted delivery of chrysin in lung cancer cells (A549) as PBA binds with sialic acid receptors which are over-expressed on the surface of A549 cells. As ZnO dissociates in acidic pH, it shows stimuli-responsive release of chrysin in tumor microenvironment. Application of ZnO-PBA-Chry nanohybrid in lung cancer cell line A549 caused oxidative stress mediated intrinsic cell death and cell cycle arrest. ZnO-PBA-Chry downregulated MMP-2 and VE-Cadherin, thereby inhibiting metastasis and the invasive property of A549 cells. pH-responsive PBA functionalized ZnO nanoparticle was fabricated. Chrysin was loaded as a bioactive anticancer agent into ZnO nanoparticle. ZnO-PBA-Chry induced intrinsic cell death and cell cycle arrest in A549 cells. It inhibited metastasis and invasive properties of A549 cells.
Collapse
|
11
|
Ozcicek I, Aysit N, Cakici C, Aydeger A. The effects of surface functionality and size of gold nanoparticles on neuronal toxicity, apoptosis, ROS production and cellular/suborgan biodistribution. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112308. [PMID: 34474859 DOI: 10.1016/j.msec.2021.112308] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/15/2021] [Accepted: 07/07/2021] [Indexed: 12/19/2022]
Abstract
Gold nanoparticles are emerging as promising nanomaterials to create nanoscale therapeutic delivery systems. The aim of the study was to synthesis of highly monodisperse and stable gold nanoparticles functionalized with polyethyleneimine (PEI) and polyethylene glycol (PEG), multiparametric investigation of their neuronal toxicological effects and evaluation of the cellular/suborgan biodistribution. Gold nanoparticles (AuNP20 and AuNP50) were synthesized and their surfaces were electrostatically modified by PEI and PEG. Dorsal root ganglion (DRG) sensory neurones were isolated from BALB/c mice. Cell viability, apoptosis and ROS production were evaluated in vitro. Cellular and suborgan biodisribution of the AuNPs were investigated using inductively coupled plasma mass spectrometry (ICP-MS) technique. PEI and PEG surface coating increased both biocompatibility and biodistribution of the AuNPs. ICP-MS measurements showed the presence of gold in liver, spleen, kidney, heart, blood and brain within a 30 days period. The size and surface chemistry of the AuNPs are important parameters for potential nanoteranostic applications in the future studies.
Collapse
Affiliation(s)
- Ilyas Ozcicek
- Department of Medical Biology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey; Health Science and Technologies Research Institute (SABITA), Istanbul Medipol University, Istanbul, Turkey.
| | - Nese Aysit
- Department of Medical Biology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey; Health Science and Technologies Research Institute (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Cagri Cakici
- Department of Medical Biochemistry, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Asel Aydeger
- Graduate School of Health Sciences, Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
12
|
The Nanosystems Involved in Treating Lung Cancer. Life (Basel) 2021; 11:life11070682. [PMID: 34357054 PMCID: PMC8307574 DOI: 10.3390/life11070682] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 12/12/2022] Open
Abstract
Even though there are various types of cancer, this pathology as a whole is considered the principal cause of death worldwide. Lung cancer is known as a heterogeneous condition, and it is apparent that genome modification presents a significant role in the occurrence of this disorder. There are conventional procedures that can be utilized against diverse cancer types, such as chemotherapy or radiotherapy, but they are hampered by the numerous side effects. Owing to the many adverse events observed in these therapies, it is imperative to continuously develop new and improved strategies for managing individuals with cancer. Nanomedicine plays an important role in establishing new methods for detecting chromosomal rearrangements and mutations for targeted chemotherapeutics or the local delivery of drugs via different types of nano-particle carriers to the lungs or other organs or areas of interest. Because of the complex signaling pathways involved in developing different types of cancer, the need to discover new methods for prevention and detection is crucial in producing gene delivery materials that exhibit the desired roles. Scientists have confirmed that nanotechnology-based procedures are more effective than conventional chemotherapy or radiotherapy, with minor side effects. Several nanoparticles, nanomaterials, and nanosystems have been studied, including liposomes, dendrimers, polymers, micelles, inorganic nanoparticles, such as gold nanoparticles or carbon nanotubes, and even siRNA delivery systems. The cytotoxicity of such nanosystems is a debatable concern, and nanotechnology-based delivery systems must be improved to increase the bioavailability, biocompatibility, and safety profiles, since these nanosystems boast a remarkable potential in many biomedical applications, including anti-tumor activity or gene therapy. In this review, the nanosystems involved in treating lung cancer and its associated challenges are discussed.
Collapse
|
13
|
Wang L, Yan Y. A Review of pH-Responsive Organic-Inorganic Hybrid Nanoparticles for RNAi-Based Therapeutics. Macromol Biosci 2021; 21:e2100183. [PMID: 34160896 DOI: 10.1002/mabi.202100183] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/04/2021] [Indexed: 12/13/2022]
Abstract
RNA interference (RNAi) shows great potential in the treatment of varying cancer and genetic disorders. The lack of safe and effective delivery methods is an ongoing challenge to realize the full potential of RNAi-based therapeutics. pH-responsive hybrid nanoparticle is a promising non-virus platform for small interfering RNA (siRNA) delivery with unique properties including the robust response to the acidic microenvironment and the capability of theranostic and combined therapeutics. The mechanism of RNAi and the delivery barriers for RNAi-based therapeutics are first discussed. Then, the general patterns of pH-response and the typical construction of hybrid nanoparticles are demonstrated. The recent advances in pH-responsive organic-inorganic hybrid nanoparticles for siRNA delivery are highlighted, in particular, how pH-response of ionizable groups, acid-labile bonds, and decomposition of inorganic components affect the physicochemical properties of hybrid nanoparticles and benefit the cellular uptake and intracellular trafficking of siRNA payloads are discussed. At last, the remaining problems and the prospects for pH-responsive hybrid nanoparticles for siRNA delivery are analyzed.
Collapse
Affiliation(s)
- Lu Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Yunfeng Yan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
14
|
Liu Y, Yin L. α-Amino acid N-carboxyanhydride (NCA)-derived synthetic polypeptides for nucleic acids delivery. Adv Drug Deliv Rev 2021; 171:139-163. [PMID: 33333206 DOI: 10.1016/j.addr.2020.12.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/06/2020] [Accepted: 12/10/2020] [Indexed: 12/17/2022]
Abstract
In recent years, gene therapy has come into the spotlight for the prevention and treatment of a wide range of diseases. Polypeptides have been widely used in mediating nucleic acid delivery, due to their versatilities in chemical structures, desired biodegradability, and low cytotoxicity. Chemistry plays an essential role in the development of innovative polypeptides to address the challenges of producing efficient and safe gene vectors. In this Review, we mainly focused on the latest chemical advances in the design and preparation of polypeptide-based nucleic acid delivery vehicles. We first discussed the synthetic approach of polypeptides via ring-opening polymerization (ROP) of N-carboxyanhydrides (NCAs), and introduced the various types of polypeptide-based gene delivery systems. The extracellular and intracellular barriers against nucleic acid delivery were then outlined, followed by detailed review on the recent advances in polypeptide-based delivery systems that can overcome these barriers to enable in vitro and in vivo gene transfection. Finally, we concluded this review with perspectives in this field.
Collapse
Affiliation(s)
- Yong Liu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
| | - Lichen Yin
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China.
| |
Collapse
|
15
|
Muhammad K, Zhao J, Gao B, Feng Y. Polymeric nano-carriers for on-demand delivery of genes via specific responses to stimuli. J Mater Chem B 2021; 8:9621-9641. [PMID: 32955058 DOI: 10.1039/d0tb01675f] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Polymeric nano-carriers have been developed as a most capable and feasible technology platform for gene therapy. As vehicles, polymeric nano-carriers are obliged to possess high gene loading capability, low immunogenicity, safety, and the ability to transfer various genetic materials into specific sites of target cells to express therapeutic proteins or block a process of gene expression. To this end, various types of polymeric nano-carriers have been prepared to release genes in response to stimuli such as pH, redox, enzymes, light and temperature. These stimulus-responsive nano-carriers exhibit high gene transfection efficiency and low cytotoxicity. In particular, dual- and multi-stimulus-responsive polymeric nano-carriers can respond to a combination of signals. Markedly, these combined responses take place either simultaneously or in a sequential manner. These dual-stimulus-responsive polymeric nano-carriers can control gene delivery with high gene transfection both in vitro and in vivo. In this review paper, we highlight the recent exciting developments in stimulus-responsive polymeric nano-carriers for gene delivery applications.
Collapse
Affiliation(s)
- Khan Muhammad
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, P. R. China.
| | - Jing Zhao
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, P. R. China.
| | - Bin Gao
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, P. R. China.
| | - Yakai Feng
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, P. R. China. and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, P. R. China and Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Tianjin 300350, P. R. China
| |
Collapse
|
16
|
Zhou Y, Zhang Y, Lin W, Kesseli SJ, Huang L, Zhao Y, Yao X, Huang K, Han G. Wavelength-Selective Light-Controlled Stepwise Photolysis from Single Gold Nanoparticles. Adv Healthc Mater 2021; 10:e2000321. [PMID: 33034955 DOI: 10.1002/adhm.202000321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/02/2020] [Indexed: 11/09/2022]
Abstract
Light-controlled sequential photolysis from a single nanoparticle is a challenge for controlled release. A wavelength-selective sequential photolysis from single gold nanoparticles is reported for the first time. In particular, it is also demonstrated that such nanoparticle can be used to sequentially release two payloads in living cells. In principle, this system can be extended to sequential release of multiple different types of payloads by rational design of diverse photocleavable linkers. It is expected that this work can provide a new tool for better orderly controlling cellular events that request high spatiotemporal manners.
Collapse
Affiliation(s)
- Yekui Zhou
- Department of Biochemistry and Molecular Pharmacology University of Massachusetts Medical School Worcester MA 01605 USA
| | - Yuanwei Zhang
- Department of Biochemistry and Molecular Pharmacology University of Massachusetts Medical School Worcester MA 01605 USA
| | - Wenhai Lin
- Department of Biochemistry and Molecular Pharmacology University of Massachusetts Medical School Worcester MA 01605 USA
| | - Samuel J. Kesseli
- Department of Biochemistry and Molecular Pharmacology University of Massachusetts Medical School Worcester MA 01605 USA
| | - Ling Huang
- Department of Biochemistry and Molecular Pharmacology University of Massachusetts Medical School Worcester MA 01605 USA
| | - Yang Zhao
- Department of Biochemistry and Molecular Pharmacology University of Massachusetts Medical School Worcester MA 01605 USA
| | - Xikuang Yao
- Department of Biochemistry and Molecular Pharmacology University of Massachusetts Medical School Worcester MA 01605 USA
| | - Kai Huang
- Department of Biochemistry and Molecular Pharmacology University of Massachusetts Medical School Worcester MA 01605 USA
| | - Gang Han
- Department of Biochemistry and Molecular Pharmacology University of Massachusetts Medical School Worcester MA 01605 USA
| |
Collapse
|
17
|
Charbe NB, Amnerkar ND, Ramesh B, Tambuwala MM, Bakshi HA, Aljabali AA, Khadse SC, Satheeshkumar R, Satija S, Metha M, Chellappan DK, Shrivastava G, Gupta G, Negi P, Dua K, Zacconi FC. Small interfering RNA for cancer treatment: overcoming hurdles in delivery. Acta Pharm Sin B 2020; 10:2075-2109. [PMID: 33304780 PMCID: PMC7714980 DOI: 10.1016/j.apsb.2020.10.005] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/24/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
In many ways, cancer cells are different from healthy cells. A lot of tactical nano-based drug delivery systems are based on the difference between cancer and healthy cells. Currently, nanotechnology-based delivery systems are the most promising tool to deliver DNA-based products to cancer cells. This review aims to highlight the latest development in the lipids and polymeric nanocarrier for siRNA delivery to the cancer cells. It also provides the necessary information about siRNA development and its mechanism of action. Overall, this review gives us a clear picture of lipid and polymer-based drug delivery systems, which in the future could form the base to translate the basic siRNA biology into siRNA-based cancer therapies.
Collapse
Key Words
- 1,3-propanediol, PEG-b-PDMAEMA-b-Ppy
- 2-propylacrylicacid, PAH-b-PDMAPMA-b-PAH
- APOB, apolipoprotein B
- AQP-5, aquaporin-5
- AZEMA, azidoethyl methacrylate
- Atufect01, β-l-arginyl-2,3-l-diaminopropionicacid-N-palmityl-N-oleyl-amide trihydrochloride
- AuNPs, gold nanoparticles
- B-PEI, branched polyethlenimine
- BMA, butyl methacrylate
- CFTR, cystic fibrosis transmembrane conductance regulator gene
- CHEMS, cholesteryl hemisuccinate
- CHOL, cholesterol
- CMC, critical micelles concentration
- Cancer
- DC-Chol, 3β-[N-(N′,N′-dimethylaminoethane)carbamoyl]cholesterol
- DMAEMA, 2-dimethylaminoethyl methacrylate
- DNA, deoxyribonucleic acid
- DOPC, dioleylphosphatidyl choline
- DOPE, dioleylphosphatidyl ethanolamine
- DOTAP, N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium methyl-sulfate
- DOTMA, N-[1-(2,3-dioleyloxy)propy]-N,N,N-trimethylammoniumchloride
- DOX, doxorubicin
- DSGLA, N,N-dis-tearyl-N-methyl-N-2[N′-(N2-guanidino-l-lysinyl)] aminoethylammonium chloride
- DSPC, 1,2-distearoyl-sn-glycero-3-phosphocholine
- DSPE, 1,2-distearoyl-sn-glycero-3-phosphorylethanolamine
- DSPE-MPEG, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (ammonium salt)
- DSPE-PEG-Mal: 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[maleimide(polyethylene glycol)-2000] (mmmonium salt), EPR
- Liposomes
- Micelles
- N-acetylgalactosamine, HIF-1α
- Nanomedicine
- PE-PCL-b-PNVCL, pentaerythritol polycaprolactone-block-poly(N-vinylcaprolactam)
- PLA, poly-l-arginine
- PLGA, poly lactic-co-glycolic acid
- PLK-1, polo-like kinase 1
- PLL, poly-l-lysine
- PPES-b-PEO-b-PPES, poly(4-(phenylethynyl)styrene)-block-PEO-block-poly(4-(phenylethynyl)styrene)
- PTX, paclitaxel
- PiRNA, piwi-interacting RNA
- Polymer
- RES, reticuloendothelial system
- RGD, Arg-Gly-Asp peptide
- RISC, RNA-induced silencing complex
- RNA, ribonucleic acid
- RNAi, RNA interference
- RNAse III, ribonuclease III enzyme
- SEM, scanning electron microscope
- SNALP, stable nucleic acid-lipid particles
- SiRNA, short interfering rNA
- Small interfering RNA (siRNA)
- S–Au, thio‒gold
- TCC, transitional cell carcinoma
- TEM, transmission electron microscopy
- Tf, transferrin
- Trka, tropomyosin receptor kinase A
- USPIO, ultra-small superparamagnetic iron oxide nanoparticles
- UV, ultraviolet
- VEGF, vascular endothelial growth factor
- ZEBOV, Zaire ebola virus
- enhanced permeability and retention, Galnac
- hypoxia-inducible factor-1α, KSP
- kinesin spindle protein, LDI
- lipid-protamine-DNA/hyaluronic acid, MDR
- lysine ethyl ester diisocyanate, LPD/LPH
- messenger RNA, MTX
- methotrexate, NIR
- methoxy polyethylene glycol-polycaprolactone, mRNA
- methoxypoly(ethylene glycol), MPEG-PCL
- micro RNA, MPEG
- multiple drug resistance, MiRNA
- nanoparticle, NRP-1
- near-infrared, NP
- neuropilin-1, PAA
- poly(N,N-dimethylacrylamide), PDO
- poly(N-isopropyl acrylamide), pentaerythritol polycaprolactone-block-poly(N-isopropylacrylamide)
- poly(acrylhydrazine)-block-poly(3-dimethylaminopropyl methacrylamide)-block-poly(acrylhydrazine), PCL
- poly(ethylene glycol)-block-poly(2-dimethylaminoethyl methacrylate)-block poly(pyrenylmethyl methacrylate), PEG-b-PLL
- poly(ethylene glycol)-block-poly(l-lysine), PEI
- poly(ethylene oxide)-block-poly(2-(diethylamino)ethyl methacrylate)-stat-poly(methoxyethyl methacrylate), PEO-b-PCL
- poly(ethylene oxide)-block-poly(Ε-caprolactone), PE-PCL-b-PNIPAM
- poly(Ε-caprolactone), PCL-PEG
- poly(Ε-caprolactone)-polyethyleneglycol-poly(l-histidine), PCL-PEI
- polycaprolactone-polyethyleneglycol, PCL-PEG-PHIS
- polycaprolactone-polyethylenimine, PDMA
- polyethylenimine, PEO-b-P(DEA-Stat-MEMA
Collapse
Affiliation(s)
- Nitin Bharat Charbe
- Departamento de Quimica Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Sri Adichunchunagiri College of Pharmacy, Sri Adichunchunagiri University, BG Nagar, Karnataka 571418, India
| | - Nikhil D. Amnerkar
- Adv V. R. Manohar Institute of Diploma in Pharmacy, Nagpur, Maharashtra 441110, India
| | - B. Ramesh
- Sri Adichunchunagiri College of Pharmacy, Sri Adichunchunagiri University, BG Nagar, Karnataka 571418, India
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| | - Hamid A. Bakshi
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| | - Alaa A.A. Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid 21163, Jordan
| | - Saurabh C. Khadse
- Department of Pharmaceutical Chemistry, R.C. Patel Institute of Pharmaceutical Education and Research, Dist. Dhule, Maharashtra 425 405, India
| | - Rajendran Satheeshkumar
- Departamento de Quimica Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Saurabh Satija
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411 Punjab, India
| | - Meenu Metha
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411 Punjab, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Garima Shrivastava
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi, New Delhi 110016, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Jaipur 302017, India
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, India
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) and School of Biomedical Sciences and Pharmacy, University of Newcastle, NSW 2308, Australia
| | - Flavia C. Zacconi
- Departamento de Quimica Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 4860, Chile
| |
Collapse
|
18
|
Pei Gong, Wu M, Zhang J, Li X, Liu J, Wan F. Comprehensive Understanding of Gold Nanoparticles Enhancing Catalytic Efficiency. COLLOID JOURNAL 2020. [DOI: 10.1134/s1061933x20050087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Luther DC, Huang R, Jeon T, Zhang X, Lee YW, Nagaraj H, Rotello VM. Delivery of drugs, proteins, and nucleic acids using inorganic nanoparticles. Adv Drug Deliv Rev 2020; 156:188-213. [PMID: 32610061 PMCID: PMC8559718 DOI: 10.1016/j.addr.2020.06.020] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 01/03/2023]
Abstract
Inorganic nanoparticles provide multipurpose platforms for a broad range of delivery applications. Intrinsic nanoscopic properties provide access to unique magnetic and optical properties. Equally importantly, the structural and functional diversity of gold, silica, iron oxide, and lanthanide-based nanocarriers provide unrivalled control of nanostructural properties for effective transport of therapeutic cargos, overcoming biobarriers on the cellular and organismal level. Taken together, inorganic nanoparticles provide a key addition to the arsenal of delivery vectors for fighting disease and improving human health.
Collapse
Affiliation(s)
- David C Luther
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Rui Huang
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Taewon Jeon
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA; Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Xianzhi Zhang
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Yi-Wei Lee
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA.
| |
Collapse
|
20
|
Lei WX, An ZS, Zhang BH, Wu Q, Gong WJ, Li JM, Chen WL. Construction of gold-siRNA NPR1 nanoparticles for effective and quick silencing of NPR1 in Arabidopsis thaliana. RSC Adv 2020; 10:19300-19308. [PMID: 35515443 PMCID: PMC9054099 DOI: 10.1039/d0ra02156c] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 05/04/2020] [Indexed: 12/20/2022] Open
Abstract
In recent years, gold nanoparticles (AuNPs) have been widely used as gene silencing agents and therapeutics for treatment of cancers due to their high transfection efficiency and lack of cytotoxicity, but their roles in gene silencing in plants have not yet been reported. Here, we report synthesis of AuNPs-branched polyethylenimine and its integration with the small interfering RNAs (siRNA) of NPR1 to form a AuNPs-siRNANPR1 compound. Our results showed that AuNPs-siRNANPR1 was capable of infiltrating into Arabidopsis cells. AuNPs-siRNANPR1 silenced 80% of the NPR1 gene in Arabidopsis. Bacteriostatic and ion leakage experiments suggest that the NPR1 gene in Arabidopsis leaves was silenced by AuNPs-siRNANPR1. In Columbia-0 plants, compared with the control group treated with buffer solution, the AuNPs-siRNANPR1 treatment significantly increased the number of colonies and cell death, and the leaves turned yellow, similar to the phenotype of the npr1 leaves. These results indicated this AuNPs-siRNANPR1 silencing the NPR1 gene method is simple, effective and quick (3 days), and a powerful tool to study gene functions in plants. Gold nanoparticles (AuNPs) have been widely used as gene silencing agents and therapeutics for treatment due to their high transfection efficiency and lack of cytotoxicity, but their roles in gene silencing in plants have not yet been reported.![]()
Collapse
Affiliation(s)
- Wen-Xue Lei
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University Guangzhou 510631 China +86-20-85216052 +86-20-85211436-8611.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University Guangzhou 510631 China
| | - Zi-Shuai An
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University Guangzhou 510631 China +86-20-85216052 +86-20-85211436-8611.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University Guangzhou 510631 China
| | - Bai-Hong Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University Guangzhou 510631 China +86-20-85216052 +86-20-85211436-8611.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University Guangzhou 510631 China
| | - Qian Wu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University Guangzhou 510631 China +86-20-85216052 +86-20-85211436-8611.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University Guangzhou 510631 China
| | - Wen-Jun Gong
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University Guangzhou 510631 China +86-20-85216052 +86-20-85211436-8611.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University Guangzhou 510631 China
| | - Jin-Ming Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University Guangzhou 510631 China +86-20-85216052 +86-20-85211436-8611.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University Guangzhou 510631 China
| | - Wen-Li Chen
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University Guangzhou 510631 China +86-20-85216052 +86-20-85211436-8611.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University Guangzhou 510631 China
| |
Collapse
|
21
|
Lee SK, Law B, Tung CH. Multifunctional Nanodelivery Platform for Maximizing Nucleic Acids Combination Therapy. Methods Mol Biol 2020; 2115:79-90. [PMID: 32006395 DOI: 10.1007/978-1-0716-0290-4_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The silencing of an oncogene with a small interfering RNA (siRNA) is a promising way for cancer therapy. Its efficacy can be further enhanced by integrating with other therapeutics; however, transporting siRNA and other active ingredients to the same location at the same time is challenging. Here, we report a novel multifunctional nanodelivery platform by sequentially layering several functional ingredients, such as siRNAs, microRNAs, peptides, and targeting ligands, onto a core through charge-charge interaction. The prepared nanovectors effectively and programmably delivered multiple active components to maximize therapeutic combination with minimal off-targeting effects.
Collapse
Affiliation(s)
- Seung Koo Lee
- Department of Radiology, Weill Cornell Medicine, Molecular Imaging Innovations Institute, New York, NY, USA
| | - Benedict Law
- Department of Radiology, Weill Cornell Medicine, Molecular Imaging Innovations Institute, New York, NY, USA
| | - Ching-Hsuan Tung
- Department of Radiology, Weill Cornell Medicine, Molecular Imaging Innovations Institute, New York, NY, USA.
| |
Collapse
|
22
|
Lee SK, Han MS, Zhang W, Tung CH. Multilayered Activatable Nanoprobe for Ultra-Bright Tumor Imaging. Macromol Biosci 2019; 19:e1900260. [PMID: 31743618 DOI: 10.1002/mabi.201900260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/02/2019] [Indexed: 11/06/2022]
Abstract
The development of tumor targeted probes with strong signal and high contrast is always challenging in cancer imaging. Here, a unique multilayered activatable nanoprobe (MAN) is prepared to fulfill this long-standing goal. MAN adopts a versatile layer-by-layer fabrication technique that sequentially assembles multifunctional polyelectrolytes onto nanoparticles via charge-charge interaction. Unlike the common one-probe-one-fluorochrome construct, MAN offers a dramatic fluorescence enhancement by transporting a large quantity of quenched fluorochromes for maximal signal and contrast. Excellent signal amplification and retention with negligible cytotoxicity is observed in cell study. Upon systemic injection into mice, MAN quickly accumulates in tumor and its fluorescent signal is turned on by proteases overexpressed in tumors, resulting in >700% tumor-to-normal-tissue contrast. This multilayered fabrication provides a simple and powerful universal platform to design sensitive tumor imaging probes.
Collapse
Affiliation(s)
- Seung Koo Lee
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, 413 East 69th Street, Box 290, New York, NY, 10021, USA
| | - Myung Shin Han
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, 413 East 69th Street, Box 290, New York, NY, 10021, USA
| | - Weiqi Zhang
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, 413 East 69th Street, Box 290, New York, NY, 10021, USA
| | - Ching-Hsuan Tung
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, 413 East 69th Street, Box 290, New York, NY, 10021, USA
| |
Collapse
|
23
|
Villar-Alvarez E, Leal BH, Cambón A, Pardo A, Martínez-Gonzalez R, Fernández-Vega J, Al-Qadi S, Mosquera VX, Bouzas A, Barbosa S, Taboada P. Triggered RNAi Therapy Using Metal Inorganic Nanovectors. Mol Pharm 2019; 16:3374-3385. [PMID: 31188622 DOI: 10.1021/acs.molpharmaceut.9b00021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The administration of small interfering RNA (siRNA) is a very interesting therapeutic option to treat genetic diseases such as Alzheimer's or some types of cancer, but its effective delivery still remains a challenge. Herein, Au nanorod (GNR)-based platforms functionalized with polyelectrolyte layers were developed and analyzed as potential siRNA nanocarriers. The polymeric layers were successfully assembled on the particle surfaces by means of the layer-by-layer assembly technique through the alternating deposition of oppositely charged poly(styrene)sulfonate, PSS, poly(lysine), PLL, and siRNA biopolymers, with a final hyaluronic acid layer in order to provide the nanoconstructs with a potential targeting ability as well as colloidal stability in physiological medium. Once the hybrid nanocarriers were obtained, the cargo release, their colloidal stability in physiological-relevant media, cytotoxicity, cellular internalization and uptake, and knockdown activity were studied. The present hybrid particles release the genetic material inside cells by means of a protease-assisted and/or a light-triggered release mechanism in order to control the delivery of the oligonucleotides on demand. In addition, the hybrid nanovectors were observed to be nontoxic to cells and could efficiently deliver the genetic material in the cell cytoplasms. The GNR-based nanocarriers proposed here can provide a suitable environment to load and protect a sufficient amount of the genetic material to allow an efficient and sustained knockdown gene expression for long (up to 93% for 72 h), thanks to the slow degradation of PLL, without the observation of adverse side toxic effects. It was also found that the silencing activity was enhanced with the number of siRNA layers assembled in the nanoplatforms.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Víctor X Mosquera
- Departamento de Cirugía Cardíaca, Complexo Hospitalario Universitario A Coruña , Instituto de Investigación Biomédica de A Coruña (INIBIC) , 15006 A Coruña , Spain
| | - Alberto Bouzas
- Departamento de Cirugía Cardíaca, Complexo Hospitalario Universitario A Coruña , Instituto de Investigación Biomédica de A Coruña (INIBIC) , 15006 A Coruña , Spain
| | | | | |
Collapse
|
24
|
Sadhukhan P, Kundu M, Chatterjee S, Ghosh N, Manna P, Das J, Sil PC. Targeted delivery of quercetin via pH-responsive zinc oxide nanoparticles for breast cancer therapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 100:129-140. [PMID: 30948047 DOI: 10.1016/j.msec.2019.02.096] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 12/12/2022]
Abstract
Naturally occurring bioactive compounds are gaining much importance as anti-tumor agents in recent times due to their high therapeutic potential and less systemic toxicity. However, different preclinical and clinical studies have noted significant shortcomings, such as nonspecific tumor targeting and low bioavailability which limit their usage in therapeutics. Therefore, a safe and compatible nanoparticle mediated controlled drug delivery system is in high demand to enable effective transport of the drug candidates in the tumor tissue. Herein, we have synthesized phenylboronic acid (PBA) conjugated Zinc oxide nanoparticles (PBA-ZnO), loaded with quercetin (a bioflavonoid widely found in plants), with zeta potential around -10.2 mV and diameter below 40 nm. Presence of PBA moieties over the nanoparticle surface facilitates targeted delivery of quercetin to the sialic acid over-expressed cancer cells. Moreover, Quercetin loaded PBA-ZnO nanoparticles (denoted as PBA-ZnO-Q) showed pH responsive drug release behavior. Results suggested that PBA-ZnO-Q induced apoptotic cell death in human breast cancer cells (MCF-7) via enhanced oxidative stress and mitochondrial damage. In line with the in vitro results, PBA-ZnO-Q was found to be effective in reducing tumor growth in EAC tumor bearing mice. Most interestingly, PBA-ZnO-Q is found to reduce tumor associated toxicity in liver, kidney and spleen. The cytotoxic potential of the nanohybrid is attributed to the combinatorial cytotoxic effects of quercetin and ZnO in the cancer cells. Overall, the presented data highlighted the chemotherapeutic potential of the novel nanohybrid, PBA-ZnO-Q which can be considered for clinical cancer treatment.
Collapse
Affiliation(s)
- Pritam Sadhukhan
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Mousumi Kundu
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Sharmistha Chatterjee
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Noyel Ghosh
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Prasenjit Manna
- Biological Science and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat, Assam 785006, India
| | - Joydeep Das
- School of Chemistry, Shoolini University of Biotechnology and Management Sciences, Bajhol, PO Sultanpur, Distt., Solan 173229, HP, India.
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India.
| |
Collapse
|
25
|
Ramchandani D, Lee SK, Yomtoubian S, Han MS, Tung CH, Mittal V. Nanoparticle Delivery of miR-708 Mimetic Impairs Breast Cancer Metastasis. Mol Cancer Ther 2019; 18:579-591. [PMID: 30679387 DOI: 10.1158/1535-7163.mct-18-0702] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 11/06/2018] [Accepted: 01/14/2019] [Indexed: 12/19/2022]
Abstract
Triple-negative breast cancer (TNBC) patients exhibit the worst clinical outcome due to its aggressive clinical course, higher rate of recurrence, and a conspicuous lack of FDA-approved targeted therapies. Here, we show that multilayered nanoparticles (NPs) carrying the metastasis suppressor microRNA miR-708 (miR708-NP) localize to orthotopic primary TNBC, and efficiently deliver the miR-708 cargo to reduce lung metastasis. Using a SOX2/OCT4 promoter reporter, we identified a population of miR-708low cancer cells with tumor-initiating properties, enhanced metastatic potential, and marked sensitivity to miR-708 treatment. In vivo, miR708-NP directly targeted the SOX2/OCT4-mCherry+ miR-708low tumor cells to impair metastasis. Together, our preclinical findings provide a mechanism-based antimetastatic therapeutic approach for TNBC, with a marked potential to generate miR-708 replacement therapy for high-risk TNBC patients in the clinic. To our knowledge, this gold nanoparticle-based delivery of microRNA mimetic is the first oligonucleotide-based targeted therapy for TNBC.
Collapse
Affiliation(s)
- Divya Ramchandani
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, New York
| | - Seung Koo Lee
- Department of Radiology, Molecular Imaging Innovations Institute, Weill Cornell Medicine, New York, New York
| | - Shira Yomtoubian
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, New York
| | - Myung Shin Han
- Department of Radiology, Molecular Imaging Innovations Institute, Weill Cornell Medicine, New York, New York
| | - Ching-Hsuan Tung
- Department of Radiology, Molecular Imaging Innovations Institute, Weill Cornell Medicine, New York, New York. .,Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - Vivek Mittal
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, New York. .,Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York.,Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, New York
| |
Collapse
|
26
|
Du B, Zhang W, Tung CH. Layer-by-layer construction of an oxygen-generating photo-responsive nanomedicine for enhanced photothermal and photodynamic combination therapy. Chem Commun (Camb) 2019; 55:5926-5929. [DOI: 10.1039/c9cc01461f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A multi-layered nanomedicine was designed for synergized oxygen-enhanced photodynamic therapy and photothermal therapy.
Collapse
Affiliation(s)
- Baoji Du
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine
- USA
| | - Weiqi Zhang
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine
- USA
| | - Ching-Hsuan Tung
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine
- USA
| |
Collapse
|
27
|
Villar-Alvarez E, Cambón A, Pardo A, Mosquera VX, Bouzas-Mosquera A, Topete A, Barbosa S, Taboada P, Mosquera V. Gold Nanorod-Based Nanohybrids for Combinatorial Therapeutics. ACS OMEGA 2018; 3:12633-12647. [PMID: 30411014 PMCID: PMC6217551 DOI: 10.1021/acsomega.8b01591] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/20/2018] [Indexed: 05/21/2023]
Abstract
In this work, multifunctional nanocarriers consisting of poly(sodium-4-styrenesulfonate) (PSS)/doxorubicin (DOXO)/poly-l-lysine hydrobromide (PLL)/hyaluronic acid (HA)-coated and (PSS/DOXO/PLL)2/HA-coated gold nanorods were assembled by the layer-by-layer technique with the aims of coupling the plasmonic photothermal properties of the metal nanoparticles for plasmonic hyperthermia and the chemoaction of drug DOXO for potential intended combinatorial cancer therapeutics in the future as well as providing different strategies for the controlled and sustained release of the cargo drug molecules. To do that, DOXO could be successfully loaded onto the hybrid nanoconstructs through electrostatic interactions with high efficiencies of up to ca. 78.3 ± 6.9% for the first formed drug layer and 56 ± 13% for the second one, with a total efficiency for the whole system [(PSS/DOXO/PLL)2/HA-coated NRs] of ca. 65.7 ± 1.4%. Nanohybrid internalization was observed to be enhanced by the outer HA layer, which is able to target the CD44 receptors widely overexpressed in some types of cancers as lung, breast, or ovarian ones. Hence, these nanohybrid systems might be versatile nanoplatforms to simultaneously deliver sufficient heat for therapeutic plasmonic hyperthermia and the anticancer drug. Two controlled mechanisms were proposed to modulate the release of the chemodrug, one by means of the enzymatic degradable character of the PLL layer and another by the modulation of the interactions between the polymeric layers through the exploitation of the optical properties of the hybrid particles under near infrared (NIR) laser irradiation. The combination of this bimodal therapeutic approach exerted a synergistic cytotoxic effect on both HeLa and MDA-MB-231 cancer cells in vitro. Cell death mechanisms were also analyzed, elucidating that plasmonic photothermal therapy induces cell necrosis, whereas DOXO activates the cell apoptotic pathway. Therefore, the present NIR laser-induced targeted cancer thermo/chemotherapy represents a novel targeted anticancer strategy with easy control on demand and suitable therapeutic efficacy.
Collapse
Affiliation(s)
- Eva Villar-Alvarez
- Grupo
de Física de Coloides y Polímeros, Departamento de Física
de Partículas, Facultad de Física e Instituto de Investigaciones
Sanitarias (IDIS), Universidad de Santiago
de Compostela, 15782 Santiago de Compostela, Spain
- E-mail: (E.V.-A.)
| | - Adriana Cambón
- Grupo
de Física de Coloides y Polímeros, Departamento de Física
de Partículas, Facultad de Física e Instituto de Investigaciones
Sanitarias (IDIS), Universidad de Santiago
de Compostela, 15782 Santiago de Compostela, Spain
| | - Alberto Pardo
- Grupo
de Física de Coloides y Polímeros, Departamento de Física
de Partículas, Facultad de Física e Instituto de Investigaciones
Sanitarias (IDIS), Universidad de Santiago
de Compostela, 15782 Santiago de Compostela, Spain
| | - Víctor X. Mosquera
- Departamento
de Cirugía Cardíaca, Complexo
Hospitalario Universitario A Coruña, Instituto
de Investigación Biomédica de A Coruña (INIBIC), A Coruña, Spain
| | - Alberto Bouzas-Mosquera
- Departamento
de Cirugía Cardíaca, Complexo
Hospitalario Universitario A Coruña, Instituto
de Investigación Biomédica de A Coruña (INIBIC), A Coruña, Spain
| | - Antonio Topete
- Departamento
de Fisiología, Centro Universitario
de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara, Jalisco, Mexico
| | - Silvia Barbosa
- Grupo
de Física de Coloides y Polímeros, Departamento de Física
de Partículas, Facultad de Física e Instituto de Investigaciones
Sanitarias (IDIS), Universidad de Santiago
de Compostela, 15782 Santiago de Compostela, Spain
| | - Pablo Taboada
- Grupo
de Física de Coloides y Polímeros, Departamento de Física
de Partículas, Facultad de Física e Instituto de Investigaciones
Sanitarias (IDIS), Universidad de Santiago
de Compostela, 15782 Santiago de Compostela, Spain
- E-mail: (P.T.)
| | - Víctor Mosquera
- Grupo
de Física de Coloides y Polímeros, Departamento de Física
de Partículas, Facultad de Física e Instituto de Investigaciones
Sanitarias (IDIS), Universidad de Santiago
de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
28
|
Munsell EV, Fang B, Sullivan MO. Histone-Mimetic Gold Nanoparticles as Versatile Scaffolds for Gene Transfer and Chromatin Analysis. Bioconjug Chem 2018; 29:3691-3704. [PMID: 30350573 DOI: 10.1021/acs.bioconjchem.8b00611] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Histone-inspired polymer assemblies (polyplexes) can regulate gene expression and subcellular transport in plasmids by harnessing the cellular machinery normally used for histone proteins. When grafted to polyplexes, histone tails promote nuclear accumulation, trigger plasmid DNA (pDNA) release, and enhance transcription. Herein, we developed multifunctional gold nanoparticles (AuNPs) decorated by histone motifs as histone-inspired scaffolds with improved pDNA binding, easy bioimaging, and increased potential for gene delivery and chromatin analysis applications. We hypothesized that polycationic AuNPs coupled to histone motifs would mimic the native presentation of these sequences on the histone octamer and thereby create structures with the capacity to both engage native histone effectors and condense pDNA into nucleosome-inspired nanostructures. AuNPs bearing ∼2 nm cores were prepared based on the well-established Brust-Schiffrin two-phase method involving tetrachloroaurate reduction in the presence of 1-pentanethiol. Solid phase peptide synthesis was employed to generate thiolated polycationic ligands and histone tail motifs, and the AuNPs and peptide ligands were combined in a two-step Murray place exchange reaction at various ratios to produce a collection of polycationic AuNPs modified with varying amounts of histone tails. Electron microscopy and thermal analyses demonstrated that these modified AuNPs exhibited tunable biochemical and biophysical properties that closely mimicked the properties of native histones. The histone-mimetic nanoscaffolds efficiently and sequence-specifically engaged histone effectors responsible for activating transcription. In addition, the nanoscaffolds condensed pDNA into complexes with high stability in the presence of physiological concentrations of heparin, a common extracellular polyanion. These combined properties of histone engagement and high stability led to a ∼6-fold enhancement in transfection efficiency as compared with typical polymeric transfection reagents, with the increased transfection efficiency correlated to the presence and amount of histone tails displayed on the surface of the nanoscaffolds. These findings demonstrate the utility of employing a biomimetic materials design approach to develop more effective and stable delivery vehicles for gene transfer and chromatin analysis applications.
Collapse
Affiliation(s)
- Erik V Munsell
- Department of Chemical and Biomolecular Engineering , University of Delaware , 150 Academy Street , Newark , Delaware 19716 , United States
| | - Bing Fang
- Department of Chemical and Biomolecular Engineering , University of Delaware , 150 Academy Street , Newark , Delaware 19716 , United States
| | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering , University of Delaware , 150 Academy Street , Newark , Delaware 19716 , United States
| |
Collapse
|
29
|
Kim HJ, Yi Y, Kim A, Miyata K. Small Delivery Vehicles of siRNA for Enhanced Cancer Targeting. Biomacromolecules 2018; 19:2377-2390. [PMID: 29864287 DOI: 10.1021/acs.biomac.8b00546] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Small interfering RNA (siRNA) drugs have been considered to treat various diseases in major organs. However, siRNA drugs developed for cancer therapy are hindered from proceeding to the clinic. To date, various delivery formulations have been developed from cationic lipids, polymers, and/or inorganic nanoparticles for systemic siRNA delivery to solid tumors. Most of these delivery vehicles do not generate small particle sizes and pharmacokinetics required for accumulation in target cancer cells compared with clinically tested anticancer drug-loaded polymeric micelles. This review describes the significance of small, long-circulating vehicles for efficient delivery of siRNA to cancer tissues via the enhanced permeability and retention (EPR) effect. We summarize recent biological evidence that supports the size effect of delivery vehicles in tumor microenvironments and introduce promising strategies for the construction of small vehicles with sizes of 10-50 nm. We then discuss the feasibility of these delivery vehicles with respect to translation to clinical trials.
Collapse
Affiliation(s)
- Hyun Jin Kim
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku, Tokyo 113-0033 , Japan
| | - Yu Yi
- Department of Materials Engineering, Graduate School of Engineering , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku, Tokyo 113-8656 , Japan.,CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety , National Center for Nanosciecne and Technology , No. 11 Beiyitiao , Zhongguancun, Beijing 100190 , China
| | - Ahram Kim
- Department of Materials Science, Graduate School of Pure and Applied Sciences , University of Tsukuba , 1-1-1 Tennoudai , Tsukuba , Ibaraki 305-8573 , Japan
| | - Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku, Tokyo 113-8656 , Japan
| |
Collapse
|
30
|
Das J, Choi YJ, Han JW, Reza AMMT, Kim JH. Nanoceria-mediated delivery of doxorubicin enhances the anti-tumour efficiency in ovarian cancer cells via apoptosis. Sci Rep 2017; 7:9513. [PMID: 28842609 PMCID: PMC5572723 DOI: 10.1038/s41598-017-09876-w] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 07/31/2017] [Indexed: 12/26/2022] Open
Abstract
Nanocarriers are widely used for effective delivery of anticancer drugs to tumours with potential to improve cancer treatment. Here, we developed a nanoceria (CeO2)-based system for delivery of the anti-cancer drug doxorubicin (DOX) to human ovarian cancer cells. Negatively charged nanoceria could conjugate with the cationic DOX via electrostatic interaction under physiological conditions, forming DOX-loaded nanoceria (CeO2/DOX). CeO2/DOX particles displayed nearly spherical shapes, along with superior drug-loading content (22.41%), loading efficiency (99.51%), and higher cellular uptake and drug release behaviours compared to free DOX. Moreover, DOX was released faster from CeO2/DOX under reductive acidic conditions (pH 5.0, 10 mM glutathione) than under physiological conditions (pH 7.4). The initial intracellular DOX concentration was higher in the free DOX groups than in the CeO2/DOX groups, but quickly reduced to 25% of the initial concentration after 24-h culture. By contrast, CeO2/DOX showed sustained DOX release over time and maintained a high intracellular DOX concentration for up to 72 h. In vitro assays showed that CeO2/DOX exhibited higher cell proliferation inhibition and apoptosis compared with free DOX. These results highlight DOX-loaded nanoceria as a promising therapeutic agent for cancer treatment.
Collapse
Affiliation(s)
- Joydeep Das
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul, 143-701, South Korea
| | - Yun-Jung Choi
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul, 143-701, South Korea
| | - Jae Woong Han
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul, 143-701, South Korea
| | - Abu Musa Md Talimur Reza
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul, 143-701, South Korea
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul, 143-701, South Korea.
| |
Collapse
|
31
|
Acharya R, Saha S, Ray S, Hazra S, Mitra MK, Chakraborty J. siRNA-nanoparticle conjugate in gene silencing: A future cure to deadly diseases? MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 76:1378-1400. [DOI: 10.1016/j.msec.2017.03.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 01/17/2017] [Accepted: 03/01/2017] [Indexed: 02/08/2023]
|
32
|
Latex nanoparticles surface modified via the layer-by-layer technique for two drugs loading. Colloids Surf A Physicochem Eng Asp 2017. [DOI: 10.1016/j.colsurfa.2017.04.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
33
|
Vinhas R, Fernandes AR, Baptista PV. Gold Nanoparticles for BCR-ABL1 Gene Silencing: Improving Tyrosine Kinase Inhibitor Efficacy in Chronic Myeloid Leukemia. MOLECULAR THERAPY. NUCLEIC ACIDS 2017. [PMID: 28624216 PMCID: PMC5436101 DOI: 10.1016/j.omtn.2017.05.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction of tyrosine kinase inhibitors for chronic myeloid leukemia treatment is associated with a 63% probability of maintaining a complete cytogenetic response, meaning that over 30% patients require an alternative methodology to overcome resistance, tolerance, or side effects. Considering the potential of nanotechnology in cancer treatment and the benefits of a combined therapy with imatinib, a nanoconjugate was designed to achieve BCR-ABL1 gene silencing. Gold nanoparticles were functionalized with a single-stranded DNA oligonucleotide that selectively targets the e14a2 BCR-ABL1 transcript expressed by K562 cells. This gold (Au)-nanoconjugate showed great efficacy in gene silencing that induced a significant increase in cell death. Variation of BCL-2 and BAX protein expression, an increase of caspase-3 activity, and apoptotic bodies in cells treated with the nanoconjugate demonstrate its aptitude for inducing apoptosis on K562 BCR-ABL1-expressing cells. Moreover, the combination of the silencing Au-nanoconjugate with imatinib prompted a decrease of imatinib IC50. This Au-nanoconjugate was also capable of inducing the loss of viability of imatinib-resistant K562 cells. This strategy shows that combination of Au-nanoconjugate and imatinib make K562 cells more vulnerable to chemotherapy and that the Au-nanoconjugate alone may overcome imatinib-resistance mechanisms, thus providing an effective treatment for chronic myeloid leukemia patients who exhibit drug tolerance.
Collapse
Affiliation(s)
- Raquel Vinhas
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus da Caparica, 2829-516 Caparica, Portugal
| | - Alexandra R Fernandes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus da Caparica, 2829-516 Caparica, Portugal.
| | - Pedro V Baptista
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus da Caparica, 2829-516 Caparica, Portugal.
| |
Collapse
|
34
|
Lin M, Gao Y, Diefenbach TJ, Shen JK, Hornicek FJ, Park YI, Xu F, Lu TJ, Amiji M, Duan Z. Facial Layer-by-Layer Engineering of Upconversion Nanoparticles for Gene Delivery: Near-Infrared-Initiated Fluorescence Resonance Energy Transfer Tracking and Overcoming Drug Resistance in Ovarian Cancer. ACS APPLIED MATERIALS & INTERFACES 2017; 9:7941-7949. [PMID: 28177223 DOI: 10.1021/acsami.6b15321] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Development of multidrug resistance (MDR) contributes to the majority of treatment failures in clinical chemotherapy. We report facial layer-by-layer engineered upconversion nanoparticles (UCNPs) for near-infrared (NIR)-initiated tracking and delivery of small interfering RNA (siRNA) to enhance chemotherapy efficacy by silencing the MDR1 gene and resensitizing resistant ovarian cancer cells to drug. Layer-by-layer engineered UCNPs were loaded with MDR1 gene-silencing siRNA (MDR1-siRNA) by electrostatic interaction. The delivery vehicle enhances MDR1-siRNA cellular uptake, protects MDR1-siRNA from nuclease degradation, and promotes endosomal escape for silencing the MDR gene. The intrinsic photon upconversion of UCNPs provides an unprecedented opportunity for monitoring intracellular attachment and release of MDR1-siRNA by NIR-initiated fluorescence resonance energy transfer occurs between donor UCNPs and acceptor fluorescence dye-labeled MDR1-siRNA. Enhanced chemotherapeutic efficacy in vitro was demonstrated by cell viability assay. The developed delivery vehicle holds great potential in delivery and imaging-guided tracking of therapeutic gene targets for effective treatment of drug-resistant cancers.
Collapse
Affiliation(s)
| | | | - Thomas J Diefenbach
- Ragon Institute of MGH, Massachusetts Institute of Technology (MIT) and Harvard University , Boston, Massachusetts 02139, United States
| | | | | | - Yong Il Park
- School of Chemical Engineering, Chonnam National University , Gwangju 61186, Republic of Korea
| | | | | | - Mansoor Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University , Boston, Massachusetts 02115, United States
| | | |
Collapse
|
35
|
Mendes R, Fernandes AR, Baptista PV. Gold Nanoparticle Approach to the Selective Delivery of Gene Silencing in Cancer-The Case for Combined Delivery? Genes (Basel) 2017; 8:E94. [PMID: 28257109 PMCID: PMC5368698 DOI: 10.3390/genes8030094] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/25/2017] [Accepted: 02/23/2017] [Indexed: 01/30/2023] Open
Abstract
Gene therapy arises as a great promise for cancer therapeutics due to its potential to silence genes involved in tumor development. In fact, there are some pivotal gene drivers that suffer critical alterations leading to cell transformation and ultimately to tumor growth. In this vein, gene silencing has been proposed as an active tool to selectively silence these molecular triggers of cancer, thus improving treatment. However, naked nucleic acid (DNA/RNA) sequences are reported to have a short lifetime in the body, promptly degraded by circulating enzymes, which in turn speed up elimination and decrease the therapeutic potential of these drugs. The use of nanoparticles for the effective delivery of these silencers to the specific target locations has allowed researchers to overcome this issue. Particularly, gold nanoparticles (AuNPs) have been used as attractive vehicles for the target-specific delivery of gene-silencing moieties, alone or in combination with other drugs. We shall discuss current trends in AuNP-based delivery of gene-silencing tools, considering the promising road ahead without overlooking existing concerns for their translation to clinics.
Collapse
Affiliation(s)
- Rita Mendes
- UCIBIO, DCV, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal.
| | - Alexandra R Fernandes
- UCIBIO, DCV, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal.
| | - Pedro V Baptista
- UCIBIO, DCV, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal.
| |
Collapse
|
36
|
Qu F, Liu Y, Lao H, Wang Y, You J. Colorimetric detection of heparin with high sensitivity based on the aggregation of gold nanoparticles induced by polymer nanoparticles. NEW J CHEM 2017. [DOI: 10.1039/c7nj02381b] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The negatively charged heparin hinders the aggregation of Au nanoparticles induced by the cationic polymer nanodots.
Collapse
Affiliation(s)
- Fei Qu
- The Key Laboratory of Life-Organic Analysis
- Qufu Normal University
- Qufu 273165
- China
- Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine
| | - Yanqun Liu
- The Key Laboratory of Life-Organic Analysis
- Qufu Normal University
- Qufu 273165
- China
- Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine
| | - Haili Lao
- Department of Clinical Laboratory
- Binzhou Central Hospital
- Binzhou Medical College
- Binzhou 256600
- China
| | - Yaping Wang
- The Key Laboratory of Life-Organic Analysis
- Qufu Normal University
- Qufu 273165
- China
- Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine
| | - Jinmao You
- The Key Laboratory of Life-Organic Analysis
- Qufu Normal University
- Qufu 273165
- China
- Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine
| |
Collapse
|
37
|
Wen MM. Getting miRNA Therapeutics into the Target Cells for Neurodegenerative Diseases: A Mini-Review. Front Mol Neurosci 2016; 9:129. [PMID: 27920668 PMCID: PMC5118630 DOI: 10.3389/fnmol.2016.00129] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/08/2016] [Indexed: 11/29/2022] Open
Abstract
miRNAs play important roles in modulating gene expression in varying cellular processes and disease pathogenesis, including neurodegenerative diseases. Several miRNAs are expressed in the brain, control brain development and are identified as important biomarkers in the pathogenesis of motor—and neuro-cognitive diseases such as Alzheimer’s (AD), Huntington’s and Parkinson’s diseases (PD) and amyotrophic lateral sclerosis. These remarkable miRNAs could be used as diagnostic markers and therapeutic targeting potential for many stressful and untreatable progressive neurodegenerative diseases. To modulate these miRNA activities, there are currently two strategies involved; first one is to therapeutically restore the suppressed miRNA level by miRNA mimics (agonist), and the other one is to inhibit miRNA function by using anti-miR (antagonist) to repress overactive miRNA function. However, RNAi-based therapeutics often faces in vivo instability because naked nucleic acids are subject to enzyme degradation before reaching the target sites. Therefore, an effective, safe and stable bio-responsive delivery system is necessary to protect the nucleic acids from serum degradation and assist their entrance to the cells. Since neuronal cells are non-regenerating, to design engineered miRNAs to be delivered to the central nervous system (CNS) for long term gene expression and knockdown is representing an enormous challenge for scientists. This article provides an insight summary on some of the innovative strategies employed to deliver miRNA into target cells. These viral and non-viral carrier systems hold promise in RNA therapy delivery for neurodegenerative diseases.
Collapse
Affiliation(s)
- Ming Ming Wen
- Department of Pharmaceutics, Pharos University in Alexandria Alexandria, Egypt
| |
Collapse
|
38
|
Correa S, Dreaden EC, Gu L, Hammond PT. Engineering nanolayered particles for modular drug delivery. J Control Release 2016; 240:364-386. [PMID: 26809005 PMCID: PMC6450096 DOI: 10.1016/j.jconrel.2016.01.040] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 01/07/2023]
Abstract
Layer-by-layer (LbL) based self-assembly of nanoparticles is an emerging and powerful method to develop multifunctional and tissue responsive nanomedicines for a broad range of diseases. This unique assembly technique is able to confer a high degree of modularity, versatility, and compositional heterogeneity to nanoparticles via the sequential deposition of alternately charged polyelectrolytes onto a colloidal template. LbL assembly can provide added functionality by directly incorporating a range of functional materials within the multilayers including nucleic acids, synthetic polymers, polypeptides, polysaccharides, and functional proteins. These materials can be used to generate hierarchically complex, heterogeneous thin films on an extensive range of both traditional and novel nanoscale colloidal templates, providing the opportunity to engineer highly precise systems capable of performing the numerous tasks required for systemic drug delivery. In this review, we will discuss the recent advancements towards the development of LbL nanoparticles for drug delivery and diagnostic applications, with a special emphasis on the incorporation of biostability, active targeting, desirable drug release kinetics, and combination therapies into LbL nanomaterials. In addition to these topics, we will touch upon the next steps for the translation of these systems towards the clinic.
Collapse
Affiliation(s)
- Santiago Correa
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, United States; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Erik C Dreaden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, United States; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Li Gu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, United States; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, United States; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States; Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, MA 02139, United States.
| |
Collapse
|
39
|
Lee SK, Law B, Tung CH. Versatile Nanodelivery Platform to Maximize siRNA Combination Therapy. Macromol Biosci 2016; 17. [PMID: 27654639 DOI: 10.1002/mabi.201600294] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 08/23/2016] [Indexed: 12/31/2022]
Abstract
The unsatisfactory outcomes of typical multiple cytotoxic chemotherapeutic combination therapies used to treat patients have fostered a need for new unconventional combinations of therapeutic agents. Among the candidates, siRNA has been widely discussed and tested. However, the right time right place codelivery of siRNA with other types of active ingredients is challenging because of the possible differences among their physiochemical and pharmacodynamics properties. To accomplish a synergistic cytotoxic effect, a nanoassembly is thus designed to codeliver siRNA with other therapeutic agents. A siRNA, targeting prosurvival gene for the p75 neurotrophin receptor, and an organelle-fusing peptide, targeting mitochondria, are layered onto a nanotemplate by charge-charge interaction, followed by a layer of CD44 targeting ligand. The formulated triple-functional nanomedicine is efficiently internalized by the CD44 expressing triple-negative breast cancer cells. The encapsulated siRNA and the pro-apoptotic peptide are released inside cells, silencing the intended prosurvival gene, and inducing apoptosis by fusing the mitochondrial membrane, respectively. A synergistic effect is achieved by this three-agent combination. The design of the developed multifunctional nanomedicine can be generalized to deliver other siRNA and drugs for a maximum therapeutic combination with minimal off-targeting effects.
Collapse
Affiliation(s)
- Seung Koo Lee
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, 413 East 69th Street, Box 290, New York, NY, 10021, USA
| | - Benedict Law
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, 413 East 69th Street, Box 290, New York, NY, 10021, USA
| | - Ching-Hsuan Tung
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, 413 East 69th Street, Box 290, New York, NY, 10021, USA
| |
Collapse
|
40
|
Das J, Han JW, Choi YJ, Song H, Cho SG, Park C, Seo HG, Kim JH. Cationic lipid-nanoceria hybrids, a novel nonviral vector-mediated gene delivery into mammalian cells: investigation of the cellular uptake mechanism. Sci Rep 2016; 6:29197. [PMID: 27380727 PMCID: PMC4933920 DOI: 10.1038/srep29197] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 06/14/2016] [Indexed: 01/30/2023] Open
Abstract
Gene therapy is a promising technique for the treatment of various diseases. The development of minimally toxic and highly efficient non-viral gene delivery vectors is the most challenging undertaking in the field of gene therapy. Here, we developed dimethyldioctadecylammonium bromide (DODAB)-nanoceria (CeO2) hybrids as a new class of non-viral gene delivery vectors. These DODAB-modified CeO2 nanoparticles (CeO2/DODAB) could effectively compact the pDNA, allowing for highly efficient gene transfection into the selected cell lines. The CeO2/DODAB nanovectors were also found to be non-toxic and did not induce ROS formation as well as any stress responsive and pro-survival signaling pathways. The overall vector performance of CeO2/DODAB nanohybrids was comparable with lipofectamine and DOTAP, and higher than calcium phosphate and DEAE-dextran for transfecting small plasmids. The increased cellular uptake of the nanovector/DNA complexes through clathrin- and caveolae-mediated endocytosis and subsequent release from the endosomes further support the increased gene transfection efficiency of the CeO2/DODAB vectors. Besides, CeO2/DODAB nanovectors could transfect genes in vivo without any sign of toxicity. Taken together, this new nano-vector has the potential to be used for gene delivery in biomedical applications.
Collapse
Affiliation(s)
- Joydeep Das
- Department of Stem Cell and Regenerative Biology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 143-701, South Korea
| | - Jae Woong Han
- Department of Stem Cell and Regenerative Biology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 143-701, South Korea
| | - Yun-Jung Choi
- Department of Stem Cell and Regenerative Biology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 143-701, South Korea
| | - Hyuk Song
- Department of Stem Cell and Regenerative Biology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 143-701, South Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 143-701, South Korea
| | - Chankyu Park
- Department of Stem Cell and Regenerative Biology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 143-701, South Korea
| | - Han Geuk Seo
- Department of Stem Cell and Regenerative Biology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 143-701, South Korea
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 143-701, South Korea
| |
Collapse
|
41
|
Wang Q, Zhou Y, Song B, Zhong Y, Wu S, Cui R, Cong H, Su Y, Zhang H, He Y. Linking Subcellular Disturbance to Physiological Behavior and Toxicity Induced by Quantum Dots in Caenorhabditis elegans. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:3143-3154. [PMID: 27121203 DOI: 10.1002/smll.201600766] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Indexed: 06/05/2023]
Abstract
The wide-ranging applications of fluorescent semiconductor quantum dots (QDs) have triggered increasing concerns about their biosafety. Most QD-related toxicity studies focus on the subcellular processes in cultured cells or global physiological effects on whole animals. However, it is unclear how QDs affect subcellular processes in living organisms, or how the subcellular disturbance contributes to the overall toxicity. Here the behavior and toxicity of QDs of three different sizes in Caenorhabditis elegans (C. elegans) are systematically investigated at both the systemic and the subcellular level. Specifically, clear size-dependent distribution and toxicity of the QDs in the digestive tract are observed. Short-term exposure of QDs leads to acute toxicity on C. elegans, yet incurring no lasting, irreversible damage. In contrast, chronic exposure of QDs severely inhibits development and shortens lifespan. Subcellular analysis reveals that endocytosis and nutrition storage are disrupted by QDs, which likely accounts for the severe deterioration in growth and longevity. This work reveals that QDs invasion disrupts key subcellular processes in living organisms, and may cause permanent damage to the tissues and organs over long-term retention. The findings provide invaluable information for safety evaluations of QD-based applications and offer new opportunities for design of novel nontoxic nanoprobes.
Collapse
Affiliation(s)
- Qin Wang
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yanfeng Zhou
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Bin Song
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yiling Zhong
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Sicong Wu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Rongrong Cui
- Laboratory of Physical Biology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
| | - Haixia Cong
- Laboratory of Physical Biology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
| | - Yuanyuan Su
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Huimin Zhang
- Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou, 215123, China
| | - Yao He
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
42
|
Zhao J, Feng SS. Nanocarriers for delivery of siRNA and co-delivery of siRNA and other therapeutic agents. Nanomedicine (Lond) 2016. [PMID: 26214357 DOI: 10.2217/nnm.15.61] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A major problem in cancer treatment is the multidrug resistance. siRNA inhibitors have great advantages to solve the problem, if the bottleneck of their delivery could be well addressed by the various nanocarriers. Moreover, co-delivery of siRNA together with the various anticancer agents in one nanocarrier may maximize their additive or synergistic effect. This review provides a comprehensive summary on the state-of-the-art of the nanocarriers, which may include prodrugs, micelles, liposomes, dendrimers, nanohydrogels, solid lipid nanoparticles, nanoparticles of biodegradable polymers and nucleic acid nanocarriers for delivery of siRNA and co-delivery of siRNA together with anticancer agents with focus on synthesis of the nanocarrier materials, design and characterization, in vitro and in vivo evaluation, and prospect and challenges of nanocarriers.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore 117576, Singapore
| | - Si-Shen Feng
- Department of Chemical & Biomolecular Engineering, Faculty of Engineering, National University of Singapore, Singapore 117576, Singapore.,International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
43
|
Badwaik VD, Aicart E, Mondjinou YA, Johnson MA, Bowman VD, Thompson DH. Structure-property relationship for in vitro siRNA delivery performance of cationic 2-hydroxypropyl-β-cyclodextrin: PEG-PPG-PEG polyrotaxane vectors. Biomaterials 2016; 84:86-98. [PMID: 26826298 PMCID: PMC4755830 DOI: 10.1016/j.biomaterials.2015.11.032] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 11/22/2015] [Accepted: 11/25/2015] [Indexed: 11/20/2022]
Abstract
Nanoparticle-mediated siRNA delivery is a promising therapeutic approach, however, the processes required for transport of these materials across the numerous extracellular and intracellular barriers are poorly understood. Efficient delivery of siRNA-containing nanoparticles would ultimately benefit from an improved understanding of how parameters associated with these barriers relate to the physicochemical properties of the nanoparticle vectors. We report the synthesis of three Pluronic(®)-based, cholesterol end-capped cationic polyrotaxanes (PR(+)) threaded with 2-hydroxypropyl-β-cyclodextrin (HPβCD) for siRNA delivery. The biological data showed that PR(+):siRNA complexes were well tolerated (∼90% cell viability) and produced efficient silencing (>80%) in HeLa-GFP and NIH 3T3-GFP cell lines. We further used a multi-parametric approach to identify relationships between the PR(+) structure, PR(+):siRNA complex physical properties, and biological activity. Small angle X-ray scattering and cryoelectron microscopy studies reveal periodicity and lamellar architectures for PR(+):siRNA complexes, whereas the biological assays, ζ potential measurements, and imaging studies suggest that silencing efficiency is influenced by the effective charge ratio (ρeff), polypropylene oxide (PO) block length, and central PO block coverage (i.e., rigidity) of the PR(+) core. We infer from our findings that more compact PR(+):siRNA nanostructures arising from lower molecular weight, rigid rod-like PR(+) polymer cores produce improved silencing efficiency relative to higher molecular weight, more flexible PR(+) vectors of similar effective charge. This study demonstrates that PR(+):siRNA complex formulations can be produced having higher performance than Lipofectamine(®) 2000, while maintaining good cell viability and siRNA sequence protection in cell culture.
Collapse
Affiliation(s)
- Vivek D Badwaik
- Department of Chemistry, Multi-disciplinary Cancer Research Facility, Bindley Bioscience Center, 1203 W. State Street, West Lafayette, IN 47907, USA
| | - Emilio Aicart
- Department of Chemistry, Multi-disciplinary Cancer Research Facility, Bindley Bioscience Center, 1203 W. State Street, West Lafayette, IN 47907, USA
| | - Yawo A Mondjinou
- Department of Chemistry, Multi-disciplinary Cancer Research Facility, Bindley Bioscience Center, 1203 W. State Street, West Lafayette, IN 47907, USA
| | - Merrell A Johnson
- Department of Physics, Indiana University-Purdue University Indianapolis, IN 46202, USA
| | - Valorie D Bowman
- Discovery Park, Hockmeyer Hall of Structural Biology, Purdue University, West Lafayette, IN 47907, USA
| | - David H Thompson
- Department of Chemistry, Multi-disciplinary Cancer Research Facility, Bindley Bioscience Center, 1203 W. State Street, West Lafayette, IN 47907, USA.
| |
Collapse
|
44
|
Shaat H, Mostafa A, Moustafa M, Gamal-Eldeen A, Emam A, El-Hussieny E, Elhefnawi M. Modified gold nanoparticles for intracellular delivery of anti-liver cancer siRNA. Int J Pharm 2016; 504:125-33. [PMID: 27036397 DOI: 10.1016/j.ijpharm.2016.03.051] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 03/25/2016] [Accepted: 03/27/2016] [Indexed: 11/25/2022]
Abstract
To overcome the rapid enzymatic degradation and low transfection efficiency of siRNA, the delivery carriers for siRNA is a therapeutic demand to increase its stability. Gold nanoparticles (AuNPs) modified by branched polyethyleneimine (bPEI) were developed as an efficient and safe intracellular delivery carriers for siRNA. The current study implied that siRNA designed against an oncogene c-Myc could be delivered by a modified AuNPs complex without significant cytotoxicity. The comparative semi-quantitative and quantitative real time PCR were used to measure the c-Myc gene expression after transfection with naked siRNA and siRNA/bPEI/AuNPs, but AuNPs interfered with PCR. However, the c-Myc protein translation was successfully detected in the transfected HuH7 cells with naked siRNA and siRNA/bPEI/AuNPs and it was found to be inhibited by siRNA/bPEI/AuNPs more than naked siRNA. The results validate the successful silencing of c-Myc gene. Accordingly, it may confirm the promising and effective delivery of siRNA by bPEI/AuNPs. The complex enhances the cellular uptake of siRNA without significant cytotoxicity and confirms that bPEI modified AuNPs could be used as a good candidate for safe cellular delivery of siRNA.
Collapse
Affiliation(s)
- Hanan Shaat
- Chemistry Department, Faculty of Science, Benha University, Benha, Egypt; Nanomedicine and Tissue Engineering Laboratory, Medical Research Centre of excellence, National Research Centre (NRC), Cairo, Egypt
| | - Amany Mostafa
- Nanomedicine and Tissue Engineering Laboratory, Medical Research Centre of excellence, National Research Centre (NRC), Cairo, Egypt; Ceramics Department, NRC, Dokki, Cairo, Egypt,.
| | - Moustafa Moustafa
- Chemistry Department, Faculty of Science, Benha University, Benha, Egypt
| | - Amira Gamal-Eldeen
- Cancer Biology and Genetics Laboratory Centre of Excellence for Advanced Sciences, NRC, Cairo, Egypt; Biochemistry Department, NRC, Dokki, Cairo, Egypt
| | - Ahmed Emam
- Nanomedicine and Tissue Engineering Laboratory, Medical Research Centre of excellence, National Research Centre (NRC), Cairo, Egypt; Ceramics Department, NRC, Dokki, Cairo, Egypt
| | - Enas El-Hussieny
- Zoology Department, Faculty of Science, Ain-Shams University, Cairo, Egypt
| | - Mahmoud Elhefnawi
- Biomedical Informatics and Chemo-Informatics Laboratory, Center of Excellence for advanced Sciences, NRC, Dokki, Cairo, Egypt,; Informatics and System Department, NRC, Dokki, Cairo, Egypt.
| |
Collapse
|
45
|
Jiang Y, Huo S, Hardie J, Liang XJ, Rotello VM. Progress and perspective of inorganic nanoparticle-based siRNA delivery systems. Expert Opin Drug Deliv 2016; 13:547-59. [PMID: 26735861 PMCID: PMC4914043 DOI: 10.1517/17425247.2016.1134486] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Small interfering RNA (siRNA) is an effective method for regulating the expression of proteins, even "undruggable" ones that are nearly impossible to target through traditional small molecule therapeutics. Delivery to the cell and then to the cytosol is the primary requirement for realization of therapeutic potential of siRNA. AREAS COVERED We summarize recent advances in the design of inorganic nanoparticle with surface functionality and physicochemical properties engineered for siRNA delivery. Specifically, we discuss the main approaches developed so far to load siRNA into/onto NPs, and NP surface chemistry engineered for enhanced intracellular siRNA delivery, endosomal escape, and targeted delivery of siRNA to disease cells and tissues. EXPERT OPINION Several challenges remain in developing inorganic NPs for efficient and effective siRNA delivery. Getting the material to the chosen site is important, however the greatest hurdle may well be delivery into the cytosol, either through efficient endosomal escape or by direct cytosolic siRNA delivery. Effective delivery at the organismic and cellular level coupled with biocompatible vehicles with low immunogenic response will facilitate the clinical translation of RNAi for the treatment of genetic diseases.
Collapse
Affiliation(s)
- Ying Jiang
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Shuaidong Huo
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
- Chinese Academy of Sciences (CAS) Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No.11, First North Road, Zhongguancun, Beijing, 100190, China
| | - Joseph Hardie
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No.11, First North Road, Zhongguancun, Beijing, 100190, China
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| |
Collapse
|
46
|
|
47
|
Abstract
Small interfering RNA (siRNA)-mediated gene silencing has shown prominent therapeutic effects in treating various diseases. However, adequate delivery and persistent gene silencing remain challenging. A nanoparticle-based delivery system which assembled by layering siRNAs between protease degradable polypeptides to show ultra-long gene silencing effect in vivo is developed. Gold nanoparticle is used as a scaffold for its unique properties including uniform size, biocompatibility, ready synthesis, and easy functionalization. A simple layer-by-layer fabrication approach, based on the electrostatic interaction between positively and negatively charged polymers, is applied to package the therapeutic siRNAs.
Collapse
Affiliation(s)
- Seung Koo Lee
- Department of Radiology, Molecular Imaging Innovations Institute, Weill Cornell Medical College, New York, NY, 10021, USA
| | - Ching-Hsuan Tung
- Department of Radiology, Molecular Imaging Innovations Institute, Weill Cornell Medical College, New York, NY, 10021, USA.
| |
Collapse
|
48
|
Affiliation(s)
- Xuan Yang
- The
Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | | | - Bo Pang
- The
Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | | | - Younan Xia
- The
Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| |
Collapse
|
49
|
Jaganathan S. Bioresorbable polyelectrolytes for smuggling drugs into cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2015; 44:1080-97. [PMID: 25961363 DOI: 10.3109/21691401.2015.1011801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
There is ample evidence that biodegradable polyelectrolyte nanocapsules are multifunctional vehicles which can smuggle drugs into cells, and release them upon endogenous activation. A large number of endogenous stimuli have already been tested in vitro, and in vivo research is escalating. Thus, the interest in the design of intelligent polyelectrolyte multilayer (PEM) drug delivery systems is clear. The need of the hour is a systematic translation of PEM-based drug delivery systems from the lab to clinical studies. Reviews on multifarious stimuli that can trigger the release of drugs from such systems already exist. This review summarizes the available literature, with emphasis on the recent progress in PEM-based drug delivery systems that are receptive in the presence of endogenous stimuli, including enzymes, glucose, glutathione, pH, and temperature, and addresses different active and passive drug targeting strategies. Insights into the current knowledge on the diversified endogenous approaches and methodological challenges may bring inspiration to resolve issues that currently bottleneck the successful implementation of polyelectrolytes into the catalog of third-generation drug delivery systems.
Collapse
Affiliation(s)
- Sripriya Jaganathan
- a SRM Research Institute, SRM University , Kattankulathur, 603203 , Chennai , Tamil Nadu , India
| |
Collapse
|
50
|
Lin CM, Kao WC, Yeh CA, Chen HJ, Lin SZ, Hsieh HH, Sun WS, Chang CH, Hung HS. Hyaluronic acid-fabricated nanogold delivery of the inhibitor of apoptosis protein-2 siRNAs inhibits benzo[a]pyrene-induced oncogenic properties of lung cancer A549 cells. NANOTECHNOLOGY 2015; 26:105101. [PMID: 25693888 DOI: 10.1088/0957-4484/26/10/105101] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Benzo[a]pyrene (BaP), a component of cooking oil fumes (COF), promotes lung cancer cell proliferation and survival via the induction of inhibitor of apoptosis protein-2 (IAP-2) proteins. Thus knockdown of IAP-2 would be a promising way to battle against lung cancer caused by COF. Functionalized gold nanoparticle (AuNP) is an effective delivery system for bio-active materials. Here, biocompatible hyaluronic acid (HA) was fabricated into nanoparticles to increase the target specificity by binding to CD44-over-expressed cancer cells. IAP-2-specific small-interfering RNA (siRNAs) or fluorescein isothiocyanate (FITC) were then incorporated into AuNP-HA. Conjugation of IAP-2 siRNA into AuNPs-HA was verified by the UV-vis spectrometer and Fourier transform infrared spectrometer. Further studies showed that AuNP-HA/FITC were effectively taken up by A549 cells through CD44-mediated endocytosis. Incubation of BaP-challenged cells with AuNP-HA-IAP-2 siRNAs silenced the expression of IAP-2, decreased cell proliferation and triggered pronounced cell apoptosis by the decrease in Bcl-2 protein and the increase in Bax protein as well as the active form of caspases-3. The BaP-elicited cell migration and enzymatic activity of the secreted matrix metalloproteinase-2 were also substantially suppressed by treatment with AuNP-HA-IAP-2 siRNAs. These results indicated that IAP-2 siRNAs can be efficiently delivered into A549 cells by functionalized AuNP-HA to repress the IAP-2 expression and BaP-induced oncogenic events, suggesting the potential therapeutic application of IAP-2 siRNA or other siRNA-conjugated AuNP-HA composites to COF-induced lung cancer and other gene-caused diseases in the future.
Collapse
Affiliation(s)
- Chung-Ming Lin
- Department of Biotechnology, Ming Chuan University, Taoyuan, Taiwan, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|