51
|
Liu D, Liu J, Ma B, Deng B, Leng X, Kong D, Liu L. A simple self-adjuvanting biomimetic nanovaccine self-assembled with the conjugate of phospholipids and nucleotides can induce a strong cancer immunotherapeutic effect. Biomater Sci 2021; 9:84-92. [PMID: 33016303 DOI: 10.1039/d0bm01333a] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Biomimetic nanoparticles have potential applications in many fields due to their favorable properties. Here, we developed a self-adjuvanting biomimetic anti-tumor nanovaccine, which was self-assembled with an amphiphilic conjugate synthesized with the phospholipids of 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) and hydrophilic Toll-like receptor (TLR9) agonist CpG ODN. The nanovaccine could not only provide effective initial antigen stimulation and sustained long-term antigen supply with a controlled release, but also induce antigen cross-presentation via the MHC-I pathway initiating CD8+ T-cell responses. Moreover, the dense nucleotide shell around the nanovaccine could promote antigen endocytosis via various receptor-mediated pathways into dendritic cells. CpG ODN interacted with TLR9 triggering the cytokine secretion of TNF-α and IL-10, which further boosted the anti-tumor humoral and cellular immune responses, which led to a significant tumor suppressive effect and remarkable survival prolongation. So, this nanovaccine self-assembled with phospholipid-nucleotide amphiphiles can serve as a safe, simple and efficient approach for anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Dan Liu
- The Tianjin Key Laboratory of Biomaterials, Institute of Biomedical Engineering, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin 300192, China.
| | | | | | | | | | | | | |
Collapse
|
52
|
Li YX, Wei Y, Zhong R, Li L, Pang HB. Transportan Peptide Stimulates the Nanomaterial Internalization into Mammalian Cells in the Bystander Manner through Macropinocytosis. Pharmaceutics 2021; 13:552. [PMID: 33920021 PMCID: PMC8070997 DOI: 10.3390/pharmaceutics13040552] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 01/12/2023] Open
Abstract
Covalent coupling with cell-penetrating peptides (CPPs) has been a common strategy to facilitate the cell entry of nanomaterial and other macromolecules. Though efficient, this strategy requires chemical modifications on nanomaterials, which is not always desired for their applications. Recent studies on a few cationic CPPs have revealed that they can stimulate the cellular uptake of nanoparticles (NPs) simply via co-administration (bystander manner), which bypasses the requirement of chemical modification. In this study, we investigated the other classes of CPPs and discovered that transportan (TP) peptide, an amphiphilic CPP, also exhibited such bystander activities. When simply co-administered, TP peptide enabled the cells to engulf a variety of NPs, as well as common solute tracers, while these payloads had little or no ability to enter the cells by themselves. This result was validated in vitro and ex vivo, and TP peptide showed no physical interaction with co-administered NPs (bystander cargo). We further explored the cell entry mechanism for TP peptide and its bystander cargo, and showed that it was mediated by a receptor-dependent macropinocytosis process. Together, our findings improve the understanding of TP-assisted cell entry, and open up a new avenue to apply this peptide for nanomaterial delivery.
Collapse
Affiliation(s)
- Yue-Xuan Li
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (Y.-X.L.); (Y.W.)
| | - Yushuang Wei
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (Y.-X.L.); (Y.W.)
| | - Rui Zhong
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA; (R.Z.); (L.L.)
| | - Ling Li
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA; (R.Z.); (L.L.)
| | - Hong-Bo Pang
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; (Y.-X.L.); (Y.W.)
| |
Collapse
|
53
|
Wang Y, Zhang H, Xiao W, Liu Y, Zhou Y, He X, Xia X, Gong T, Wang L, Gao H. Unmasking CSF protein corona: Effect on targeting capacity of nanoparticles. J Control Release 2021; 333:352-361. [PMID: 33823221 DOI: 10.1016/j.jconrel.2021.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 03/25/2021] [Accepted: 04/01/2021] [Indexed: 12/19/2022]
Abstract
Among biological fluids, cerebrospinal fluid (CSF) not only protects and support brain, but also plays a pivotal role in intracerebral interaction of various nano-drug carriers. However, it is still uncertain how protein corona from CSF affects the targeting capability of functionalized nanoparticles (NPs). So, two types of polystyrene NPs, including PEGylated polystyrene NPs (PN) and transferrin (Tf)-modified PN (PT), were used to obtain protein corona-coated NPs, by incubating with CSF in vivo and in vitro. Strikingly, both the corona-coated NPs recovered in vivo and in vitro completely lost their active targeting characteristics towards bEnd.3 and C6 cells. Charge-, clathrin- and energy-mediated endocytosis contributed to the improved uptake efficiency of PT, whereas this enhancement in uptake of PT was disappeared after the formation of CSF protein corona. Moreover, serum albumin, which were found both in vivo and in vitro CSF corona, could mediate and facilitate the internalization of corona-coated NPs. Overall, these results have distinctly confirmed that the formation of CSF protein corona could cause the loss of active targeting specificity by shielding the targeting groups on the surface of polystyrene NPs and alter their cellular uptake by other non-specific internalization pathways.
Collapse
Affiliation(s)
- Yazhen Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China
| | - Huilin Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China
| | - Wei Xiao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China
| | - Yuwei Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China
| | - Yang Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China
| | - Xueqin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China
| | - Xue Xia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China
| | - Ling Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China.
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China.
| |
Collapse
|
54
|
Jia Y, Chen L, Liu J, Li W, Gu H. DNA-catalyzed efficient production of single-stranded DNA nanostructures. Chem 2021. [DOI: 10.1016/j.chempr.2020.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
55
|
Lacroix A, Sleiman HF. DNA Nanostructures: Current Challenges and Opportunities for Cellular Delivery. ACS NANO 2021; 15:3631-3645. [PMID: 33635620 DOI: 10.1021/acsnano.0c06136] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
DNA nanotechnology has produced a wide range of self-assembled structures, offering unmatched possibilities in terms of structural design. Because of their programmable assembly and precise control of size, shape, and function, DNA particles can be used for numerous biological applications, including imaging, sensing, and drug delivery. While the biocompatibility, programmability, and ease of synthesis of nucleic acids have rapidly made them attractive building blocks, many challenges remain to be addressed before using them in biological conditions. Enzymatic hydrolysis, low cellular uptake, immune cell recognition and degradation, and unclear biodistribution profiles are yet to be solved. Rigorous methodologies are needed to study, understand, and control the fate of self-assembled DNA structures in physiological conditions. In this review, we describe the current challenges faced by the field as well as recent successes, highlighting the potential to solve biology problems or develop smart drug delivery tools. We then propose an outlook to drive the translation of DNA constructs toward preclinical design. We particularly believe that a detailed understanding of the fate of DNA nanostructures within living organisms, achieved through thorough characterization, is the next required step to reach clinical maturity.
Collapse
Affiliation(s)
- Aurélie Lacroix
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec H3A 0B8, Canada
| | - Hanadi F Sleiman
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec H3A 0B8, Canada
| |
Collapse
|
56
|
Vincent MP, Bobbala S, Karabin NB, Frey M, Liu Y, Navidzadeh JO, Stack T, Scott EA. Surface chemistry-mediated modulation of adsorbed albumin folding state specifies nanocarrier clearance by distinct macrophage subsets. Nat Commun 2021; 12:648. [PMID: 33510170 PMCID: PMC7844416 DOI: 10.1038/s41467-020-20886-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 12/21/2020] [Indexed: 01/30/2023] Open
Abstract
Controlling nanocarrier interactions with the immune system requires a thorough understanding of the surface properties that modulate protein adsorption in biological fluids, since the resulting protein corona redefines cellular interactions with nanocarrier surfaces. Albumin is initially one of the dominant proteins to adsorb to nanocarrier surfaces, a process that is considered benign or beneficial by minimizing opsonization or inflammation. Here, we demonstrate the surface chemistry of a model nanocarrier can be engineered to stabilize or denature the three-dimensional conformation of adsorbed albumin, which respectively promotes evasion or non-specific clearance in vivo. Interestingly, certain common chemistries that have long been considered to convey stealth properties denature albumin to promote nanocarrier recognition by macrophage class A1 scavenger receptors, providing a means for their eventual removal from systemic circulation. We establish that the surface chemistry of nanocarriers can be specified to modulate adsorbed albumin structure and thereby tune clearance by macrophage scavenger receptors.
Collapse
Affiliation(s)
- Michael P Vincent
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Sharan Bobbala
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Nicholas B Karabin
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Molly Frey
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, 60208, USA
| | - Yugang Liu
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Justin O Navidzadeh
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Trevor Stack
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA.
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL, 60208, USA.
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA.
- Simpson Querrey Institute, Northwestern University, Chicago, IL, 60611, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
57
|
Thai HB, Kim KR, Hong KT, Voitsitskyi T, Lee JS, Mao C, Ahn DR. Kidney-Targeted Cytosolic Delivery of siRNA Using a Small-Sized Mirror DNA Tetrahedron for Enhanced Potency. ACS CENTRAL SCIENCE 2020; 6:2250-2258. [PMID: 33376785 PMCID: PMC7760472 DOI: 10.1021/acscentsci.0c00763] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Indexed: 05/30/2023]
Abstract
A proper intracellular delivery method with target tissue specificity is critical to utilize the full potential of therapeutic molecules including siRNAs while minimizing their side effects. Herein, we prepare four small-sized DNA tetrahedrons (sTds) by self-assembly of different sugar backbone-modified oligonucleotides and screened them to develop a platform for kidney-targeted cytosolic delivery of siRNA. An in vivo biodistribution study revealed the kidney-specific accumulation of mirror DNA tetrahedron (L-sTd). Low opsonization of L-sTd in serum appeared to avoid liver clearance and keep its size small enough to be filtered through the glomerular basement membrane (GBM). After GBM filtration, L-sTd could be delivered into tubular cells by endocytosis. The kidney preference and the tubular cell uptake property of the mirror DNA nanostructure could be successfully harnessed for kidney-targeted intracellular delivery of p53 siRNA to treat acute kidney injury (AKI) in mice. Therefore, L-sTd could be a promising platform for kidney-targeted cytosolic delivery of siRNA to treat renal diseases.
Collapse
Affiliation(s)
- Hien Bao
Dieu Thai
- Center
for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Kyoung-Ran Kim
- Center
for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Kyung Tae Hong
- Division
of Biomedical Science and Technology, KIST School, Korea University of Science and Technology (UST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Taras Voitsitskyi
- Division
of Biomedical Science and Technology, KIST School, Korea University of Science and Technology (UST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Jun-Seok Lee
- Division
of Biomedical Science and Technology, KIST School, Korea University of Science and Technology (UST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Korea
- Molecular
Recognition Research Center, Korea Institute
of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Korea
| | - Chengde Mao
- Department
of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Dae-Ro Ahn
- Center
for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Korea
- Division
of Biomedical Science and Technology, KIST School, Korea University of Science and Technology (UST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Korea
| |
Collapse
|
58
|
Ponedal A, Zhu S, Sprangers AJ, Wang XQ, Yeo DC, Lio DCS, Zheng M, Capek M, Narayan SP, Meckes B, Paller AS, Xu C, Mirkin CA. Attenuation of Abnormal Scarring Using Spherical Nucleic Acids Targeting Transforming Growth Factor Beta 1. ACS APPLIED BIO MATERIALS 2020; 3:8603-8610. [PMID: 33709070 DOI: 10.1021/acsabm.0c00990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Abnormal scarring is a consequence of dysregulation in the wound healing process, with limited options for effective and noninvasive therapies. Given the ability of spherical nucleic acids (SNAs) to penetrate skin and regulate gene expression within, we investigated whether gold-core SNAs (AuSNAs) and liposome-core SNAs (LSNAs) bearing antisense oligonucleotides targeting transforming growth factor beta 1 (TGF-β1) can function as a topical therapy for scarring. Importantly, both SNA constructs appreciably downregulated TGF-β1 protein expression in primary hypertrophic and keloid scar fibroblasts in vitro. In vivo, topically applied AuSNAs and LSNAs downregulated TGF-β1 protein expression levels and improved scar histology as determined by the scar elevation index. These data underscore the potential of SNAs as a localized, self-manageable treatment for skin-related diseases and disorders that are driven by increased gene expression.
Collapse
Affiliation(s)
- Adam Ponedal
- Department of Chemical and Biological Engineering and International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
| | - Shengshuang Zhu
- International Institute for Nanotechnology and Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Anthony J Sprangers
- International Institute for Nanotechnology and Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Xiao-Qi Wang
- Department of Dermatology, Northwestern University, Chicago, Illinois 60611, United States
| | - David C Yeo
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 639798
| | - Daniel C S Lio
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 639798
| | - Mengjia Zheng
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 639798
| | - Matthew Capek
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
| | - Suguna P Narayan
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
| | - Brian Meckes
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States; Department of Biomedical Engineering, University of North Texas, Denton, Texas 76203, United States
| | - Amy S Paller
- Department of Dermatology, Northwestern University, Chicago, Illinois 60611, United States
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Chad A Mirkin
- Department of Chemical and Biological Engineering, International Institute for Nanotechnology, Department of Materials Science and Engineering, and Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
59
|
Yan J, Tan YL, Lin MJ, Xing H, Jiang JH. A DNA-mediated crosslinking strategy to enhance cellular delivery and sensor performance of protein spherical nucleic acids. Chem Sci 2020; 12:1803-1809. [PMID: 34163943 PMCID: PMC8179099 DOI: 10.1039/d0sc04977h] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Intracellular delivery of enzymes is essential for protein-based diagnostic and therapeutic applications. Protein-spherical nucleic acids (ProSNAs) defined by protein core and dense shell of oligonucleotides have been demonstrated as a promising vehicle-free enzyme delivery platform. In this work, we reported a crosslinking strategy to vastly improve both delivery efficiency and intracellular sensor performance of ProSNA. By assembling individual ProSNA with lactate oxidase (LOX) core into a nanoscale particle, termed as crosslinked SNA (X-SNA), the enzyme delivery efficiency increased up to 5-6 times higher. The LOX X-SNA was later demonstrated as a ratiometric probe for quantitative detection of lactate in living cells. More importantly, X-SNA probe showed significantly improved sensor performance with signal-to-noise ratio 4 times as high as ProSNA when detecting intracellular lactate.
Collapse
Affiliation(s)
- Jing Yan
- Institute of Chemical Biology and Nanomedicine, Hunan University Changsha 410082 P. R. China .,State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University Changsha 410082 P. R. China
| | - Ya-Ling Tan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University Changsha 410082 P. R. China
| | - Min-Jie Lin
- Institute of Chemical Biology and Nanomedicine, Hunan University Changsha 410082 P. R. China .,State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University Changsha 410082 P. R. China
| | - Hang Xing
- Institute of Chemical Biology and Nanomedicine, Hunan University Changsha 410082 P. R. China .,State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University Changsha 410082 P. R. China
| | - Jian-Hui Jiang
- Institute of Chemical Biology and Nanomedicine, Hunan University Changsha 410082 P. R. China .,State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University Changsha 410082 P. R. China
| |
Collapse
|
60
|
Liessi N, Maragliano L, Castagnola V, Bramini M, Benfenati F, Armirotti A. Isobaric Labeling Proteomics Allows a High-Throughput Investigation of Protein Corona Orientation. Anal Chem 2020; 93:784-791. [PMID: 33285070 PMCID: PMC7818227 DOI: 10.1021/acs.analchem.0c03134] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
The
formation of the biomolecular corona represents a crucial factor
in controlling the biological interactions and trafficking of nanomaterials.
In this context, the availability of key epitopes exposed on the surface
of the corona, and able to engage the biological machinery, is important
to define the biological fate of the material. While the full biomolecular
corona composition can be investigated by conventional bottom-up proteomics,
the assessment of the spatial orientation of proteins in the corona
in a high-throughput fashion is still challenging. In this work, we
show that labeling corona proteins with isobaric tags in their native
conditions and analyzing the MS/MS spectra of tryptic peptides allow
an easy and high-throughput assessment of the inner/outer orientation
of the corresponding proteins in the original corona. We put our results
in the context of what is currently known of the protein corona of
graphene-based nanomaterials. Our conclusions are in line with previous
data and were confirmed by in silico calculations.
Collapse
Affiliation(s)
- Nara Liessi
- Analytical Chemistry Lab, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Luca Maragliano
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy.,Department of Life and Environmental Sciences, Marche Polytechnic University, Via Brecce Bianche, 60131 Ancona, Italy
| | - Valentina Castagnola
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy.,IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Mattia Bramini
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy.,Department of Applied Physics, Faculty of Sciences, University of Granada, Fuente Nueva s/n, 18071 Granada, Spain
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy.,IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Andrea Armirotti
- Analytical Chemistry Lab, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| |
Collapse
|
61
|
Li H, Fan J, Buhl EM, Huo S, Loznik M, Göstl R, Herrmann A. DNA hybridization as a general method to enhance the cellular uptake of nanostructures. NANOSCALE 2020; 12:21299-21305. [PMID: 33064117 DOI: 10.1039/d0nr02405h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The biomedical application of nanoparticles (NPs) for diagnosis and therapy is considerably stalled by their inefficient cellular internalization. Many strategies to overcome this obstacle have been developed but are not generally applicable to different NP systems, consequently underlining the need for a universal method that enhances NP entry into cells. Here we describe a method to increase NP cellular uptake via strand hybridization between DNA-functionalized NPs and cells that bear the respective complementary sequence incorporated into the membrane. By this, the NPs bind efficiently to the cellular surface enhancing internalization of three completely different NP types: DNA tetrahedrons, gold (Au) NPs, and polystyrene (PS) NPs. We show that our approach is a simple and generalizable strategy that can be applied to virtually every functionalizable NP system.
Collapse
Affiliation(s)
- Hongyan Li
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52056 Aachen, Germany.
| | | | | | | | | | | | | |
Collapse
|
62
|
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: 108] [Impact Index Per Article: 27.0] [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
|
63
|
Breznica P, Koliqi R, Daka A. A review of the current understanding of nanoparticles protein corona composition. Med Pharm Rep 2020; 93:342-350. [PMID: 33225259 PMCID: PMC7664725 DOI: 10.15386/mpr-1756] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/22/2020] [Accepted: 06/30/2020] [Indexed: 12/25/2022] Open
Abstract
Upon entering into the biological environments, the surface of the nanoparticles is immediately coated with proteins and form the so-called a protein corona due to which a nanoparticle changes its “synthetic” identity to a new “biological” identity. Different types of nanoparticles have different protein binding profiles, which is why they have different protein corona composition and therefore it cannot be said that there is a universal protein corona. The composition and amount of protein in the corona depends on the physical and chemical characteristics of the nanoparticles, the type of biological medium and the exposure time. Protein corona increases the diameter but also changes the composition of the surface of the nanoparticles and these changes affect biodistribution, efficacy, and toxicity of the nanoparticles.
Collapse
Affiliation(s)
- Pranvera Breznica
- Department of Pharmaceutical Chemistry, Pharmacy Division, Faculty of Medicine, "Hasan Prishtina" University, Prishtina, Republic of Kosovo
| | - Rozafa Koliqi
- Department of Clinical Pharmacy and Biopharmacy, Pharmacy Division, Faculty of Medicine, "Hasan Prishtina" University, Prishtina, Republic of Kosovo
| | - Arlinda Daka
- Department of Clinical Pharmacy and Biopharmacy, Pharmacy Division, Faculty of Medicine, "Hasan Prishtina" University, Prishtina, Republic of Kosovo
| |
Collapse
|
64
|
Griffith DM, Jayaram DT, Spencer DM, Pisetsky DS, Payne CK. DNA-nanoparticle interactions: Formation of a DNA corona and its effects on a protein corona. Biointerphases 2020; 15:051006. [PMID: 33003950 PMCID: PMC7863680 DOI: 10.1116/6.0000439] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/29/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022] Open
Abstract
There has been much recent interest in the protein "corona," the nonspecific adsorption of proteins on the surface of nanoparticles used in biological applications. This research investigates an analogous DNA corona. We find that particles (200 nm and 1 μm) incubated with DNA form a DNA corona, with a higher concentration of DNA adsorbed on the surface of cationic nanoparticles. With protein present, a combined DNA and protein corona is formed although DNA in solution displaces protein from the nanoparticle surface. Displacement of protein from the nanoparticle surface is dependent on the concentration of DNA in solution and was also observed for planar surfaces. Overall, we expect this investigation of the DNA corona to be important for nanomedicine applications, as well as disease states, especially systemic lupus erythematosus, in which biological particles with bound DNA are important mediators of inflammation and thrombosis.
Collapse
Affiliation(s)
- Darbi M Griffith
- Department of Mechanical Engineering and Materials Science, Duke University, Hudson Hall, Durham, North Carolina 27708
| | - Dhanya T Jayaram
- Department of Mechanical Engineering and Materials Science, Duke University, Hudson Hall, Durham, North Carolina 27708
| | - Diane M Spencer
- Division of Rheumatology and Immunology, Duke University Medical Center, 508 Fulton St, Durham, North Carolina 27707
| | - David S Pisetsky
- Division of Rheumatology and Immunology, Duke University Medical Center, and Medical Research Service, Durham VA Medical Center, 508 Fulton St, Durham, North Carolina 27707
| | - Christine K Payne
- Department of Mechanical Engineering and Materials Science, Duke University, Hudson Hall, Durham, North Carolina 27708
| |
Collapse
|
65
|
Hosseinzadeh L, Nemati H, Nemati N, Sadeghi M. Spherical Gold Nanoparticles: Small Interfering RNA Delivery in Regulation of the Tumor Necrosis Factor-Alpha Gene Expression. J Interferon Cytokine Res 2020; 40:490-496. [PMID: 32865449 DOI: 10.1089/jir.2020.0090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Proinflammatory cytokines are signaling molecules that are expelled from immune cells like macrophages and other types of cells. Tumor necrosis factor-alpha (TNF-α) is overexpressed during inflammation caused by inflammatory diseases. Therefore, the regulation of TNF-α has a key role in inflammation. The use and target delivery of small interfering RNAs (siRNAs) provide many effectual treatment benefits in the regulation of gene expression in cells. In this study, we used siRNA nanoparticle conjugates in the regulation of gene expression and inflammation. We first prepared safe fusion ribonucleic acid interference carrier, spherical nucleic acid nanoparticle conjugates (SNA-NCs), to enhance the perforation of siRNA into the macrophages and their ability to target TNF-α gene regulation. Furthermore, the suppression of the TNF-α gene was monitored after curing macrophages by SNA-NCs. Gene expression was carried out by real-time polymerase chain reaction in cells and the levels of TNF-α were investigated by the enzyme-linked immunosorbent assay (ELISA) method. This study indicated that the SNA-NCs were safe and very stable. TNF-α siRNA could significantly regulate gene expression in cells to form SNA-NCs. The results indicated that TNF-α gene expression downregulated to 93.40% ± 1.45%, 66.06% ± 0.95%, and 35.76% ± 1.09% in the presence of 0.1, 1, and 10 nM siRNA, respectively. The proliferation of macrophages and subsequently expression of TNF-α were significant for the formation of inflammation. These findings showed that the use of SNA-NC siRNA might ameliorate the inflammatory disease by suppression of gene expression and functional activity of macrophage generation.
Collapse
Affiliation(s)
- Leila Hosseinzadeh
- Pharmaceutical Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Houshang Nemati
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Narges Nemati
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoud Sadeghi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
66
|
Zhang J, Ma R, Blanchard A, Petree J, Jo H, Salaita K. Conditional Deoxyribozyme-Nanoparticle Conjugates for miRNA-Triggered Gene Regulation. ACS APPLIED MATERIALS & INTERFACES 2020; 12:37851-37861. [PMID: 32803952 PMCID: PMC8287654 DOI: 10.1021/acsami.0c07609] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
DNA-nanoparticle (NP) conjugates have been used to knockdown gene expression transiently and effectively, making them desirable tools for gene regulation therapy. Because DNA-NPs are constitutively active and are rapidly taken up by most cell types, they offer limited control in terms of tissue or cell type specificity. To take a step toward solving this issue, we incorporate toehold-mediated strand exchange, a versatile molecular programming modality, to switch the DNA-NPs from an inactive state to an active state in the presence of a specific RNA input. Because many transcripts are unique to cell subtype or disease state, this approach could one day lead to responsive nucleic acid therapeutics with enhanced specificity. As a proof of concept, we designed conditional deoxyribozyme-nanoparticles (conditional DzNPs) that knockdown tumor necrosis factor α (TNFα) mRNA upon miR-33 triggering. We demonstrate toehold-mediated strand exchange and restoration of TNFα DNAzyme activity in the presence of miR-33 trigger, with optimization of the preparation, configuration, and toehold length of conditional DzNPs. Our results indicate specific and strong ON/OFF response of conditional DzNPs to the miR-33 trigger in buffer. Furthermore, we demonstrate endogenous miR-33-triggered knockdown of TNFα mRNA in mouse macrophages, implying the potential of conditional gene regulation applications using these DzNPs.
Collapse
Affiliation(s)
- Jiahui Zhang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Rong Ma
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | - Aaron Blanchard
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Jessica Petree
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Khalid Salaita
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Department of Chemistry, Emory University, Atlanta, GA, USA
| |
Collapse
|
67
|
Awotunde O, Okyem S, Chikoti R, Driskell JD. Role of Free Thiol on Protein Adsorption to Gold Nanoparticles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:9241-9249. [PMID: 32686419 DOI: 10.1021/acs.langmuir.0c01550] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Protein-gold nanoparticle (AuNP) bioconjugates have many potential applications in nanomedicine. A thorough understanding of the interaction between the protein and the AuNP is critical to engineering these functional bioconjugates with desirable properties. In this work, we investigate the role of free thiols presented by the protein on the stability of the protein-AuNP conjugate. Human serum albumin (HSA) was modified with 2-iminothiolane (Traut's reagent) to introduce additional thiols onto the protein surface, and three variants of HSA were synthesized to present 1, 5, and 20 free thiols by controlling the molar excess of the chemical modifier. Protein exchange studies on AuNPs were conducted using these HSA species and an IgG antibody which exhibited 10 free thiols. Antibody-AuNP conjugates were synthesized, purified, and dispersed in solutions containing each of the HSA species. No protein exchange was detected with the HSA or modified HSA containing 5 thiols; however, 85% of the antibody was displaced on the AuNP surface by the extensively thiolated HSA presenting 20 free thiols. Furthermore, the impact of the protein adsorption sequence was probed in which each of the HSA species were preadsorbed onto the AuNP and dispersed in a solution of antibody. The antibody fully displaced the HSA with a single thiol from the AuNP within 3 h, required 24 h to completely displace the modified HSA containing 5 thiols, and was unable to displace the modified HSA containing 20 thiols. These results indicate that the number of Au-S interactions governs the binding interaction between the protein and the AuNP. This work provides further insight into the protein-AuNP binding mechanism and identifies important design principles for engineered proteins to optimize bioconjugates.
Collapse
Affiliation(s)
- Olatunde Awotunde
- Department of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| | - Samuel Okyem
- Department of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| | - Rishika Chikoti
- Department of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| | - Jeremy D Driskell
- Department of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| |
Collapse
|
68
|
The Impact of Ag Nanoparticles and CdTe Quantum Dots on Expression and Function of Receptors Involved in Amyloid-β Uptake by BV-2 Microglial Cells. MATERIALS 2020; 13:ma13143227. [PMID: 32698417 PMCID: PMC7412234 DOI: 10.3390/ma13143227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/09/2020] [Accepted: 07/16/2020] [Indexed: 02/08/2023]
Abstract
Microglial cells clear the brain of pathogens and harmful debris, including amyloid-β (Aβ) deposits that are formed during Alzheimer’s disease (AD). We studied the expression of Msr1, Ager and Cd36 receptors involved in Aβ uptake and expression of Cd33 protein, which is considered a risk factor in AD. The effect of silver nanoparticles (AgNP) and cadmium telluride quantum dots (CdTeQD) on the expression of the above receptors and Aβ uptake by microglial cells was investigated. Absorption of Aβ and NP was confirmed by confocal microscopy. AgNP, but not CdTeQD, caused a decrease in Aβ accumulation. By using a specific inhibitor—polyinosinic acid—we demonstrated that Aβ and AgNP compete for scavenger receptors. Real-time PCR showed up-regulation of Cd33 and Cd36 gene expression after treatment with CdTeQD for 24 h. Analysis of the abundance of the receptors on the cell surface revealed that AgNP treatment significantly reduced the presence of Msr1, Cd33, Ager and Cd36 receptors (6 and 24 h), whereas CdTeQD increased the levels of Msr1 and Cd36 (24 h). To summarize, we showed that AgNP uptake competes with Aβ uptake by microglial cells and consequently can impair the removal of the aggregates. In turn, CdTeQD treatment led to the accumulation of proinflammatory Cd36 protein on the cell surface.
Collapse
|
69
|
Ribbon of DNA Lattice on Gold Nanoparticles for Selective Drug Delivery to Cancer Cells. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202005624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
70
|
Zhang S, Chen C, Xue C, Chang D, Xu H, Salena BJ, Li Y, Wu Z. Ribbon of DNA Lattice on Gold Nanoparticles for Selective Drug Delivery to Cancer Cells. Angew Chem Int Ed Engl 2020; 59:14584-14592. [DOI: 10.1002/anie.202005624] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/21/2020] [Indexed: 12/25/2022]
Affiliation(s)
- Shuxin Zhang
- Cancer Metastasis Alert and Prevention Center Fujian Provincial Key Laboratory of Cancer Metastasis, Chemoprevention and Chemotherapy National & Local Joint Biomedical Engineering Research Center on, Photodynamic Technologies Pharmaceutical Photocatalysis of State Key Laboratory of, Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou 350002 China
| | - Chang Chen
- Cancer Metastasis Alert and Prevention Center Fujian Provincial Key Laboratory of Cancer Metastasis, Chemoprevention and Chemotherapy National & Local Joint Biomedical Engineering Research Center on, Photodynamic Technologies Pharmaceutical Photocatalysis of State Key Laboratory of, Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou 350002 China
| | - Chang Xue
- Cancer Metastasis Alert and Prevention Center Fujian Provincial Key Laboratory of Cancer Metastasis, Chemoprevention and Chemotherapy National & Local Joint Biomedical Engineering Research Center on, Photodynamic Technologies Pharmaceutical Photocatalysis of State Key Laboratory of, Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou 350002 China
| | - Dingran Chang
- Department of Biochemistry and Biomedical Sciences McMaster University 1280 Main Street West Hamilton Ontario L8S4K1 Canada
| | - Huo Xu
- Cancer Metastasis Alert and Prevention Center Fujian Provincial Key Laboratory of Cancer Metastasis, Chemoprevention and Chemotherapy National & Local Joint Biomedical Engineering Research Center on, Photodynamic Technologies Pharmaceutical Photocatalysis of State Key Laboratory of, Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou 350002 China
| | - Bruno J. Salena
- Department of Medicine McMaster University 1280 Main Street West Hamilton Ontario L8S4K1 Canada
| | - Yingfu Li
- Department of Biochemistry and Biomedical Sciences McMaster University 1280 Main Street West Hamilton Ontario L8S4K1 Canada
| | - Zai‐Sheng Wu
- Cancer Metastasis Alert and Prevention Center Fujian Provincial Key Laboratory of Cancer Metastasis, Chemoprevention and Chemotherapy National & Local Joint Biomedical Engineering Research Center on, Photodynamic Technologies Pharmaceutical Photocatalysis of State Key Laboratory of, Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou 350002 China
| |
Collapse
|
71
|
Qin L, Wang S, Dominguez D, Long A, Chen S, Fan J, Ahn J, Skakuj K, Huang Z, Lee A, Mirkin C, Zhang B. Development of Spherical Nucleic Acids for Prostate Cancer Immunotherapy. Front Immunol 2020; 11:1333. [PMID: 32733447 PMCID: PMC7362897 DOI: 10.3389/fimmu.2020.01333] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/26/2020] [Indexed: 01/02/2023] Open
Abstract
Although the strategy of therapeutic vaccination for the treatment of prostate cancer has advanced to and is available in the clinic (Sipuleucel-T), the efficacy of such therapy remains limited. Here, we develop Immunostimulatory Spherical Nucleic Acid (IS-SNA) nanostructures comprised of CpG oligonucleotides as adjuvant and prostate cancer peptide antigens, and evaluate their antitumor efficacy in syngeneic mouse models of prostate cancer. IS-SNAs with the specific structural feature of presenting both antigen and adjuvant CpG on the surface (hybridized model (HM) SNAs) induce stronger cytotoxic T lymphocyte (CTL) mediated antigen-specific killing of target cells than that for IS-SNAs with CpG on the surface and antigen encapsulated within the core (encapsulated model (EM) SNAs). Mechanistically, HM SNAs increase the co-delivery of CpG and antigen to dendritic cells over that for EM SNAs or admixtures of linear CpG and peptide, thereby improving cross-priming of antitumor CD8+ T cells. As a result, vaccination with HM SNAs leads to more effective antitumor immune responses in two prostate cancer models. These data demonstrate the importance of the structural positioning of peptide antigens together with adjuvants within IS-SNAs to the efficacy of IS-SNA-based cancer immunotherapy.
Collapse
Affiliation(s)
- Lei Qin
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Shuya Wang
- Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, IL, United States
| | - Donye Dominguez
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Alan Long
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Siqi Chen
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jie Fan
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jihae Ahn
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Kacper Skakuj
- Department of Chemistry, Northwestern University, Evanston, IL, United States
| | - Ziyin Huang
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, United States
| | - Andrew Lee
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, United States
| | - Chad Mirkin
- Department of Chemistry, Northwestern University, Evanston, IL, United States.,The International Institute for Nanotechnology, Northwestern University, Evanston, IL, United States
| | - Bin Zhang
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
72
|
Taschauer A, Polzer W, Pöschl S, Metz S, Tepe N, Decker S, Cyran N, Scholda J, Maier J, Bloß H, Anton M, Hofmann T, Ogris M, Sami H. Combined Chemisorption and Complexation Generate siRNA Nanocarriers with Biophysics Optimized for Efficient Gene Knockdown and Air-Blood Barrier Crossing. ACS APPLIED MATERIALS & INTERFACES 2020; 12:30095-30111. [PMID: 32515194 PMCID: PMC7467563 DOI: 10.1021/acsami.0c06608] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Current nucleic acid (NA) nanotherapeutic approaches face challenges because of shortcomings such as limited control on loading efficiency, complex formulation procedure involving purification steps, low load of NA cargo per nanoparticle, endosomal trapping, and hampered release inside the cell. When combined, these factors significantly limit the amount of biologically active NA delivered per cell in vitro, delivered dosages in vivo for a prolonged biological effect, and the upscalability potential, thereby warranting early consideration in the design and developmental phase. Here, we report a versatile nanotherapeutic platform, termed auropolyplexes, for improved and efficient delivery of small interfering RNA (siRNA). Semitelechelic, thiolated linear polyethylenimine (PEI) was chemisorbed onto gold nanoparticles to endow them with positive charge. A simple two-step complexation method offers tunable loading of siRNA at concentrations relevant for in vivo studies and the flexibility for inclusion of multiple functionalities without any purification steps. SiRNA was electrostatically complexed with these cationic gold nanoparticles and further condensed with polycation or polyethyleneglycol-polycation conjugates. The resulting auropolyplexes ensured complete complexation of siRNA into nanoparticles with a high load of ∼15,500 siRNA molecules/nanoparticle. After efficient internalization into the tumor cell, an 80% knockdown of the luciferase reporter gene was achieved. Auropolyplexes were applied intratracheally in Balb/c mice for pulmonary delivery, and their biodistribution were studied spatio-temporally and quantitatively by optical tomography. Auropolyplexes were well tolerated with ∼25% of the siRNA dose remaining in the lungs after 24 h. Importantly, siRNA was released from auropolyplexes in vivo and a fraction also crossed the air-blood barrier, which was then excreted via kidneys, whereas >97% of gold nanoparticles were retained in the lung. Linear PEI-based auropolyplexes offer a combination of successful endosomal escape and better biocompatibility profile in vivo. Taken together, combined chemisorption and complexation endow auropolyplexes with crucial biophysical attributes, enabling a versatile and upscalable nanogold-based platform for siRNA delivery in vitro and in vivo.
Collapse
Affiliation(s)
- Alexander Taschauer
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Wolfram Polzer
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Stefan Pöschl
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Slavica Metz
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Nathalie Tepe
- Department of Environmental Geosciences, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
| | - Simon Decker
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Norbert Cyran
- Core Facility Cell
Imaging and Ultrastructure Research (CIUS), University of Vienna, 1090 Vienna, Austria
| | - Julia Scholda
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Julia Maier
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Hermann Bloß
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Martina Anton
- Institutes of Molecular Immunology and Experimental Oncology, Klinikum
rechts der Isar, Technische Universität
München, 81675 Munich, Germany
| | - Thilo Hofmann
- Department of Environmental Geosciences, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
| | - Manfred Ogris
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
- Center for NanoScience (CeNS), Ludwig Maximilians
University, 80539 Munich, Germany
| | - Haider Sami
- Faculty of Life
Sciences, Center of Pharmaceutical Sciences, Department of Pharmaceutical
Chemistry, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| |
Collapse
|
73
|
Meyer CE, Liu J, Craciun I, Wu D, Wang H, Xie M, Fussenegger M, Palivan CG. Segregated Nanocompartments Containing Therapeutic Enzymes and Imaging Compounds within DNA-Zipped Polymersome Clusters for Advanced Nanotheranostic Platform. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1906492. [PMID: 32130785 DOI: 10.1002/smll.201906492] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/22/2019] [Indexed: 06/10/2023]
Abstract
Nanotheranostics is an emerging field that brings together nanoscale-engineered materials with biological systems providing a combination of therapeutic and diagnostic strategies. However, current theranostic nanoplatforms have serious limitations, mainly due to a mismatch between the physical properties of the selected nanomaterials and their functionalization ease, loading ability, or overall compatibility with bioactive molecules. Herein, a nanotheranostic system is proposed based on nanocompartment clusters composed of two different polymersomes linked together by DNA. Careful design and procedure optimization result in clusters segregating the therapeutic enzyme human Dopa decarboxylase (DDC) and fluorescent probes for the detection unit in distinct but colocalized nanocompartments. The diagnostic compartment provides a twofold function: trackability via dye loading as the imaging component and the ability to attach the cluster construct to the surface of cells. The therapeutic compartment, loaded with active DDC, triggers the cellular expression of a secreted reporter enzyme via production of dopamine and activation of dopaminergic receptors implicated in atherosclerosis. This two-compartment nanotheranostic platform is expected to provide the basis of a new treatment strategy for atherosclerosis, to expand versatility and diversify the types of utilizable active molecules, and thus by extension expand the breadth of attainable applications.
Collapse
Affiliation(s)
- Claire E Meyer
- Department of Chemistry, University of Basel, Mattenstrasse 24a, Basel, 4002, Switzerland
| | - Juan Liu
- Department of Chemistry, University of Basel, Mattenstrasse 24a, Basel, 4002, Switzerland
| | - Ioana Craciun
- Department of Chemistry, University of Basel, Mattenstrasse 24a, Basel, 4002, Switzerland
| | - Dalin Wu
- Department of Chemistry, University of Basel, Mattenstrasse 24a, Basel, 4002, Switzerland
| | - Hui Wang
- Department of Biosystems Science Engineering, ETHZ, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Mingqi Xie
- Department of Biosystems Science Engineering, ETHZ, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science Engineering, ETHZ, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Cornelia G Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 24a, Basel, 4002, Switzerland
| |
Collapse
|
74
|
Akpe V, Shiddiky MJA, Kim TH, Brown CL, Yamauchi Y, Cock IE. Cancer biomarker profiling using nanozyme containing iron oxide loaded with gold particles. J R Soc Interface 2020; 17:20200180. [PMID: 32574540 DOI: 10.1098/rsif.2020.0180] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Nanozymes are nanomaterials with intrinsic magnetism and superparamagnetic properties. In the presence of an external magnet, nanozyme particles aggregate and redisperse without a foreign attraction. We evaluated the performances of nanozyme by changing the biosensing platforms and substituting other biological variants for a complete cancer assay detection. We investigated the expression of morphological variants in the transmission of signals using an electrochemical method. The signal responses, including signal enhancement with the nanozyme (Au-Fe2O3), showed a wide capturing range (greater than 80%, from 102 to 105 cells ml-1 in phosphate-buffered saline buffer, pH 7.4). The platform showed a fast response time within a dynamic range of 10-105 cells ml-1 for the investigated T47D cancer cell line. We also obtained higher responses for anti-HER2 (human epidermal receptor 2)/streptavidin interface as the biosensing electrode in the presence of T47D cancer cells. The positive assay produced a sixfold increase in current output compared to the negative target or negative biological variant. We calculated the limit of detection at 0.4 U ml-1, and of quantitation at 4 U ml-1 (units per millilitre). However, blood volume amounts in clinical settings may constrain diagnosis and increase detection limit value significantly.
Collapse
Affiliation(s)
- Victor Akpe
- School of Environment and Science, Griffith University, Nathan Campus, Queensland 4111, Australia.,Environmental Futures Research Institute, Griffith University, Nathan Campus, Queensland 4111, Australia
| | - Muhammad J A Shiddiky
- School of Environment and Science, Griffith University, Nathan Campus, Queensland 4111, Australia.,Queensland Micro and Nanotechnology Centre, Griffith University, Nathan Campus, Queensland 4111, Australia
| | - Tak H Kim
- School of Environment and Science, Griffith University, Nathan Campus, Queensland 4111, Australia.,Environmental Futures Research Institute, Griffith University, Nathan Campus, Queensland 4111, Australia
| | - Christopher L Brown
- School of Environment and Science, Griffith University, Nathan Campus, Queensland 4111, Australia.,Environmental Futures Research Institute, Griffith University, Nathan Campus, Queensland 4111, Australia
| | - Yusuke Yamauchi
- School of Chemical Engineering and Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, Brisbane, Queensland 4072, Australia
| | - Ian E Cock
- School of Environment and Science, Griffith University, Nathan Campus, Queensland 4111, Australia.,Environmental Futures Research Institute, Griffith University, Nathan Campus, Queensland 4111, Australia
| |
Collapse
|
75
|
Franch O, Gutiérrez-Corbo C, Domínguez-Asenjo B, Boesen T, Jensen PB, Nejsum LN, Keller JG, Nielsen SP, Singh PR, Jha RK, Nagaraja V, Balaña-Fouce R, Ho YP, Reguera RM, Knudsen BR. DNA flowerstructure co-localizes with human pathogens in infected macrophages. Nucleic Acids Res 2020; 48:6081-6091. [PMID: 32402089 PMCID: PMC7293011 DOI: 10.1093/nar/gkaa341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 04/16/2020] [Accepted: 04/24/2020] [Indexed: 01/07/2023] Open
Abstract
Herein, we characterize the cellular uptake of a DNA structure generated by rolling circle DNA amplification. The structure, termed nanoflower, was fluorescently labeled by incorporation of ATTO488-dUTP allowing the intracellular localization to be followed. The nanoflower had a hydrodynamic diameter of approximately 300 nanometer and was non-toxic for all mammalian cell lines tested. It was internalized specifically by mammalian macrophages by phagocytosis within a few hours resulting in specific compartmentalization in phagolysosomes. Maximum uptake was observed after eight hours and the nanoflower remained stable in the phagolysosomes with a half-life of 12 h. Interestingly, the nanoflower co-localized with both Mycobacterium tuberculosis and Leishmania infantum within infected macrophages although these pathogens escape lysosomal degradation by affecting the phagocytotic pathway in very different manners. These results suggest an intriguing and overlooked potential application of DNA structures in targeted treatment of infectious diseases such as tuberculosis and leishmaniasis that are caused by pathogens that escape the human immune system by modifying macrophage biology.
Collapse
Affiliation(s)
- Oskar Franch
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark
| | | | | | - Thomas Boesen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark
- DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Pia Bomholt Jensen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark
| | - Lene N Nejsum
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Josephine Geertsen Keller
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Prakruti R Singh
- Department of Microbiology and Cell Biology, Indian Institute of Science & Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Rajiv Kumar Jha
- Department of Microbiology and Cell Biology, Indian Institute of Science & Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Valakunja Nagaraja
- Department of Microbiology and Cell Biology, Indian Institute of Science & Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | | | - Yi-Ping Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR
- Centre for Novel Biomaterials, The Chinese University of Hong Kong, Hong Kong SAR
| | | | - Birgitta Ruth Knudsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark
| |
Collapse
|
76
|
Kusmierz C, Bujold KE, Callmann CE, Mirkin CA. Defining the Design Parameters for in Vivo Enzyme Delivery Through Protein Spherical Nucleic Acids. ACS CENTRAL SCIENCE 2020; 6:815-822. [PMID: 32490197 PMCID: PMC7256953 DOI: 10.1021/acscentsci.0c00313] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Indexed: 05/19/2023]
Abstract
The translation of proteins as effective intracellular drug candidates is limited by the challenge of cellular entry and their vulnerability to degradation. To advance their therapeutic potential, cell-impermeable proteins can be readily transformed into protein spherical nucleic acids (ProSNAs) by densely functionalizing their surfaces with DNA, yielding structures that are efficiently taken up by cells. Because small structural changes in the chemical makeup of a conjugated ligand can affect the bioactivity of the associated protein, structure-activity relationships of the linker bridging the DNA and the protein surface and the DNA sequence itself are investigated on the ProSNA system. In terms of attachment chemistry, DNA-based linkers promote a sevenfold increase in cellular uptake while maintaining enzymatic activity in vitro as opposed to hexaethylene glycol (HEG, Spacer18) linkers. Additionally, the employment of G-quadruplex-forming sequences increases cellular uptake in vitro up to fourfold. When translating to murine models, the ProSNA with a DNA-only shell exhibits increased blood circulation times and higher accumulation in major organs, including lung, kidney, and spleen, regardless of sequence. Importantly, ProSNAs with an all-oligonucleotide shell retain their enzymatic activity in tissue, whereas the native protein loses all function. Taken together, these results highlight the value of structural design in guiding ProSNA biological fate and activity and represent a significant step forward in the development of intracellular protein-based therapeutics.
Collapse
Affiliation(s)
- Caroline
D. Kusmierz
- Department
of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- International
Institute for Nanotechnology, Northwestern
University, 2145 Sheridan
Road, Evanston, Illinois 60208, United States
| | - Katherine E. Bujold
- Department
of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- International
Institute for Nanotechnology, Northwestern
University, 2145 Sheridan
Road, Evanston, Illinois 60208, United States
| | - Cassandra E. Callmann
- Department
of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- International
Institute for Nanotechnology, Northwestern
University, 2145 Sheridan
Road, Evanston, Illinois 60208, United States
| | - Chad A. Mirkin
- Department
of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- International
Institute for Nanotechnology, Northwestern
University, 2145 Sheridan
Road, Evanston, Illinois 60208, United States
| |
Collapse
|
77
|
Thorp EB, Boada C, Jarbath C, Luo X. Nanoparticle Platforms for Antigen-Specific Immune Tolerance. Front Immunol 2020; 11:945. [PMID: 32508829 PMCID: PMC7251028 DOI: 10.3389/fimmu.2020.00945] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
Innovative approaches in nanoparticle design have facilitated the creation of new formulations of nanoparticles that are capable of selectively calibrating the immune response. These nanomaterials may be engineered to interact with specific cellular and molecular targets. Recent advancements in nanoparticle synthesis have enabled surface functionalization of particles that mimic the diversity of ligands on the cell surface. Platforms synthesized using these design principles, called "biomimetic" nanoparticles, have achieved increasingly sophisticated targeting specificity and cellular trafficking capabilities. This holds great promise for next generation therapies that seek to achieve immune tolerance. In this review, we discuss the importance of physical design parameters including size, shape, and biomimetic surface functionalization, on the biodistribution, safety and efficacy of biologic nanoparticles. We will also explore potential applications for immune tolerance for organ or stem cell transplantation.
Collapse
Affiliation(s)
- Edward B. Thorp
- Departments of Pathology & Pediatrics at Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Christian Boada
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, United States
| | - Clarens Jarbath
- Departments of Pathology & Pediatrics at Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Xunrong Luo
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, United States
- Duke Transplant Center, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
78
|
Liu J, Craciun I, Belluati A, Wu D, Sieber S, Einfalt T, Witzigmann D, Chami M, Huwyler J, Palivan CG. DNA-directed arrangement of soft synthetic compartments and their behavior in vitro and in vivo. NANOSCALE 2020; 12:9786-9799. [PMID: 32328600 DOI: 10.1039/d0nr00361a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
DNA has been widely used as a key tether to promote self-organization of super-assemblies with emergent properties. However, control of this process is still challenging for compartment assemblies and to date the resulting assemblies have unstable membranes precluding in vitro and in vivo testing. Here we present our approach to overcome these limitations, by manipulating molecular factors such as compartment membrane composition and DNA surface density, thereby controlling the size and stability of the resulting DNA-linked compartment clusters. The soft, flexible character of the polymer membrane and low number of ssDNA remaining exposed after cluster formation determine the interaction of these clusters with the cell surface. These clusters exhibit in vivo stability and lack of toxicity in a zebrafish model. To display the breadth of therapeutic applications attainable with our system, we encapsulated the medically established enzyme laccase within the inner compartment and demonstrated its activity within the clustered compartments. Most importantly, these clusters can interact selectively with different cell lines, opening a new strategy to modify and expand cellular functions by attaching such pre-organized soft DNA-mediated compartment clusters on cell surfaces for cell engineering or therapeutic applications.
Collapse
Affiliation(s)
- Juan Liu
- Department of Chemistry, University of Basel, Mattenstrasse 24a, Basel-4058, Switzerland.
| | - Ioana Craciun
- Department of Chemistry, University of Basel, Mattenstrasse 24a, Basel-4058, Switzerland.
| | - Andrea Belluati
- Department of Chemistry, University of Basel, Mattenstrasse 24a, Basel-4058, Switzerland.
| | - Dalin Wu
- Department of Chemistry, University of Basel, Mattenstrasse 24a, Basel-4058, Switzerland.
| | - Sandro Sieber
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, Basel-4056, Switzerland
| | - Tomaz Einfalt
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, Basel-4056, Switzerland
| | - Dominik Witzigmann
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, Basel-4056, Switzerland
| | - Mohamed Chami
- BioEM lab, Biozentrum, University of Basel, Mattenstrasse 26, Basel-4058, Switzerland
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, Basel-4056, Switzerland
| | - Cornelia G Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 24a, Basel-4058, Switzerland.
| |
Collapse
|
79
|
Dykman LA. Gold nanoparticles for preparation of antibodies and vaccines against infectious diseases. Expert Rev Vaccines 2020; 19:465-477. [PMID: 32306785 PMCID: PMC7196924 DOI: 10.1080/14760584.2020.1758070] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/16/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Vaccination remains very effective in stimulating protective immune responses against infections. An important task in antibody and vaccine preparation is to choose an optimal carrier that will ensure a high immune response. Particularly promising in this regard are nanoscale particle carriers. An antigen that is adsorbed or encapsulated by nanoparticles can be used as an adjuvant to optimize the immune response during vaccination. a very popular antigen carrier used for immunization and vaccination is gold nanoparticles, with are being used to make new vaccines against viral, bacterial, and parasitic infections. AREAS COVERED This review summarizes what is currently known about the use of gold nanoparticles as an antigen carrier and adjuvant to prepare antibodies in vivo and design vaccines against viral, bacterial, and parasitic infections. The basic principles, recent advances, and current problems in the use of gold nanoparticles are discussed. EXPERT OPINION Gold nanoparticles can be used as adjuvants to increase the effectiveness of vaccines by stimulating antigen-presenting cells and ensuring controlled antigen release. Studying the characteristics of the immune response obtained from the use of gold nanoparticles as a carrier and an adjuvant will permit the particles' potential for vaccine design to be increased.
Collapse
Affiliation(s)
- Lev A. Dykman
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, Saratov, Russia
| |
Collapse
|
80
|
Grafals-Ruiz N, Rios-Vicil CI, Lozada-Delgado EL, Quiñones-Díaz BI, Noriega-Rivera RA, Martínez-Zayas G, Santana-Rivera Y, Santiago-Sánchez GS, Valiyeva F, Vivas-Mejía PE. Brain Targeted Gold Liposomes Improve RNAi Delivery for Glioblastoma. Int J Nanomedicine 2020; 15:2809-2828. [PMID: 32368056 PMCID: PMC7185647 DOI: 10.2147/ijn.s241055] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 03/27/2020] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Glioblastoma (GBM) is the most common and lethal of the central nervous system (CNS) malignancies. The initiation, progression, and infiltration ability of GBMs are attributed in part to the dysregulation of microRNAs (miRNAs). Thus, targeting dysregulated miRNAs with RNA oligonucleotides (RNA interference, RNAi) has been proposed for GBM treatment. Despite promising results in the laboratory, RNA oligonucleotides have clinical limitations that include poor RNA stability and off-target effects. RNAi therapies against GBM confront an additional obstacle, as they need to cross the blood-brain barrier (BBB). METHODS Here, we developed gold-liposome nanoparticles conjugated with the brain targeting peptides apolipoprotein E (ApoE) and rabies virus glycoprotein (RVG). First, we functionalized gold nanoparticles with oligonucleotide miRNA inhibitors (OMIs), creating spherical nucleic acids (SNAs). Next, we encapsulated SNAs into ApoE, or RVG-conjugated liposomes, to obtain SNA-Liposome-ApoE and SNA-Liposome-RVG, respectively. We characterized each nanoparticle in terms of their size, charge, encapsulation efficiency, and delivery efficiency into U87 GBM cells in vitro. Then, they were administered intravenously (iv) in GBM syngeneic mice to evaluate their delivery efficiency to brain tumor tissue. RESULTS SNA-Liposomes of about 30-50 nm in diameter internalized U87 GBM cells and inhibited the expression of miRNA-92b, an aberrantly overexpressed miRNA in GBM cell lines and GBM tumors. Conjugating SNA-Liposomes with ApoE or RVG peptides increased their systemic delivery to the brain tumors of GBM syngeneic mice. SNA-Liposome-ApoE demonstrated to accumulate at higher extension in brain tumor tissues, when compared with non-treated controls, SNA-Liposomes, or SNA-Liposome-RVG. DISCUSSION SNA-Liposome-ApoE has the potential to advance the translation of miRNA-based therapies for GBM as well as other CNS disorders.
Collapse
Affiliation(s)
- Nilmary Grafals-Ruiz
- Department of Physiology, University of Puerto Rico, San Juan, Puerto Rico
- Comprehensive Cancer Center, University of Puerto Rico, San Juan, Puerto Rico
| | - Christian I Rios-Vicil
- Comprehensive Cancer Center, University of Puerto Rico, San Juan, Puerto Rico
- Department of Neurosurgery, University of Puerto Rico, San Juan, Puerto Rico
| | - Eunice L Lozada-Delgado
- Comprehensive Cancer Center, University of Puerto Rico, San Juan, Puerto Rico
- Department of Biology, University of Puerto Rico, San Juan, Puerto Rico
| | - Blanca I Quiñones-Díaz
- Comprehensive Cancer Center, University of Puerto Rico, San Juan, Puerto Rico
- Department of Biochemistry, University of Puerto Rico, San Juan, Puerto Rico
| | - Ricardo A Noriega-Rivera
- Comprehensive Cancer Center, University of Puerto Rico, San Juan, Puerto Rico
- Department of Biochemistry, University of Puerto Rico, San Juan, Puerto Rico
| | - Gabriel Martínez-Zayas
- Comprehensive Cancer Center, University of Puerto Rico, San Juan, Puerto Rico
- Department of Chemistry, University of Puerto Rico, San Juan, Puerto Rico
| | | | - Ginette S Santiago-Sánchez
- Comprehensive Cancer Center, University of Puerto Rico, San Juan, Puerto Rico
- Department of Biochemistry, University of Puerto Rico, San Juan, Puerto Rico
| | - Fatma Valiyeva
- Comprehensive Cancer Center, University of Puerto Rico, San Juan, Puerto Rico
| | - Pablo E Vivas-Mejía
- Comprehensive Cancer Center, University of Puerto Rico, San Juan, Puerto Rico
- Department of Biochemistry, University of Puerto Rico, San Juan, Puerto Rico
| |
Collapse
|
81
|
Lacroix A, Fakih HH, Sleiman HF. Detailed cellular assessment of albumin-bound oligonucleotides: Increased stability and lower non-specific cell uptake. J Control Release 2020; 324:34-46. [PMID: 32330572 DOI: 10.1016/j.jconrel.2020.04.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 04/11/2020] [Indexed: 01/04/2023]
Abstract
Conjugation of lipid moieties to nucleic-acid therapeutics increases their interaction with cellular membranes, enhances their uptake and influences in vivo distribution. Once injected in biological fluids, such modifications trigger the binding of various serum proteins, which in turn play a major role in determining the fate of oligonucleotides. Yet, the role played by each of these proteins, more than 300 in serum, remains to be elucidated. Albumin, the most abundant circulating protein is an attractive candidate to study, as it was previously used to enhance the therapeutic effect of various drugs. Herein, we present a thorough fluorescent-based methodology to study the effect of strong and specific albumin-binding on the fate and cellular uptake of DNA oligonucleotides. We synthesized a library of molecules that exhibit non-covalent binding to albumin, with affinities ranging from high (nanomolar) to none. Our results revealed that strong albumin binding can be used as a strategy to reduce degradation of oligonucleotides in physiological conditions caused by enzymes (nucleases), to reduce uptake and degradation by immune cells (macrophages) and to prevent non-specific uptake by cells. We believe that introducing protein-binding domains in oligonucleotides can be used as a strategy to control the fate of oligonucleotides in physiological environments. While our study focuses on albumin, we believe that such systematic studies, which elucidate the role of serum proteins systematically, will ultimately provide a toolbox to engineer the next-generation of therapeutic oligonucleotides, overcoming many of the barriers encountered by these therapeutics, such as stability, immunogenicity and off-target effects.
Collapse
Affiliation(s)
- Aurélie Lacroix
- Department of Chemistry, McGill University, 801 Sherbrooke St. W., Montréal, Québec H3A 0B8, Canada
| | - Hassan H Fakih
- Department of Chemistry, McGill University, 801 Sherbrooke St. W., Montréal, Québec H3A 0B8, Canada
| | - Hanadi F Sleiman
- Department of Chemistry, McGill University, 801 Sherbrooke St. W., Montréal, Québec H3A 0B8, Canada.
| |
Collapse
|
82
|
Karimi S, Fouani MH, Moshaii A, Nikkhah M, Hosseinkhani S, Sheikhnejad R. Development of Dual Functional Nucleic Acid Delivery Nanosystem for DNA Induced Silencing of Bcl-2 Oncogene. Int J Nanomedicine 2020; 15:1693-1708. [PMID: 32210560 PMCID: PMC7073599 DOI: 10.2147/ijn.s236217] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/27/2020] [Indexed: 12/30/2022] Open
Abstract
Introduction Cancer treatment using functionalized vehicles in order to block involved genes has attracted a remarkable interest. In this study, we investigated the cellular uptake and cytotoxic effects of three sizes of anti Bcl-2 DNAi-conjugated gold nanoparticles by MCF-7 cells. Methods Three different sizes of gold nanoparticles were synthesized by citrate reduction method and after characterization, the nanoparticles were functionalized by Bcl-2 targeted DNAi. Cell internalization of the nanoparticles was analyzed by atomic absorption spectroscopy and light microscopy. The cytotoxic effects of the nanoparticles were investigated by MTT assay, flow cytometry and RT-PCR of the target gene. Results While poor cell internalization of bare gold nanoparticles was observed, the results demonstrated that cellular uptake of DNAi-conjugated gold nanoparticles is completely size-dependent, and the largest nanoparticle (~42 nm) revealed the highest internalization rate compared to other sizes (~14 and ~26 nm). Experimental findings showed that the DNAi-conjugated gold nanoparticles induced apoptotic pathway by silencing of the targeted Bcl-2 gene. In addition, supplementary theoretical studies demonstrated that the 42 nm DNAi-conjugated gold nanoparticles have great photothermal conversion efficiency for treatment under external illumination and these nanoparticles can be induced further cytotoxic effect by approximately 10°C temperature elevations. Conclusion Remarkable photothermal properties of DNAi-conjugated 42 nm Au-NPs in parallel with their high cell internalization and cytotoxic effects introduce them as potential dual functional anticancer nanosystems.
Collapse
Affiliation(s)
- Somayeh Karimi
- Department of Physics, Tarbiat Modares University, Tehran, Iran
| | | | - Ahmad Moshaii
- Department of Physics, Tarbiat Modares University, Tehran, Iran
| | - Maryam Nikkhah
- Department of Nanobiotechnology, Tarbiat Modares University, Tehran, Iran
| | | | - Reza Sheikhnejad
- Department of Molecular Biology, Tofigh Daru Engineering-Research Co., Tehran, Iran
| |
Collapse
|
83
|
Sarker SR, Takikawa M, Takeoka S. In Vitro Delivery of Cell Impermeable Phallotoxin Using Cationic Liposomes Composed of Lipids Bearing Lysine Headgroup. ACS APPLIED BIO MATERIALS 2020; 3:2048-2057. [DOI: 10.1021/acsabm.9b01167] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Satya Ranjan Sarker
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), Shinjuku-ku, Tokyo 162-8480, Japan
- Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - Masato Takikawa
- Department of Advanced Science and Engineering, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), Shinjuku-ku, Tokyo 162-8480, Japan
| | - Shinji Takeoka
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), Shinjuku-ku, Tokyo 162-8480, Japan
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda University, Shinjuku-ku, Tokyo 169-8555, Japan
| |
Collapse
|
84
|
Jiang Z, Thayumanavan S. Non-cationic Material Design for Nucleic Acid Delivery. ADVANCED THERAPEUTICS 2020; 3:1900206. [PMID: 34164572 PMCID: PMC8218910 DOI: 10.1002/adtp.201900206] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Indexed: 12/16/2022]
Abstract
Nucleic acid delivery provides effective options to control intracellular gene expression and protein production. Efficient delivery of nucleic acid typically requires delivery vehicles to facilitate the entry of nucleic acid into cells. Among non-viral delivery vehicles, cationic materials are favored because of their high loading capacity of nucleic acids and prominent cellular uptake efficiency through electrostatic interaction. However, cationic moieties at high dosage tend to induce severe cytotoxicity due to the interference on cell membrane integrity. In contrast, non-cationic materials present alternative delivery approaches with less safety concerns than cationic materials. In this Progress Report, principles of non-cationic material design for nucleic acid delivery are discussed. Examples of such non-cationic platforms are highlighted, including complexation or conjugation with nucleic acids and self-assembled nucleic acid structures.
Collapse
Affiliation(s)
- Ziwen Jiang
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| |
Collapse
|
85
|
Rafiee SD, Kocabey S, Mayer M, List J, Rüegg C. Detection of HER2 + Breast Cancer Cells using Bioinspired DNA-Based Signal Amplification. ChemMedChem 2020; 15:661-666. [PMID: 31943804 PMCID: PMC7187270 DOI: 10.1002/cmdc.201900697] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 12/17/2019] [Indexed: 01/25/2023]
Abstract
Circulating tumor cells (CTC) are promising biomarkers for metastatic cancer detection and monitoring progression. However, detection of CTCs remains challenging due to their low frequency and heterogeneity. Herein, we report a bioinspired approach to detect individual cancer cells, based on a signal amplification cascade using a programmable DNA hybridization chain reaction (HCR) circuit. We applied this approach to detect HER2+ cancer cells using the anti‐HER2 antibody (trastuzumab) coupled to initiator DNA eliciting a HCR cascade that leads to a fluorescent signal at the cell surface. At 4 °C, this HCR detection scheme resulted in highly efficient, specific and sensitive signal amplification of the DNA hairpins specifically on the membrane of the HER2+ cells in a background of HER2− cells and peripheral blood leukocytes, which remained almost non‐fluorescent. The results indicate that this system offers a new strategy that may be further developed toward an in vitro diagnostic platform for the sensitive and efficient detection of CTC.
Collapse
Affiliation(s)
- Sarah D Rafiee
- Department of Oncology, Microbiology and Immunology Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 18, PER17, 1700, Fribourg, Switzerland
| | - Samet Kocabey
- Department of Oncology, Microbiology and Immunology Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 18, PER17, 1700, Fribourg, Switzerland
| | - Michael Mayer
- BioPhysics, Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, PER 18, 1700, Fribourg, Switzerland
| | - Jonathan List
- BioPhysics, Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, PER 18, 1700, Fribourg, Switzerland.,Physics of Synthetic Biological Systems, Technical University Munich, Am Coulombwall 4a, 85748, Garching, Germany
| | - Curzio Rüegg
- Department of Oncology, Microbiology and Immunology Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 18, PER17, 1700, Fribourg, Switzerland
| |
Collapse
|
86
|
Graczyk A, Pawlowska R, Jedrzejczyk D, Chworos A. Gold Nanoparticles in Conjunction with Nucleic Acids as a Modern Molecular System for Cellular Delivery. Molecules 2020; 25:E204. [PMID: 31947834 PMCID: PMC6982881 DOI: 10.3390/molecules25010204] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/23/2019] [Accepted: 12/26/2019] [Indexed: 02/07/2023] Open
Abstract
Development of nanotechnology has become prominent in many fields, such as medicine, electronics, production of materials, and modern drugs. Nanomaterials and nanoparticles have gained recognition owing to the unique biochemical and physical properties. Considering cellular application, it is speculated that nanoparticles can transfer through cell membranes following different routes exclusively owing to their size (up to 100 nm) and surface functionalities. Nanoparticles have capacity to enter cells by themselves but also to carry other molecules through the lipid bilayer. This quality has been utilized in cellular delivery of substances like small chemical drugs or nucleic acids. Different nanoparticles including lipids, silica, and metal nanoparticles have been exploited in conjugation with nucleic acids. However, the noble metal nanoparticles create an alternative, out of which gold nanoparticles (AuNP) are the most common. The hybrids of DNA or RNA and metal nanoparticles can be employed for functional assemblies for variety of applications in medicine, diagnostics or nano-electronics by means of biomarkers, specific imaging probes, or gene expression regulatory function. In this review, we focus on the conjugates of gold nanoparticles and nucleic acids in the view of their potential application for cellular delivery and biomedicine. This review covers the current advances in the nanotechnology of DNA and RNA-AuNP conjugates and their potential applications. We emphasize the crucial role of metal nanoparticles in the nanotechnology of nucleic acids and explore the role of such conjugates in the biological systems. Finally, mechanisms guiding the process of cellular intake, essential for delivery of modern therapeutics, will be discussed.
Collapse
Affiliation(s)
| | | | | | - Arkadiusz Chworos
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Lodz, Poland; (A.G.); (R.P.); (D.J.)
| |
Collapse
|
87
|
Bhowmik D, Culver KSB, Liu T, Odom TW. Resolving Single-Nanoconstruct Dynamics during Targeting and Nontargeting Live-Cell Membrane Interactions. ACS NANO 2019; 13:13637-13644. [PMID: 31398007 PMCID: PMC7830831 DOI: 10.1021/acsnano.9b03144] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
This paper describes how differences in the dynamics of targeting and nontargeting constructs can provide information on nanoparticle (NP)-cell interactions. We probed translational and rotational dynamics of functionalized Au nanostar (AuNS) nanoconstructs interacting with cells in serum-containing medium. We found that AuNS with targeting ligands had a larger dynamical footprint and faster rotational speed on cell membranes expressing human epidermal growth factor receptor 2 (HER-2) receptors compared to that of AuNS with nontargeting ligands. Targeting and nontargeting nanoconstructs displayed distinct membrane dynamics despite their similar protein adsorption profiles, which suggests that targeted interactions are preserved even in the presence of a protein corona. The high sensitivity of single-NP dynamics can be used to compare different nanoconstruct properties (such as NP size, shape, and surface chemistry) to improve their design as delivery vehicles.
Collapse
Affiliation(s)
- Debanjan Bhowmik
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Kayla S. B. Culver
- Department of Materials Science & Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Tingting Liu
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Teri W. Odom
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Department of Materials Science & Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Corresponding Author: Phone: +1 (847) 491-7674.
| |
Collapse
|
88
|
Jiang Z, He H, Liu H, Thayumanavan S. Cellular Uptake Evaluation of Amphiphilic Polymer Assemblies: Importance of Interplay between Pharmacological and Genetic Approaches. Biomacromolecules 2019; 20:4407-4418. [PMID: 31609589 PMCID: PMC6901731 DOI: 10.1021/acs.biomac.9b01073] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Understanding the cellular uptake mechanism of materials is of fundamental importance that would be beneficial for materials design with enhanced biological functions. Herein, we report the interplay of pharmacological and genetic approaches to minimize the possible misinterpretation on cellular uptake mechanism. A library of amphiphilic polymers was used as a model system to evaluate the reliability of such methodological interplay. To probe the cellular uptake of amphiphilic polymers, we utilized an orthogonal end-group labeling strategy to conjugate one fluorescent molecule on each polymer chain. The results from the methodological interplay with these labeled polymers revealed the off-target effects of dynasore, a well-known dynamin inhibitor. Instead of dynamin, actin was found to be an essential cellular component during the cellular uptake of these amphiphilic polymers. Our study demonstrates the importance of interplaying pharmacological and genetic approaches when evaluating the endocytic mechanism of functional materials, providing insights on understanding the cellular uptake of future therapeutic materials.
Collapse
|
89
|
Applications of Spherical Nucleic Acid Nanoparticles as Delivery Systems. Trends Mol Med 2019; 25:1066-1079. [DOI: 10.1016/j.molmed.2019.08.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 08/26/2019] [Accepted: 08/28/2019] [Indexed: 12/13/2022]
|
90
|
Kobos L, Shannahan J. Biocorona‐induced modifications in engineered nanomaterial–cellular interactions impacting biomedical applications. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 12:e1608. [PMID: 31788989 DOI: 10.1002/wnan.1608] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/18/2019] [Accepted: 09/29/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Lisa Kobos
- School of Health Sciences College of Human and Health Sciences, Purdue University West Lafayette Indiana
| | - Jonathan Shannahan
- School of Health Sciences College of Human and Health Sciences, Purdue University West Lafayette Indiana
| |
Collapse
|
91
|
Shreffler JW, Pullan JE, Dailey KM, Mallik S, Brooks AE. Overcoming Hurdles in Nanoparticle Clinical Translation: The Influence of Experimental Design and Surface Modification. Int J Mol Sci 2019; 20:E6056. [PMID: 31801303 PMCID: PMC6928924 DOI: 10.3390/ijms20236056] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/11/2019] [Accepted: 11/23/2019] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles are becoming an increasingly popular tool for biomedical imaging and drug delivery. While the prevalence of nanoparticle drug-delivery systems reported in the literature increases yearly, relatively little translation from the bench to the bedside has occurred. It is crucial for the scientific community to recognize this shortcoming and re-evaluate standard practices in the field, to increase clinical translatability. Currently, nanoparticle drug-delivery systems are designed to increase circulation, target disease states, enhance retention in diseased tissues, and provide targeted payload release. To manage these demands, the surface of the particle is often modified with a variety of chemical and biological moieties, including PEG, tumor targeting peptides, and environmentally responsive linkers. Regardless of the surface modifications, the nano-bio interface, which is mediated by opsonization and the protein corona, often remains problematic. While fabrication and assessment techniques for nanoparticles have seen continued advances, a thorough evaluation of the particle's interaction with the immune system has lagged behind, seemingly taking a backseat to particle characterization. This review explores current limitations in the evaluation of surface-modified nanoparticle biocompatibility and in vivo model selection, suggesting a promising standardized pathway to clinical translation.
Collapse
Affiliation(s)
| | | | | | | | - Amanda E. Brooks
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA; (J.W.S.); (J.E.P.); (K.M.D.); (S.M.)
| |
Collapse
|
92
|
Sikorska K, Grądzka I, Sochanowicz B, Presz A, Męczyńska-Wielgosz S, Brzóska K, Kruszewski MK. Diminished amyloid-β uptake by mouse microglia upon treatment with quantum dots, silver or cerium oxide nanoparticles: Nanoparticles and amyloid-β uptake by microglia. Hum Exp Toxicol 2019; 39:147-158. [DOI: 10.1177/0960327119880586] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Alzheimer’s disease (AD) is a chronic neurodegenerative disease leading to progressive dementia in elderly people. The disease is characterized, among others, by formation of amyloid- β (A β) polypeptide plaques in the brain. Although etiology of the disease is not fully understood, recent research suggest that nanomaterials may affect AD development. Here, we described the consequences of exposure of mouse BV-2 microglia to silver nanoparticles (AgNPs, 50 µg/mL), cerium oxide nanoparticles (CeO2NPs, 100 µg/mL), and cadmium telluride quantum dots (CdTeQDs, 3 or 10 µg/mL) in the context of its ability to clear A β plaques. The brain microglial cells play an important role in removing A β plaques from the brain. Cell viability and cycle progression were assessed by trypan blue test and propidium iodide binding, respectively. The uptake of A β and NPs was measured by flow cytometry. Secretion of proinflammatory cytokines was measured with the use of cytometric bead array. A β (0.1 μM) did not affect viability, whereas NPs decreased microglia growth by arresting the cells in G1 phase (CdTeQDs) or in S phase (AgNPs and CeO2NPs) of cell cycle. The uptake of A β was significantly reduced in the presence of AgNPs and CeO2NPs. In addition, the least toxic CeO2NPs induced the release of proinflammatory cytokine, tumor necrosis factor α. In summary, each of the NPs tested affected either the microglia phagocytic activity (AgNPs and CeO2NPs) and/or its viability (AgNPs and CdTeQDs) that may favor the occurrence of AD and accelerate its development.
Collapse
Affiliation(s)
- K Sikorska
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - I Grądzka
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - B Sochanowicz
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - A Presz
- Laboratory of Nanostructures, Institute of High Pressure Physics, Warsaw, Poland
| | - S Męczyńska-Wielgosz
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - K Brzóska
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - MK Kruszewski
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland
- Department of Molecular Biology and Translational Research, Institute of Rural Health, Lublin, Poland
| |
Collapse
|
93
|
Gigante A, Li M, Junghänel S, Hirschhäuser C, Knauer S, Schmuck C. Non-viral transfection vectors: are hybrid materials the way forward? MEDCHEMCOMM 2019; 10:1692-1718. [PMID: 32180915 PMCID: PMC7053704 DOI: 10.1039/c9md00275h] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/12/2019] [Indexed: 12/18/2022]
Abstract
Transfection is a process by which oligonucleotides (DNA or RNA) are delivered into living cells. This allows the synthesis of target proteins as well as their inhibition (gene silencing). However, oligonucleotides cannot cross the plasma membrane by themselves; therefore, efficient carriers are needed for successful gene delivery. Recombinant viruses are among the earliest described vectors. Unfortunately, they have severe drawbacks such as toxicity and immunogenicity. In this regard, the development of non-viral transfection vectors has attracted increasing interests, and has become an important field of research. In the first part of this review we start with a tutorial introduction into the biological backgrounds of gene transfection followed by the classical non-viral vectors (cationic organic carriers and inorganic nanoparticles). In the second part we highlight selected recent reports, which demonstrate that hybrid vectors that combine key features of classical carriers are a remarkable strategy to address the current challenges in gene delivery.
Collapse
Affiliation(s)
- A Gigante
- Institute of Organic Chemistry , University of Duisburg-Essen , 45141 Essen , Germany .
| | - M Li
- Institute of Organic Chemistry , University of Duisburg-Essen , 45141 Essen , Germany .
| | - S Junghänel
- Institute of Organic Chemistry , University of Duisburg-Essen , 45141 Essen , Germany .
- Biomedical Technology Center of the Medical Faculty , University of Muenster , Muenster , Germany
| | - C Hirschhäuser
- Institute of Organic Chemistry , University of Duisburg-Essen , 45141 Essen , Germany .
| | - S Knauer
- Faculty of Biology , University of Duisburg-Essen , 45141 Essen , Germany
| | - C Schmuck
- Institute of Organic Chemistry , University of Duisburg-Essen , 45141 Essen , Germany .
| |
Collapse
|
94
|
Guan CM, Chinen AB, Ferrer JR, Ko CH, Mirkin CA. Impact of Sequence Specificity of Spherical Nucleic Acids on Macrophage Activation in Vitro and in Vivo. Mol Pharm 2019; 16:4223-4229. [PMID: 31536368 DOI: 10.1021/acs.molpharmaceut.9b00561] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The effects of spherical nucleic acid (SNA) gold nanoparticle conjugates on the activation of macrophages in vitro and release of cytokines in vivo were explored. Herein, we show that G-quadruplexes, the formation of which is enhanced on gold nanoparticle surfaces, elicit an increase in cytokine release from mouse and human macrophages and induce the upregulation of activation receptors as well as NO2 production in vitro. Moreover, these G-rich SNAs can induce cytokine release when injected intravenously, though there were no severe, long-term effects observed. These results further reinforce the notion that nucleic acid sequence and structure play an important role in how SNAs interact in biological milieu and highlight a key design parameter.
Collapse
Affiliation(s)
- Chenxia M Guan
- International Institute for Nanotechnology , Evanston , Illinois 60208 , United States
| | - Alyssa B Chinen
- International Institute for Nanotechnology , Evanston , Illinois 60208 , United States
| | - Jennifer R Ferrer
- Department of Surgery , Northwestern Feinberg School of Medicine , Chicago , Illinois 60611 , United States.,International Institute for Nanotechnology , Evanston , Illinois 60208 , United States
| | - Caroline H Ko
- International Institute for Nanotechnology , Evanston , Illinois 60208 , United States
| | - Chad A Mirkin
- International Institute for Nanotechnology , Evanston , Illinois 60208 , United States
| |
Collapse
|
95
|
Wang H, Dardir K, Lee KB, Fabris L. Impact of Protein Corona in Nanoflare-Based Biomolecular Detection and Quantification. Bioconjug Chem 2019; 30:2555-2562. [PMID: 31479244 DOI: 10.1021/acs.bioconjchem.9b00495] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Selective detection and precise quantification of biomolecules in intracellular settings play a pivotal role in the diagnostics and therapeutics of diseases, including various cancers and infectious epidemics. Because of this clinical relevance, nanoprobes with high sensitivity, wide tunability, and excellent biological stability have become of high demand. In particular, nanoflares based on gold nanoparticles have emerged as an attractive candidate for intracellular detection due to their efficient cellular uptake, enhanced binding affinity with complementary targets, and improved biological compatibility. However, nanoprobes, including these nanoflares, are known to be susceptible to the adsorption of proteins present in the biological environment, which leads to the formation of a so-called protein corona layer on their surface, leading to an altered targeting efficiency and cellular uptake. In this work, we leverage the nanoflares platform to demonstrate the effect of protein corona on biomolecular detection, quantification, as well as biological stability against enzymatic degradation. Nanoflares incubated in a biologically relevant concentration of serum albumin proteins (0.50 wt %) were shown to result in more than 20% signal reduction in target detection, with a decrease varying proportionally with the protein concentrations. In addition, similar signal reduction was observed for different serum proteins, and PEG backfilling was found to be ineffective in mitigating the negative impact induced by the corona formation. Furthermore, nuclease resistance in nanoflares was also severely compromised by the presence of the corona shell (∼2-fold increase in hydrolysis activity). This work demonstrates the consequences of an in situ formed protein corona layer on molecular detection/quantification and biological stability of nanoflares in the presence of nuclease enzymes, highlighting the importance of calibrating similar nanoprobes in proper biological media to improve the accuracy of molecular detection and quantification.
Collapse
Affiliation(s)
- Hao Wang
- Department of Materials Science and Engineering , Rutgers University , 607 Taylor Road , Piscataway , New Jersey 08854 , United States
| | - Kholud Dardir
- Department of Materials Science and Engineering , Rutgers University , 607 Taylor Road , Piscataway , New Jersey 08854 , United States
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology , Rutgers University , 123 Bevier Road , Piscataway , New Jersey 08854 , United States.,Department of Life and Nanopharmaceutical Science, College of Pharmacy , Kyung Hee University , Seoul 02447 , Republic of Korea
| | - Laura Fabris
- Department of Materials Science and Engineering , Rutgers University , 607 Taylor Road , Piscataway , New Jersey 08854 , United States
| |
Collapse
|
96
|
Towards Self-Transfecting Nucleic Acid Nanostructures for Gene Regulation. Trends Biotechnol 2019; 37:983-994. [DOI: 10.1016/j.tibtech.2019.01.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 01/06/2023]
|
97
|
Hu X, Ke G, Liu L, Fu X, Kong G, Xiong M, Chen M, Zhang XB. Valency-Controlled Molecular Spherical Nucleic Acids with Tunable Biosensing Performances. Anal Chem 2019; 91:11374-11379. [PMID: 31402646 DOI: 10.1021/acs.analchem.9b02614] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Spherical nucleic acids (SNAs) play critical roles in many fields, such as molecular diagnostics, disease therapeutics, and materials application. Due to the important role of DNA density on the properties of SNAs, the controlled synthesis of monodisperse SNAs with precise DNA density is an important approach for the structure-function relationship study and finite functions regulation of SNAs. In particular, the construction of monodisperse SNAs in a valency-tunable and site-specific manner is highly important; however, it is still challenging. Herein, on the basis of the high controllability, nanometer precision, and addressable modification ability of framework nucleic acid (FNA), we develop the concept of valency-controlled framework nucleic acid core-based molecular spherical nucleic acids (FNA-mSNAs) with tunable biosensing performances. The FNA-mSNAs consist of a valency-tunable FNA-based DNA nanocube as the core and a controlled, precise number of DNA strands per core. By simply alternating the binding site number for shell DNA strands on the DNA nanocube, homogeneous FNA-mSNAs with different valencies were easily designed, which enabled the molecular level study of the effect of valency on their properties, such as nuclease stability and cellular uptake. Furthermore, taking advantage of the addressable modification ability of FNA, the first heterogeneous molecular SNAs with tunable valency were demonstrated. Importantly, the valency of heterogeneous FNA-mSNAs was able to tune their biosensing performance, such as response dynamics, detection sensitivity, and response range. With these remarkable features, FNA-mSNAs provide new research methods for the development of functional SNAs at the molecular level for a wide range of biological applications.
Collapse
Affiliation(s)
- Xue Hu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering , Hunan University , Changsha , Hunan 410082 , China
| | - Guoliang Ke
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering , Hunan University , Changsha , Hunan 410082 , China
| | - Lu Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering , Hunan University , Changsha , Hunan 410082 , China
| | - Xiaoyi Fu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering , Hunan University , Changsha , Hunan 410082 , China
| | - Gezhi Kong
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering , Hunan University , Changsha , Hunan 410082 , China
| | - Mengyi Xiong
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering , Hunan University , Changsha , Hunan 410082 , China
| | - Mei Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering , Hunan University , Changsha , Hunan 410082 , China
| | - Xiao-Bing Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering , Hunan University , Changsha , Hunan 410082 , China
| |
Collapse
|
98
|
Ruiz G, Ryan N, Rutschke K, Awotunde O, Driskell JD. Antibodies Irreversibly Adsorb to Gold Nanoparticles and Resist Displacement by Common Blood Proteins. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:10601-10609. [PMID: 31335148 DOI: 10.1021/acs.langmuir.9b01900] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Gold nanoparticles (AuNPs) functionalized with proteins to impart desirable surface properties have been developed for many nanobiotechnology applications. A strong interaction between the protein and nanoparticle is critical to the formation of a stable conjugate to realize the potential of these emerging technologies. In this work, we examine the robustness of a protein layer adsorbed onto gold nanoparticles while under the stress of a physiological environment that could potentially lead to protein exchange on the nanoparticle surface. The adsorption interaction of common blood plasma proteins (transferrin, human serum albumin, and fibrinogen) and anti-horseradish peroxidase antibody onto AuNPs is investigated by nanoparticle tracking analysis. Our data show that a monolayer of protein is formed at saturation for each protein, and the maximum size increase for the conjugate, relative to the AuNP core, correlates with the protein size. The binding affinity of each protein to the AuNP is extracted from a best fit of the adsorption isotherm to the Hill equation. The antibody displays the greatest affinity (Kd = 15.2 ± 0.8 nM) that is ∼20-65 times stronger than the affinity of the other plasma proteins. Antibody-AuNP conjugates were prepared, purified, and suspended in solutions of blood plasma proteins to evaluate the stability of the antibody layer. An enzyme-mediated assay confirms that the antibody-AuNP interaction is irreversible, and the adsorbed antibody resists displacement by the plasma proteins. This work provides insight into the capabilities and potential limitations of antibody-AuNP-enabled technologies in biological systems.
Collapse
Affiliation(s)
- Guadalupe Ruiz
- Department of Chemistry , Illinois State University , Normal , Illinois 61790 , United States
| | - Nicki Ryan
- Department of Chemistry , Illinois State University , Normal , Illinois 61790 , United States
| | - Kylie Rutschke
- Department of Chemistry , Illinois State University , Normal , Illinois 61790 , United States
| | - Olatunde Awotunde
- Department of Chemistry , Illinois State University , Normal , Illinois 61790 , United States
| | - Jeremy D Driskell
- Department of Chemistry , Illinois State University , Normal , Illinois 61790 , United States
| |
Collapse
|
99
|
Sakhtianchi R, Darvishi B, Mirzaie Z, Dorkoosh F, Shanehsazzadeh S, Dinarvand R. Pegylated magnetic mesoporous silica nanoparticles decorated with AS1411 Aptamer as a targeting delivery system for cytotoxic agents. Pharm Dev Technol 2019; 24:1063-1075. [PMID: 30654677 DOI: 10.1080/10837450.2019.1569678] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Fulfilling the purpose of developing a NP with theragnostic capabilities, the current study describes the synthesis of an aptamer-functionalized PEG-coated SPION/mesoporous silica core-shell nanoparticle for concurrent cancer targeted therapy and magnetic resonance imaging. SPIONs were synthesized according to a thermal decomposition method and served as cores for SPION/mesoporous silica core/shell nanoparticles (MMSNs). Doxorubicin was then successfully loaded in MMSNs which were then coated with di-carboxylic acid functionalized polyethylene glycol (PEG-MMSNs). AS1411 aptamers were at the end covalently attached to NPs (APT-PEG-MMSNs). The mean diameter of synthesized NPs was about 89 nm and doxorubicin encapsulation efficacy was ≈67.47%. Results of MTT based cell cytotoxicity assay demonstrated a significantly higher toxicity profile for APT-PEG-MMSNs against MCF7 cells compared to non-decorated MMSNs, while no significant differences were spotted against NIH-3T3 cells. Meanwhile, formation of protein corona around APT-PEG-MMSNs in biological medium significantly attenuated observed cytotoxicity against MCF7 cell line. Examining NPs uptake by MCF7 cells using confocal laser scanning microscopy also confirmed superiority of APT-PEG-MMSNs over PEG-MMSNs. Finally, APT decorated NPs induced highest signal intensity reduction in T2-weighted images during in vitro MRI assay. In conclusion, developed NPs may serve as promising multifunctional vehicles for simultaneous cancer targeted therapy and MRI imaging.
Collapse
Affiliation(s)
- Ramin Sakhtianchi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences , Tehran , Iran.,Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences , Tehran , Iran
| | - Behrad Darvishi
- Recombinant Proteins Department, Breast Cancer Research Center, ACECR , Tehran , Iran
| | - Zahra Mirzaie
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences , Tehran , Iran
| | - Farid Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences , Tehran , Iran
| | - Saeed Shanehsazzadeh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences , Tehran , Iran
| | - Rassoul Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences , Tehran , Iran.,Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
100
|
Wang H, Chen B, He M, Li X, Chen P, Hu B. Study on uptake of gold nanoparticles by single cells using droplet microfluidic chip-inductively coupled plasma mass spectrometry. Talanta 2019; 200:398-407. [DOI: 10.1016/j.talanta.2019.03.075] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/19/2019] [Accepted: 03/19/2019] [Indexed: 11/29/2022]
|