351
|
Abstract
Nanoparticle-mediated gene and siRNA delivery has been an appealing area to gene therapists when they attempt to treat the diseases by manipulating the genetic information in the target cells. However, the advances in materials science could not keep up with the demand for multifunctional nanomaterials to achieve desired delivery efficiency. Researchers have thus taken an alternative approach to incorporate various materials into single composite nanoparticle using different fabrication methods. This approach allows nanoparticles to possess defined nanostructures as well as multiple functionalities to overcome the critical extracellular and intracellular barriers to successful gene delivery. This chapter will highlight the advances of fabrication methods that have the most potential to translate nanoparticles from bench to bedside. Furthermore, a major class of composite nanoparticle-lipid-based composite nanoparticles will be classified based on the components and reviewed in details.
Collapse
|
352
|
Abstract
We report a novel spherical nucleic acid (SNA) gold nanoparticle conjugate, termed the Sticky-flare, which enables facile quantification of RNA expression in live cells and spatiotemporal analysis of RNA transport and localization. The Sticky-flare is capable of entering live cells without the need for transfection agents and recognizing target RNA transcripts in a sequence-specific manner. On recognition, the Sticky-flare transfers a fluorophore-conjugated reporter to the transcript, resulting in a turning on of fluorescence in a quantifiable manner and the fluorescent labeling of targeted transcripts. The latter allows the RNA to be tracked via fluorescence microscopy as it is transported throughout the cell. We use this novel nanoconjugate to analyze the expression level and spatial distribution of β-actin mRNA in HeLa cells and to observe the real-time transport of β-actin mRNA in mouse embryonic fibroblasts. Furthermore, we investigate the application of Sticky-flares for tracking transcripts that undergo more extensive compartmentalization by fluorophore-labeling U1 small nuclear RNA and observing its distribution in the nucleus of live cells.
Collapse
|
353
|
Ming X, Laing B. Bioconjugates for targeted delivery of therapeutic oligonucleotides. Adv Drug Deliv Rev 2015; 87:81-9. [PMID: 25689735 DOI: 10.1016/j.addr.2015.02.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/04/2015] [Accepted: 02/06/2015] [Indexed: 01/05/2023]
Abstract
Bioconjugates have been used to deliver therapeutic oligonucleotides to their pharmacological targets in diseased cells. Molecular-scale conjugates can be prepared by directly linking targeting ligands with oligonucleotides and the resultant conjugates can selectively bind to cell surface receptors in target cells in diseased tissues. Besides targeted delivery, additional functionality can be incorporated in the conjugates by utilization of carrier molecules, and these larger conjugates are called carrier-associated conjugates. Both molecular and carrier-associated conjugates have achieved initial successes in clinical trials for treating liver diseases; therefore, currently the greater challenge is to deliver oligonucleotides to extrahepatic tissues such as tumors. This review will provide an update on the application of oligonucleotide conjugates for targeted delivery during the last decade. By identifying key elements for successful delivery, it is suggested that oligonucleotide conjugates with intermediate size, cell targeting ability, and endosomal release functionality are superior systems to advance oligonucleotides to achieve their full therapeutic potentials.
Collapse
Affiliation(s)
- Xin Ming
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Brian Laing
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
354
|
Reed AN, Putman T, Sullivan C, Jin L. Application of a nanoflare probe specific to a latency associated transcript for isolation of KHV latently infected cells. Virus Res 2015; 208:129-35. [PMID: 26087404 DOI: 10.1016/j.virusres.2015.06.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/29/2015] [Accepted: 06/01/2015] [Indexed: 11/18/2022]
Abstract
One of the unique features of herpesvirus infection is latent infection following an initial exposure, which is characterized by viral genome persistence in a small fraction of cells within the latently infected tissue. Investigation of the mechanisms of herpesvirus latency has been very challenging in tissues with only a small fraction of cells that are latently infected. Cyprinid herpesvirus 3, also known as koi herpesvirus (KHV), is an important and deadly pathogen of koi and common carp, Cyprinus carpio. Acute infection can cause up to 100% mortality in exposed fish, and fish that survive the infection become latently infected. KHV becomes latent in a small percentage of B lymphocytes and can reactivate under stressful conditions. During latency, KHV ORF6 transcript is expressed in the latently infected B lymphocytes. In order to study KHV latent infection in cells that are only latently infected, a nanoflare probe specific to ORF6 RNA was used to separate KHV latently infected cells from total peripheral white blood cells (WBC). Using the ORF6 nanoflare probe, less than 1% of peripheral WBC was isolated from KHV latently infected koi. When this enriched population of WBC was examined by real-time PCR specific for KHV, it was estimated that about 1-2 copies of viral genome persists in the sorted cells. In addition, KHV ORF6 transcript was shown to be the major transcript expressed during latency by RNA-seq analysis. This study demonstrated that an RNA nanoflare probe could be used to enrich latently infected cells, which can subsequently be used to investigate the molecular mechanisms of KHV latency.
Collapse
Affiliation(s)
- Aimee N Reed
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, United States; Department of Microbiology, College of Science, Oregon State University, Corvallis, OR 97331, United States
| | - Timothy Putman
- Department of Microbiology, College of Science, Oregon State University, Corvallis, OR 97331, United States
| | - Christopher Sullivan
- Center for Genome Research and Biocomputing, Oregon State University, Corvallis, OR 97331, United States
| | - Ling Jin
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, United States; Department of Microbiology, College of Science, Oregon State University, Corvallis, OR 97331, United States.
| |
Collapse
|
355
|
Choi CKK, Li J, Wei K, Xu YJ, Ho LWC, Zhu M, To KKW, Choi CHJ, Bian L. A gold@polydopamine core-shell nanoprobe for long-term intracellular detection of microRNAs in differentiating stem cells. J Am Chem Soc 2015; 137:7337-46. [PMID: 25996312 DOI: 10.1021/jacs.5b01457] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The capability of monitoring the differentiation process in living stem cells is crucial to the understanding of stem cell biology and the practical application of stem-cell-based therapies, yet conventional methods for the analysis of biomarkers related to differentiation require a large number of cells as well as cell lysis. Such requirements lead to the unavoidable loss of cell sources and preclude real-time monitoring of cellular events. In this work, we report the detection of microRNAs (miRNAs) in living human mesenchymal stem cells (hMSCs) by using polydopamine-coated gold nanoparticles (Au@PDA NPs). The PDA shell facilitates the immobilization of fluorescently labeled hairpin DNA strands (hpDNAs) that can recognize specific miRNA targets. The gold core and PDA shell quench the fluorescence of the immobilized hpDNAs, and subsequent binding of the hpDNAs to the target miRNAs leads to their dissociation from Au@PDA NPs and the recovery of fluorescence signals. Remarkably, these Au@PDA-hpDNA nanoprobes can naturally enter stem cells, which are known for their poor transfection efficiency, without the aid of transfection agents. Upon cellular uptake of these nanoprobes, we observe intense and time-dependent fluorescence responses from two important osteogenic marker miRNAs, namely, miR-29b and miR-31, only in hMSCs undergoing osteogenic differentiation and living primary osteoblasts but not in undifferentiated hMSCs and 3T3 fibroblasts. Strikingly, our nanoprobes can afford long-term tracking of miRNAs (5 days) in the differentiating hMSCs without the need of continuously replenishing cell culture medium with fresh nanoprobes. Our results demonstrate the capability of our Au@PDA-hpDNA nanoprobes for monitoring the differentiation status of hMSCs (i.e., differentiating versus undifferentiated) via the detection of specific miRNAs in living stem cells. Our nanoprobes show great promise in the investigation of the long-term dynamics of stem cell differentiation, identification and isolation of specific cell types, and high-throughput drug screening.
Collapse
Affiliation(s)
- Chun Kit K Choi
- †Department of Mechanical and Automation Engineering (Biomedical Engineering), ‡Department of Electronic Engineering (Biomedical Engineering), §School of Pharmacy, ∥Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Jinming Li
- †Department of Mechanical and Automation Engineering (Biomedical Engineering), ‡Department of Electronic Engineering (Biomedical Engineering), §School of Pharmacy, ∥Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Kongchang Wei
- †Department of Mechanical and Automation Engineering (Biomedical Engineering), ‡Department of Electronic Engineering (Biomedical Engineering), §School of Pharmacy, ∥Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Yang J Xu
- †Department of Mechanical and Automation Engineering (Biomedical Engineering), ‡Department of Electronic Engineering (Biomedical Engineering), §School of Pharmacy, ∥Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Lok Wai C Ho
- †Department of Mechanical and Automation Engineering (Biomedical Engineering), ‡Department of Electronic Engineering (Biomedical Engineering), §School of Pharmacy, ∥Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Meiling Zhu
- †Department of Mechanical and Automation Engineering (Biomedical Engineering), ‡Department of Electronic Engineering (Biomedical Engineering), §School of Pharmacy, ∥Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Kenneth K W To
- †Department of Mechanical and Automation Engineering (Biomedical Engineering), ‡Department of Electronic Engineering (Biomedical Engineering), §School of Pharmacy, ∥Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Chung Hang J Choi
- †Department of Mechanical and Automation Engineering (Biomedical Engineering), ‡Department of Electronic Engineering (Biomedical Engineering), §School of Pharmacy, ∥Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Liming Bian
- †Department of Mechanical and Automation Engineering (Biomedical Engineering), ‡Department of Electronic Engineering (Biomedical Engineering), §School of Pharmacy, ∥Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| |
Collapse
|
356
|
Wu Z, Liu GQ, Yang XL, Jiang JH. Electrostatic nucleic acid nanoassembly enables hybridization chain reaction in living cells for ultrasensitive mRNA imaging. J Am Chem Soc 2015; 137:6829-36. [PMID: 25969953 DOI: 10.1021/jacs.5b01778] [Citation(s) in RCA: 264] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Efficient approaches for intracellular delivery of nucleic acid reagents to achieve sensitive detection and regulation of gene and protein expressions are essential for chemistry and biology. We develop a novel electrostatic DNA nanoassembly that, for the first time, realizes hybridization chain reaction (HCR), a target-initiated alternating hybridization reaction between two hairpin probes, for signal amplification in living cells. The DNA nanoassembly has a designed structure with a core gold nanoparticle, a cationic peptide interlayer, and an electrostatically assembled outer layer of fluorophore-labeled hairpin DNA probes. It is shown to have high efficiency for cellular delivery of DNA probes via a unique endocytosis-independent mechanism that confers a significant advantage of overcoming endosomal entrapment. Moreover, electrostatic assembly of DNA probes enables target-initialized release of the probes from the nanoassembly via HCR. This intracellular HCR offers efficient signal amplification and enables ultrasensitive fluorescence activation imaging of mRNA expression with a picomolar detection limit. The results imply that the developed nanoassembly may provide an invaluable platform in low-abundance biomarker discovery and regulation for cell biology and theranostics.
Collapse
Affiliation(s)
- Zhan Wu
- State Key Laboratory of Chemeo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Gao-Qin Liu
- State Key Laboratory of Chemeo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Xiao-Li Yang
- State Key Laboratory of Chemeo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| | - Jian-Hui Jiang
- State Key Laboratory of Chemeo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China
| |
Collapse
|
357
|
Peptide-mediated delivery: an overview of pathways for efficient internalization. Ther Deliv 2015; 5:1203-22. [PMID: 25491671 DOI: 10.4155/tde.14.72] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Poor cellular delivery and low bioavailability of novel potent therapeutic molecules continue to remain the bottleneck of modern cancer and gene therapy. Cell-penetrating peptides have provided immense opportunities for the intracellular delivery of bioactive cargos and have led to the first exciting successes in experimental therapy of muscular dystrophies. This review focuses on the mechanisms by which cell-penetrating peptides gain access to the cell interior and deliver cargos. Recent advances in augmenting delivery efficacy and facilitation of endosomal escape of cargo are presented, and the cell-penetrating peptide-mediated delivery of two of the most popular classes of cargo molecules, oligonucleotides and proteins, is analyzed. The arsenal of tools for oligonucleotide delivery has dramatically expanded in the last decade enabling harnessing of cell-surface receptors for targeted delivery.
Collapse
|
358
|
Tan X, Li BB, Lu X, Jia F, Santori C, Menon P, Li H, Zhang B, Zhao JJ, Zhang K. Light-triggered, self-immolative nucleic Acid-drug nanostructures. J Am Chem Soc 2015; 137:6112-5. [PMID: 25924099 DOI: 10.1021/jacs.5b00795] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The simultaneous intracellular delivery of multiple types of payloads, such as hydrophobic drugs and nucleic acids, typically requires complex carrier systems. Herein, we demonstrate a self-deliverable form of nucleic acid-drug nanostructure that is composed almost entirely of payload molecules. Upon light activation, the nanostructure sheds the nucleic acid shell, while the core, which consists of prodrug molecules, disintegrates via an irreversible self-immolative process, releasing free drug molecules and small molecule fragments. We demonstrate that the nanostructures exhibit enhanced stability against DNase I compared with free DNA, and that the model drug (camptothecin) released exhibits similar efficacy as free, unmodified drugs toward cancer cells.
Collapse
Affiliation(s)
- Xuyu Tan
- †Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Ben B Li
- §Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts 02215, United States.,∥Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Xueguang Lu
- †Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Fei Jia
- †Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Clarissa Santori
- †Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Priyanka Menon
- †Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Hui Li
- ‡Institute of Chemical Biology and Nanomedicine, Hunan University, Changsha 410081, China
| | - Bohan Zhang
- ‡Institute of Chemical Biology and Nanomedicine, Hunan University, Changsha 410081, China
| | - Jean J Zhao
- §Department of Cancer Biology, Dana Farber Cancer Institute, Boston, Massachusetts 02215, United States.,∥Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Ke Zhang
- †Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States.,‡Institute of Chemical Biology and Nanomedicine, Hunan University, Changsha 410081, China
| |
Collapse
|
359
|
siRNA-based spherical nucleic acids reverse impaired wound healing in diabetic mice by ganglioside GM3 synthase knockdown. Proc Natl Acad Sci U S A 2015; 112:5573-8. [PMID: 25902507 DOI: 10.1073/pnas.1505951112] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Spherical nucleic acid (SNA) gold nanoparticle conjugates (13-nm-diameter gold cores functionalized with densely packed and highly oriented nucleic acids) dispersed in Aquaphor have been shown to penetrate the epidermal barrier of both intact mouse and human skin, enter keratinocytes, and efficiently down-regulate gene targets. ganglioside-monosialic acid 3 synthase (GM3S) is a known target that is overexpressed in diabetic mice and responsible for causing insulin resistance and impeding wound healing. GM3S SNAs increase keratinocyte migration and proliferation as well as insulin and insulin-like growth factor-1 (IGF1) receptor activation under both normo- and hyperglycemic conditions. The topical application of GM3S SNAs (50 nM) to splinted 6-mm-diameter full-thickness wounds in diet-induced obese diabetic mice decreases local GM3S expression by >80% at the wound edge through an siRNA pathway and fully heals wounds clinically and histologically within 12 d, whereas control-treated wounds are only 50% closed. Granulation tissue area, vascularity, and IGF1 and EGF receptor phosphorylation are increased in GM3S SNA-treated wounds. These data capitalize on the unique ability of SNAs to naturally penetrate the skin and enter keratinocytes without the need for transfection agents. Moreover, the data further validate GM3 as a mediator of the delayed wound healing in type 2 diabetes and support regional GM3 depletion as a promising therapeutic direction.
Collapse
|
360
|
Hong CA, Eltoukhy AA, Lee H, Langer R, Anderson DG, Nam YS. Dendrimeric siRNA for Efficient Gene Silencing. Angew Chem Int Ed Engl 2015; 54:6740-4. [PMID: 25892329 DOI: 10.1002/anie.201412493] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 02/09/2015] [Indexed: 11/09/2022]
Abstract
Programmable molecular self-assembly of siRNA molecules provides precisely controlled generation of dendrimeric siRNA nanostructures. The second-generation dendrimers of siRNA can be effectively complexed with a low-molecular-weight, cationic polymer (poly(β-amino ester), PBAE) to generate stable nanostructures about 160 nm in diameter via strong electrostatic interactions. Condensation and gene silencing efficiencies increase with the increased generation of siRNA dendrimers due to a high charge density and structural flexibility.
Collapse
Affiliation(s)
- Cheol Am Hong
- Department of Biological Sciences, Department of Materials Science and Engineering, KI for NanoCentury (KINC CNiT), Korea Advanced Institute of Science and Technology, Daejeon 305-701 (Republic of Korea)
| | - Ahmed A Eltoukhy
- Department of Biological Engineering, The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 (USA)
| | - Hyukjin Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul (Republic of Korea)
| | - Robert Langer
- Department of Biological Engineering, The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 (USA)
| | - Daniel G Anderson
- Department of Biological Engineering, The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 (USA).
| | - Yoon Sung Nam
- Department of Biological Sciences, Department of Materials Science and Engineering, KI for NanoCentury (KINC CNiT), Korea Advanced Institute of Science and Technology, Daejeon 305-701 (Republic of Korea).
| |
Collapse
|
361
|
Hong CA, Eltoukhy AA, Lee H, Langer R, Anderson DG, Nam YS. Dendrimeric siRNA for Efficient Gene Silencing. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201412493] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
362
|
Smith JA, Leonardi T, Huang B, Iraci N, Vega B, Pluchino S. Extracellular vesicles and their synthetic analogues in aging and age-associated brain diseases. Biogerontology 2015; 16:147-85. [PMID: 24973266 PMCID: PMC4578234 DOI: 10.1007/s10522-014-9510-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 06/16/2014] [Indexed: 02/06/2023]
Abstract
Multicellular organisms rely upon diverse and complex intercellular communications networks for a myriad of physiological processes. Disruption of these processes is implicated in the onset and propagation of disease and disorder, including the mechanisms of senescence at both cellular and organismal levels. In recent years, secreted extracellular vesicles (EVs) have been identified as a particularly novel vector by which cell-to-cell communications are enacted. EVs actively and specifically traffic bioactive proteins, nucleic acids, and metabolites between cells at local and systemic levels, modulating cellular responses in a bidirectional manner under both homeostatic and pathological conditions. EVs are being implicated not only in the generic aging process, but also as vehicles of pathology in a number of age-related diseases, including cancer and neurodegenerative and disease. Thus, circulating EVs-or specific EV cargoes-are being utilised as putative biomarkers of disease. On the other hand, EVs, as targeted intercellular shuttles of multipotent bioactive payloads, have demonstrated promising therapeutic properties, which can potentially be modulated and enhanced through cellular engineering. Furthermore, there is considerable interest in employing nanomedicinal approaches to mimic the putative therapeutic properties of EVs by employing synthetic analogues for targeted drug delivery. Herein we describe what is known about the origin and nature of EVs and subsequently review their putative roles in biology and medicine (including the use of synthetic EV analogues), with a particular focus on their role in aging and age-related brain diseases.
Collapse
Affiliation(s)
- J A Smith
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, CB2 0PY, UK
| | | | | | | | | | | |
Collapse
|
363
|
Abstract
Immunomodulatory nucleic acids have extraordinary promise for treating disease, yet clinical progress has been limited by a lack of tools to safely increase activity in patients. Immunomodulatory nucleic acids act by agonizing or antagonizing endosomal toll-like receptors (TLR3, TLR7/8, and TLR9), proteins involved in innate immune signaling. Immunomodulatory spherical nucleic acids (SNAs) that stimulate (immunostimulatory, IS-SNA) or regulate (immunoregulatory, IR-SNA) immunity by engaging TLRs have been designed, synthesized, and characterized. Compared with free oligonucleotides, IS-SNAs exhibit up to 80-fold increases in potency, 700-fold higher antibody titers, 400-fold higher cellular responses to a model antigen, and improved treatment of mice with lymphomas. IR-SNAs exhibit up to eightfold increases in potency and 30% greater reduction in fibrosis score in mice with nonalcoholic steatohepatitis (NASH). Given the clinical potential of SNAs due to their potency, defined chemical nature, and good tolerability, SNAs are attractive new modalities for developing immunotherapies.
Collapse
|
364
|
Lindberg S, Regberg J, Eriksson J, Helmfors H, Muñoz-Alarcón A, Srimanee A, Figueroa RA, Hallberg E, Ezzat K, Langel Ü. A convergent uptake route for peptide- and polymer-based nucleotide delivery systems. J Control Release 2015; 206:58-66. [PMID: 25769688 DOI: 10.1016/j.jconrel.2015.03.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 02/16/2015] [Accepted: 03/09/2015] [Indexed: 11/19/2022]
Abstract
Cell-penetrating peptides (CPPs) have been used as vehicles to deliver various cargos into cells and are promising as tools to deliver therapeutic biomolecules such as oligonucleotides both in vitro and in vivo. CPPs are positively charged and it is believed that CPPs deliver their cargo in a receptor-independent manner by interacting with the negatively charged plasma membrane and thereby inducing endocytosis. In this study we examine the mechanism of uptake of several different, well known, CPPs that form complexes with oligonucleotides. We show that these CPP:oligonucleotide complexes are negatively charged in transfection-media and their uptake is mediated by class A scavenger receptors (SCARA). These receptors are known to promiscuously bind to, and mediate uptake of poly-anionic macromolecules. Uptake of CPP:oligonucleotide complexes was abolished using pharmacological SCARA inhibitors as well as siRNA-mediated knockdown of SCARA. Additionally, uptake of CPP:oligonucleotide was significantly increased by transiently overexpressing SCARA. Furthermore, SCARA inhibitors also blocked internalization of cationic polymer:oligonucleotide complexes. Our results demonstrate that the previous held belief that CPPs act receptor independently does not hold true for CPP:oligonucleotide complexes, as scavenger receptor class A (SCARA) mediates the uptake of all the examined CPP:oligonucleotide complexes in this study.
Collapse
Affiliation(s)
- Staffan Lindberg
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, 10691 Stockholm, Sweden.
| | - Jakob Regberg
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, 10691 Stockholm, Sweden
| | - Jonas Eriksson
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, 10691 Stockholm, Sweden
| | - Henrik Helmfors
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, 10691 Stockholm, Sweden
| | - Andrés Muñoz-Alarcón
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, 10691 Stockholm, Sweden
| | - Artita Srimanee
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, 10691 Stockholm, Sweden
| | - Ricardo A Figueroa
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, 10691 Stockholm, Sweden
| | - Einar Hallberg
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, 10691 Stockholm, Sweden
| | - Kariem Ezzat
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, 10691 Stockholm, Sweden
| | - Ülo Langel
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, 10691 Stockholm, Sweden; Tartu University, Institute of Technology, 504 11 Tartu, Estonia.
| |
Collapse
|
365
|
Adjei IM, Blanka S. Modulation of the tumor microenvironment for cancer treatment: a biomaterials approach. J Funct Biomater 2015; 6:81-103. [PMID: 25695337 PMCID: PMC4384103 DOI: 10.3390/jfb6010081] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 10/07/2014] [Accepted: 02/12/2015] [Indexed: 12/26/2022] Open
Abstract
Tumors are complex tissues that consist of stromal cells, such as fibroblasts, immune cells and mesenchymal stem cells, as well as non-cellular components, in addition to neoplastic cells. Increasingly, there is evidence to suggest that these non-neoplastic cell components support cancer initiation, progression and metastasis and that their ablation or reprogramming can inhibit tumor growth. Our understanding of the activities of different parts of the tumor stroma in advancing cancer has been improved by the use of scaffold and matrix-based 3D systems originally developed for regenerative medicine. Additionally, drug delivery systems made from synthetic and natural biomaterials deliver drugs to kill stromal cells or reprogram the microenvironment for tumor inhibition. In this article, we review the impact of 3D tumor models in increasing our understanding of tumorigenesis. We also discuss how different drug delivery systems aid in the reprogramming of tumor stroma for cancer treatment.
Collapse
Affiliation(s)
- Isaac M Adjei
- Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| | - Sharma Blanka
- Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
366
|
Calabrese CM, Merkel TJ, Briley WE, Randeria PS, Narayan SP, Rouge JL, Walker DA, Scott AW, Mirkin CA. Biocompatible infinite-coordination-polymer nanoparticle-nucleic-acid conjugates for antisense gene regulation. Angew Chem Int Ed Engl 2015; 54:476-480. [PMID: 25393766 PMCID: PMC4314394 DOI: 10.1002/anie.201407946] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/20/2014] [Indexed: 01/15/2023]
Abstract
Herein, we report the synthesis of DNA-functionalized infinite-coordination-polymer (ICP) nanoparticles as biocompatible gene-regulation agents. ICP nanoparticles were synthesized from ferric nitrate and a ditopic 3-hydroxy-4-pyridinone (HOPO) ligand bearing a pendant azide. Addition of Fe(III) to a solution of the ligand produced nanoparticles, which were colloidally unstable in the presence of salts. Conjugation of DNA to the Fe(III)-HOPO ICP particles by copper-free click chemistry afforded colloidally stable nucleic-acid nanoconstructs. The DNA-ICP particles, when cross-linked through sequence-specific hybridization, exhibited narrow, highly cooperative melting transitions consistent with dense DNA surface loading. The ability of the DNA-ICP particles to enter cells and alter protein expression was also evaluated. Our results indicate that these novel particles carry nucleic acids into mammalian cells without the need for transfection agents and are capable of efficient gene knockdown.
Collapse
Affiliation(s)
| | - Timothy J. Merkel
- International Institute for Nanotechnology Northwestern University, Evanston, IL (USA)
| | - William E. Briley
- Interdepartmental Biological Sciences Northwestern University, Evanston, IL (USA)
| | - Pratik S. Randeria
- Department of Biomedical Engineering Northwestern University, Evanston, IL (USA)
| | - Suguna P. Narayan
- Department of Biomedical Engineering Northwestern University, Evanston, IL (USA)
| | - Jessica L. Rouge
- International Institute for Nanotechnology Northwestern University, Evanston, IL (USA)
| | - David A. Walker
- International Institute for Nanotechnology Northwestern University, Evanston, IL (USA)
- Department of Chemical and Biological Engineering Northwestern University, Evanston, IL (USA)
| | - Alexander W. Scott
- Department of Biomedical Engineering Northwestern University, Evanston, IL (USA)
| | - Chad A. Mirkin
- Department of Chemistry Northwestern University, Evanston, IL (USA)
- International Institute for Nanotechnology Northwestern University, Evanston, IL (USA)
- Department of Chemical and Biological Engineering Northwestern University, Evanston, IL (USA)
| |
Collapse
|
367
|
Goda T, Ishihara K, Miyahara Y. Critical update on 2-methacryloyloxyethyl phosphorylcholine (MPC) polymer science. J Appl Polym Sci 2015. [DOI: 10.1002/app.41766] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Tatsuro Goda
- Institute of Biomaterials and Bioengineering; Tokyo Medical and Dental University; 2-3-10 Kanda-Surugadai Chiyoda Tokyo 101-0062 Japan
| | - Kazuhiko Ishihara
- Department of Materials Engineering; The University of Tokyo; 7-3-1 Hongo Bunkyo Tokyo 113-8656 Japan
- Department of Bioengineering; The University of Tokyo; 7-3-1 Hongo Bunkyo Tokyo 113-8656 Japan
| | - Yuji Miyahara
- Institute of Biomaterials and Bioengineering; Tokyo Medical and Dental University; 2-3-10 Kanda-Surugadai Chiyoda Tokyo 101-0062 Japan
| |
Collapse
|
368
|
Randeria PS, Briley WE, Chinen AB, Guan CM, Petrosko SH, Mirkin CA. Nanoflares as probes for cancer diagnostics. Cancer Treat Res 2015; 166:1-22. [PMID: 25895862 DOI: 10.1007/978-3-319-16555-4_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Patients whose cancer is detected early are much more likely to have a positive prognosis and outcome. Nanoflares hold promise as a practical diagnostic platform for the early detection of cancer markers in living cells. These probes are based on spherical nucleic acid (SNAs) and are typically composed of gold nanoparticle cores and densely packed and highly oriented oligonucleotide shells; these sequences are complementary to specific mRNA targets and are hybridized to fluorophore-labeled reporter strands. Nanoflares take advantage of the highly efficient fluorescence quenching properties of gold, the rapid cellular uptake of SNAs that occurs without the use of transfection agents, and the enzymatic stability of such constructs to report a highly sensitive and specific signal in the presence of intracellular target mRNA. In this chapter, we will focus on the synthesis, characterization, and diagnostic applications of nanoflares as they relate to cancer markers.
Collapse
Affiliation(s)
- Pratik S Randeria
- Department of Biomedical Engineering, Northwestern University, Sheridan Road, 2145, Evanston, IL, 60208, USA
| | | | | | | | | | | |
Collapse
|
369
|
Pilapong C, Raiputta C, Chaisupa J, Sittichai S, Thongtem S, Thongtem T. Magnetic-EpCAM nanoprobe as a new platform for efficient targeting, isolating and imaging hepatocellular carcinoma. RSC Adv 2015. [DOI: 10.1039/c5ra01566a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Herein, magnetic-EpCAM nanoparticle (EpCAM-MNP) was developed and exploited as nanoprobe for targeting, isolating and imaging hepatocellular carcinoma.
Collapse
Affiliation(s)
- C. Pilapong
- Center of Excellence for Molecular Imaging (CEMI)
- Department of Radiologic Technology
- Faculty of Associated Medical Sciences
- Chiang Mai University
- Chiang Mai 50200
| | - C. Raiputta
- Center of Excellence for Molecular Imaging (CEMI)
- Department of Radiologic Technology
- Faculty of Associated Medical Sciences
- Chiang Mai University
- Chiang Mai 50200
| | - J. Chaisupa
- Center of Excellence for Molecular Imaging (CEMI)
- Department of Radiologic Technology
- Faculty of Associated Medical Sciences
- Chiang Mai University
- Chiang Mai 50200
| | - S. Sittichai
- Department of Physics and Material Science
- Faculty of Science
- Chiang Mai University
- Chiang Mai 50200
- Thailand
| | - S. Thongtem
- Department of Physics and Material Science
- Faculty of Science
- Chiang Mai University
- Chiang Mai 50200
- Thailand
| | - T. Thongtem
- Department of Chemistry
- Faculty of Science
- Chiang Mai University
- Chiang Mai 50200
- Thailand
| |
Collapse
|
370
|
Maddala SP, Mastroianni G, Velluto D, Sullivan AC. Intracellular delivery of BSA by phosphonate@silica nanoparticles. J Mater Chem B 2015; 3:6057-6070. [DOI: 10.1039/c5tb00555h] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Folate receptor mediated delivery of BSA to HeLa cells by a mesoporous phosphonate@silica nanoparticle carrier is described.
Collapse
Affiliation(s)
- Sai Prakash Maddala
- School of Biological and Chemical Science
- Queen Mary University of London
- London E14NS
- UK
| | - Giulia Mastroianni
- School of Biological and Chemical Science
- Queen Mary University of London
- London E14NS
- UK
| | - Diana Velluto
- School of Biological and Chemical Science
- Queen Mary University of London
- London E14NS
- UK
| | - Alice C. Sullivan
- School of Biological and Chemical Science
- Queen Mary University of London
- London E14NS
- UK
| |
Collapse
|
371
|
Barnaby SN, Sita TL, Petrosko SH, Stegh AH, Mirkin CA. Therapeutic applications of spherical nucleic acids. Cancer Treat Res 2015; 166:23-50. [PMID: 25895863 DOI: 10.1007/978-3-319-16555-4_2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Spherical nucleic acids (SNAs) represent an emerging class of nanoparticle-based therapeutics. SNAs consist of densely functionalized and highly oriented oligonucleotides on the surface of a nanoparticle which can either be inorganic (such as gold or platinum) or hollow (such as liposomal or silica-based). The spherical architecture of the oligonucleotide shell confers unique advantages over traditional nucleic acid delivery methods, including entry into nearly all cells independent of transfection agents and resistance to nuclease degradation. Furthermore, SNAs can penetrate biological barriers, including the blood-brain and blood-tumor barriers as well as the epidermis, and have demonstrated efficacy in several murine disease models in the absence of significant adverse side effects. In this chapter, we will focus on the applications of SNAs in cancer therapy as well as discuss multimodal SNAs for drug delivery and imaging.
Collapse
Affiliation(s)
- Stacey N Barnaby
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, IL, 60208, USA
| | | | | | | | | |
Collapse
|
372
|
Abstract
There are currently no nanoparticle formulations that optimally target diseased cells in the body. A small percentage of nanoparticles reach these cells and most accumulate in cells of the mononuclear phagocytic system. This chapter explores the interactions between nanoparticles and cells that may explain the causes for off-target accumulation of nanoparticles. A greater understanding of the nanoparticle-cellular interactions will lead to improvements in particle design for improved therapeutic outcome.
Collapse
Affiliation(s)
- Abdullah Syed
- Institute of Biomaterials and Biomedical Engineering, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 164 College St., 407, Toronto, ON, M5S 3G9, Canada
| | | |
Collapse
|
373
|
Abstract
Intracellular delivery of functional proteins using nanoparticles can be a game-changing approach for cancer therapy. However, cytosolic release of functional protein is still a major challenge. In addition, formation of protein corona on the surface of the nanoparticles can also alter the behavior of the nanoparticles. Here, we will review recent strategies for protein delivery into the cell. Finally we will discuss the issue of protein corona formation in light of nanoparticle-protein interactions.
Collapse
|
374
|
Wang Z, Zhang R, Wang Z, Wang HF, Wang Y, Zhao J, Wang F, Li W, Niu G, Kiesewetter DO, Chen X. Bioinspired nanocomplex for spatiotemporal imaging of sequential mRNA expression in differentiating neural stem cells. ACS NANO 2014; 8:12386-96. [PMID: 25494492 PMCID: PMC4278684 DOI: 10.1021/nn505047n] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 12/10/2014] [Indexed: 05/19/2023]
Abstract
Messenger RNA plays a pivotal role in regulating cellular activities. The expression dynamics of specific mRNA contains substantial information on the intracellular milieu. Unlike the imaging of stationary mRNAs, real-time intracellular imaging of the dynamics of mRNA expression is of great value for investigating mRNA biology and exploring specific cellular cascades. In addition to advanced imaging methods, timely extracellular stimulation is another key factor in regulating the mRNA expression repertoire. The integration of effective stimulation and imaging into a single robust system would significantly improve stimulation efficiency and imaging accuracy, producing fewer unwanted artifacts. In this study, we developed a multifunctional nanocomplex to enable self-activating and spatiotemporal imaging of the dynamics of mRNA sequential expression during the neural stem cell differentiation process. This nanocomplex showed improved enzymatic stability, fast recognition kinetics, and high specificity. With a mechanism regulated by endogenous cell machinery, this nanocomplex realized the successive stimulating motif release and the dynamic imaging of chronological mRNA expression during neural stem cell differentiation without the use of transgenetic manipulation. The dynamic imaging montage of mRNA expression ultimately facilitated genetic heterogeneity analysis. In vivo lateral ventricle injection of this nanocomplex enabled endogenous neural stem cell activation and labeling at their specific differentiation stages. This nanocomplex is highly amenable as an alternative tool to explore the dynamics of intricate mRNA activities in various physiological and pathological conditions.
Collapse
Affiliation(s)
- Zhe Wang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Ruili Zhang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi 710071, China
| | - Zhongliang Wang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi 710071, China
- Address correspondence to ,
| | - He-Fang Wang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Yu Wang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Jun Zhao
- Unit on Synapse Development and Plasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Fu Wang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi 710071, China
| | - Weitao Li
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Dale O. Kiesewetter
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
- Address correspondence to ,
| |
Collapse
|
375
|
Fleischer C, Payne CK. Secondary structure of corona proteins determines the cell surface receptors used by nanoparticles. J Phys Chem B 2014; 118:14017-26. [PMID: 24779411 PMCID: PMC4266332 DOI: 10.1021/jp502624n] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Revised: 04/27/2014] [Indexed: 01/04/2023]
Abstract
Nanoparticles used for biological and biomedical applications encounter a host of extracellular proteins. These proteins rapidly adsorb onto the nanoparticle surface, creating a protein corona. Poly(ethylene glycol) can reduce, but not eliminate, the nonspecific adsorption of proteins. As a result, the adsorbed proteins, rather than the nanoparticle itself, determine the cellular receptors used for binding, the internalization mechanism, the intracellular transport pathway, and the subsequent immune response. Using fluorescence microscopy and flow cytometry, we first characterize a set of polystyrene nanoparticles in which the same adsorbed protein, bovine serum albumin, leads to binding to two different cell surface receptors: native albumin receptors and scavenger receptors. Using a combination of circular dichroism spectroscopy, isothermal titration calorimetry, and fluorescence spectroscopy, we demonstrate that the secondary structure of the adsorbed bovine serum albumin protein controls the cellular receptors used by the protein-nanoparticle complexes. These results show that protein secondary structure is a key parameter in determining the cell surface receptor used by a protein-nanoparticle complex. We expect this link between protein structure and cellular outcomes will provide a molecular basis for the design of nanoparticles for use in biological and biomedical applications.
Collapse
Affiliation(s)
- Candace
C. Fleischer
- School of Chemistry and Biochemistry and Petit Institute
for Bioengineering
and Bioscience, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, Georgia 30332, United States
| | - Christine K. Payne
- School of Chemistry and Biochemistry and Petit Institute
for Bioengineering
and Bioscience, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, Georgia 30332, United States
| |
Collapse
|
376
|
Halo TL, McMahon KM, Angeloni NL, Xu Y, Wang W, Chinen AB, Malin D, Strekalova E, Cryns VL, Cheng C, Mirkin CA, Thaxton CS. NanoFlares for the detection, isolation, and culture of live tumor cells from human blood. Proc Natl Acad Sci U S A 2014; 111:17104-9. [PMID: 25404304 PMCID: PMC4260589 DOI: 10.1073/pnas.1418637111] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Metastasis portends a poor prognosis for cancer patients. Primary tumor cells disseminate through the bloodstream before the appearance of detectable metastatic lesions. The analysis of cancer cells in blood—so-called circulating tumor cells (CTCs)—may provide unprecedented opportunities for metastatic risk assessment and investigation. NanoFlares are nanoconstructs that enable live-cell detection of intracellular mRNA. NanoFlares, when coupled with flow cytometry, can be used to fluorescently detect genetic markers of CTCs in the context of whole blood. They allow one to detect as few as 100 live cancer cells per mL of blood and subsequently culture those cells. This technique can also be used to detect CTCs in a murine model of metastatic breast cancer. As such, NanoFlares provide, to our knowledge, the first genetic-based approach for detecting, isolating, and characterizing live cancer cells from blood and may provide new opportunities for cancer diagnosis, prognosis, and personalized therapy.
Collapse
MESH Headings
- Base Sequence
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/blood
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Cadherins/genetics
- Cadherins/metabolism
- Carbocyanines/chemistry
- Carbocyanines/metabolism
- Cell Line, Tumor
- DNA, Antisense/chemistry
- DNA, Antisense/genetics
- DNA, Antisense/metabolism
- Female
- Fibronectins/genetics
- Fibronectins/metabolism
- Gold/chemistry
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Humans
- Luminescent Proteins/genetics
- Luminescent Proteins/metabolism
- Metal Nanoparticles/chemistry
- Microscopy, Fluorescence
- Nanotechnology/methods
- Neoplastic Cells, Circulating/chemistry
- Neoplastic Cells, Circulating/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Transplantation, Heterologous
- Vimentin/genetics
- Vimentin/metabolism
- Red Fluorescent Protein
Collapse
Affiliation(s)
- Tiffany L Halo
- Department of Chemistry, International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208-3113
| | - Kaylin M McMahon
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611; Simpson Querrey Institute for BioNanotechnology in Medicine, Northwestern University, Chicago, IL, 60611; Walter S. and Lucienne Driskill Graduate Training Program in Life Sciences, Northwestern University, Chicago, IL 60611
| | - Nicholas L Angeloni
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611; Simpson Querrey Institute for BioNanotechnology in Medicine, Northwestern University, Chicago, IL, 60611; Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Yilin Xu
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611; Department of Medicine, Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Wei Wang
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611; Department of Medicine, Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Alyssa B Chinen
- Department of Chemistry, International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208-3113
| | - Dmitry Malin
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705; and
| | - Elena Strekalova
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705; and
| | - Vincent L Cryns
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705; and
| | - Chonghui Cheng
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611; Department of Medicine, Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Chad A Mirkin
- Department of Chemistry, International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208-3113; Department of Materials Science and Engineering, and
| | - C Shad Thaxton
- International Institute for Nanotechnology, Northwestern University, Evanston, IL 60208-3113; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611; Simpson Querrey Institute for BioNanotechnology in Medicine, Northwestern University, Chicago, IL, 60611; Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611;
| |
Collapse
|
377
|
Chinen AB, Guan CM, Mirkin CA. Spherical nucleic acid nanoparticle conjugates enhance G-quadruplex formation and increase serum protein interactions. Angew Chem Int Ed Engl 2014; 54:527-31. [PMID: 25393322 DOI: 10.1002/anie.201409211] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 10/13/2014] [Indexed: 01/15/2023]
Abstract
To understand the effect of three-dimensional oligonucleotide structure on protein corona formation, we studied the identity and quantity of human serum proteins that bind to spherical nucleic acid (SNA) nanoparticle conjugates. SNAs exhibit cellular uptake properties that are remarkably different from those of linear nucleic acids, which have been related to their interaction with certain classes of proteins. Through a proteomic analysis, this work shows that the protein binding properties of SNAs are sequence-specific and supports the conclusion that the oligonucleotide tertiary structure can significantly alter the chemical composition of the SNA protein corona. This knowledge will impact our understanding of how nucleic acid-based nanostructures, and SNAs in particular, function in complex biological milieu.
Collapse
Affiliation(s)
- Alyssa B Chinen
- Department of Chemistry, Northwestern University, Evanston, IL 60201 (USA)
| | | | | |
Collapse
|
378
|
Calabrese CM, Merkel TJ, Briley WE, Randeria PS, Narayan SP, Rouge JL, Walker DA, Scott AW, Mirkin CA. Biocompatible Infinite-Coordination-Polymer Nanoparticle-Nucleic-Acid Conjugates for Antisense Gene Regulation. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201407946] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
379
|
Chinen AB, Guan CM, Mirkin CA. Spherical Nucleic Acid Nanoparticle Conjugates Enhance G-Quadruplex Formation and Increase Serum Protein Interactions. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201409211] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
380
|
Kim J, Lee YM, Kang Y, Kim WJ. Tumor-homing, size-tunable clustered nanoparticles for anticancer therapeutics. ACS NANO 2014; 8:9358-9367. [PMID: 25184691 DOI: 10.1021/nn503349g] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
We present herein a pH-responsive dynamic DNA nanocluster based on gold nanoparticles with highly packed nucleic acid assembly and evaluate its potential as a drug delivery vehicle with tumor-specific accumulation. Each gold nanoparticle was readily functionalized with various functional DNA sequences; in particular, we modified the surface of gold nanoparticles with bcl-2 antisense and i-motif binding sequences. Clustering of the gold nanoparticles induced by hybridization of each DNA sequence via i-motif DNA provided tumor targeting and drug loading capabilities. After cellular uptake, the drug was released by disassembly of the gold nanoparticle cluster into single gold nanoparticles in response to the pH decrease in the late endosome. Furthermore, the antiapoptotic Bcl-2 protein was down-regulated by the antisense-modified gold nanoparticles; thus, drug-mediated apoptosis was significantly accelerated by sensitizing the cancer cells to the drug. Our size-tunable clustered nucleic acid-grafted gold nanoparticles provide tumor homing in the blood circulation and are thus a potential multifunctional therapeutic agent in vivo as well as in vitro.
Collapse
Affiliation(s)
- Jinhwan Kim
- Center for Self-assembly and Complexity, Institute for Basic Science (IBS) , Pohang 790-784, Korea
| | | | | | | |
Collapse
|
381
|
Jeong EH, Ryu JH, Jeong H, Jang B, Lee HY, Hong S, Lee H, Lee H. Efficient delivery of siRNAs by a photothermal approach using plant flavonoid-inspired gold nanoshells. Chem Commun (Camb) 2014; 50:13388-90. [PMID: 25234123 DOI: 10.1039/c4cc07155g] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Polymer gold nanoshells (PGNs) are prepared by a novel plant-inspired flavonoid surface modification method. The PGNs show dramatic photothermal properties, which can facilitate the endosomal escape and delivery of siRNA into the cytoplasm of cells. Efficient gene silencing has been achieved using siRNA immobilized PGNs, suggesting the potential applications of in vitro gene regulation by an external NIR stimulus.
Collapse
Affiliation(s)
- Eun Hye Jeong
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 120-750, Republic of Korea.
| | | | | | | | | | | | | | | |
Collapse
|
382
|
Banga R, Chernyak N, Narayan SP, Nguyen ST, Mirkin CA. Liposomal spherical nucleic acids. J Am Chem Soc 2014; 136:9866-9. [PMID: 24983505 PMCID: PMC4280063 DOI: 10.1021/ja504845f] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Indexed: 12/17/2022]
Abstract
A novel class of metal-free spherical nucleic acid nanostructures was synthesized from readily available starting components. These particles consist of 30 nm liposomal cores, composed of an FDA-approved 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) lipid monomer. The surface of the liposomes was functionalized with DNA strands modified with a tocopherol tail that intercalates into the phospholipid layer of the liposomal core via hydrophobic interactions. The spherical nucleic acid architecture not only stabilizes these constructs but also facilitates cellular internalization and gene regulation in SKOV-3 cells.
Collapse
Affiliation(s)
- Resham
J. Banga
- International Institute of Nanotechnology, Department of Chemical
and Biological
Engineering, Department of Chemistry, and Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Natalia Chernyak
- International Institute of Nanotechnology, Department of Chemical
and Biological
Engineering, Department of Chemistry, and Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Suguna P. Narayan
- International Institute of Nanotechnology, Department of Chemical
and Biological
Engineering, Department of Chemistry, and Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - SonBinh T. Nguyen
- International Institute of Nanotechnology, Department of Chemical
and Biological
Engineering, Department of Chemistry, and Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chad A. Mirkin
- International Institute of Nanotechnology, Department of Chemical
and Biological
Engineering, Department of Chemistry, and Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
383
|
Jensen SA, Day ES, Ko CH, Hurley LA, Luciano JP, Kouri FM, Merkel TJ, Luthi AJ, Patel PC, Cutler JI, Daniel WL, Scott AW, Rotz MW, Meade TJ, Giljohann DA, Mirkin CA, Stegh AH. Spherical nucleic acid nanoparticle conjugates as an RNAi-based therapy for glioblastoma. Sci Transl Med 2014; 5:209ra152. [PMID: 24174328 DOI: 10.1126/scitranslmed.3006839] [Citation(s) in RCA: 420] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Glioblastoma multiforme (GBM) is a neurologically debilitating disease that culminates in death 14 to 16 months after diagnosis. An incomplete understanding of how cataloged genetic aberrations promote therapy resistance, combined with ineffective drug delivery to the central nervous system, has rendered GBM incurable. Functional genomics efforts have implicated several oncogenes in GBM pathogenesis but have rarely led to the implementation of targeted therapies. This is partly because many "undruggable" oncogenes cannot be targeted by small molecules or antibodies. We preclinically evaluate an RNA interference (RNAi)-based nanomedicine platform, based on spherical nucleic acid (SNA) nanoparticle conjugates, to neutralize oncogene expression in GBM. SNAs consist of gold nanoparticles covalently functionalized with densely packed, highly oriented small interfering RNA duplexes. In the absence of auxiliary transfection strategies or chemical modifications, SNAs efficiently entered primary and transformed glial cells in vitro. In vivo, the SNAs penetrated the blood-brain barrier and blood-tumor barrier to disseminate throughout xenogeneic glioma explants. SNAs targeting the oncoprotein Bcl2Like12 (Bcl2L12)--an effector caspase and p53 inhibitor overexpressed in GBM relative to normal brain and low-grade astrocytomas--were effective in knocking down endogenous Bcl2L12 mRNA and protein levels, and sensitized glioma cells toward therapy-induced apoptosis by enhancing effector caspase and p53 activity. Further, systemically delivered SNAs reduced Bcl2L12 expression in intracerebral GBM, increased intratumoral apoptosis, and reduced tumor burden and progression in xenografted mice, without adverse side effects. Thus, silencing antiapoptotic signaling using SNAs represents a new approach for systemic RNAi therapy for GBM and possibly other lethal malignancies.
Collapse
Affiliation(s)
- Samuel A Jensen
- Ken and Ruth Davee Department of Neurology, The Northwestern Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, 303 East Superior, Chicago, IL 60611, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
384
|
Probing the inherent stability of siRNA immobilized on nanoparticle constructs. Proc Natl Acad Sci U S A 2014; 111:9739-44. [PMID: 24946803 DOI: 10.1073/pnas.1409431111] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Small interfering RNA (siRNA) is a powerful and highly effective method to regulate gene expression in vitro and in vivo. However, the susceptibility to serum nuclease-catalyzed degradation is a major challenge and it remains unclear whether the strategies developed to improve the stability of siRNA free in serum solution are ideal for siRNA conjugated to nanoparticle surfaces. Herein, we use spherical nucleic acid nanoparticle conjugates, consisting of gold nanoparticles (AuNPs) with siRNA chemisorbed to the surface, as a platform to study how a model siRNA targeting androgen receptor degrades in serum (SNA-siRNAAR). In solutions of 10% (vol/vol) FBS, we find rapid endonuclease hydrolysis at specific sites near the AuNP-facing terminus of siRNAAR, which were different from those of siRNAAR free in solution. These data indicate that the chemical environment of siRNA on a nanoparticle surface can alter the recognition of siRNA by serum nucleases and change the inherent stability of the nucleic acid. Finally, we demonstrate that incorporation of 2'-O-methyl RNA nucleotides at sites of nuclease hydrolysis on SNA-siRNAAR results in a 10-fold increase in siRNA lifetime. These data suggest that strategies for enhancing the serum stability of siRNA immobilized to nanoparticles must be developed from a dedicated analysis of the siRNA-nanoparticle conjugate, rather than a reliance on strategies developed for siRNA free in solution. We believe these findings are important for fundamentally understanding interactions between biological media and oligonucleotides conjugated to nanoparticles for the development of gene regulatory and therapeutic agents in a variety of disease models.
Collapse
|
385
|
Liang H, Zhang XB, Lv Y, Gong L, Wang R, Zhu X, Yang R, Tan W. Functional DNA-containing nanomaterials: cellular applications in biosensing, imaging, and targeted therapy. Acc Chem Res 2014; 47:1891-901. [PMID: 24780000 PMCID: PMC4066909 DOI: 10.1021/ar500078f] [Citation(s) in RCA: 248] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
DNA performs
a vital function as a carrier of genetic code, but in the field of
nanotechnology, DNA molecules can catalyze chemical reactions in the
cell, that is, DNAzymes, or bind with target-specific ligands, that
is, aptamers. These functional DNAs with different modifications have
been developed for sensing, imaging, and therapeutic systems. Thus,
functional DNAs hold great promise for future applications in nanotechnology
and bioanalysis. However, these functional DNAs face challenges, especially
in the field of biomedicine. For example, functional DNAs typically
require the use of cationic transfection reagents to realize cellular
uptake. Such reagents enter the cells, increasing the difficulty of
performing bioassays in vivo and potentially damaging the cell’s
nucleus. To address this obstacle, nanomaterials, such as metallic,
carbon, silica, or magnetic materials, have been utilized as DNA carriers
or assistants. In this Account, we describe selected examples of functional
DNA-containing nanomaterials and their applications from our recent
research and those of others. As models, we have chosen to highlight
DNA/nanomaterial complexes consisting of gold nanoparticles, graphene oxides, and aptamer–micelles, and we illustrate the potential
of such complexes in biosensing, imaging, and medical diagnostics. Under proper conditions, multiple ligand–receptor interactions,
decreased steric hindrance, and increased surface roughness can be
achieved from a high density of DNA that is bound to the surface of
nanomaterials, resulting in a higher affinity for complementary DNA
and other targets. In addition, this high density of DNA causes a
high local salt concentration and negative charge density, which can
prevent DNA degradation. For example, DNAzymes assembled on gold nanoparticles
can effectively catalyze chemical reactions even in living cells.
And it has been confirmed that DNA–nanomaterial complexes can
enter cells more easily than free single-stranded DNA. Nanomaterials
can be designed and synthesized in needed sizes and shapes, and they
possess unique chemical and physical properties, which make them useful
as DNA carriers or assistants, excellent signal reporters, transducers,
and amplifiers. When nanomaterials are combined with functional DNAs
to create novel assay platforms, highly sensitive biosensing and high-resolution
imaging result. For example, gold nanoparticles and graphene oxides
can quench fluorescence efficiently to achieve low background and
effectively increase the signal-to-background ratio. Meanwhile, gold
nanoparticles themselves can be colorimetric reporters because of
their different optical absorptions between monodispersion and aggregation. DNA self-assembled nanomaterials contain several properties of
both DNA and nanomaterials. Compared with DNA–nanomaterial
complexes, DNA self-assembled nanomaterials more closely resemble
living beings, and therefore they have lower cytotoxicity at high
concentrations. Functional DNA self-assemblies also have high density
of DNA for multivalent reaction and three-dimensional nanostructures
for cell uptake. Now and in the future, we envision the use of DNA
bases in making designer molecules for many challenging applications
confronting chemists. With the further development of artificial DNA
bases using smart organic synthesis, DNA macromolecules based on elegant
molecular assembly approaches are expected to achieve great diversity,
additional versatility, and advanced functions.
Collapse
Affiliation(s)
- Hao Liang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory
of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Collaborative Innovation Center of Molecular Engineering for Theranostics, Hunan University, Changsha, Hunan 410082, China
| | - Xiao-Bing Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory
of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Collaborative Innovation Center of Molecular Engineering for Theranostics, Hunan University, Changsha, Hunan 410082, China
| | - Yifan Lv
- Molecular Science and Biomedicine Laboratory, State Key Laboratory
of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Collaborative Innovation Center of Molecular Engineering for Theranostics, Hunan University, Changsha, Hunan 410082, China
| | - Liang Gong
- Molecular Science and Biomedicine Laboratory, State Key Laboratory
of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Collaborative Innovation Center of Molecular Engineering for Theranostics, Hunan University, Changsha, Hunan 410082, China
| | - Ruowen Wang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory
of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Collaborative Innovation Center of Molecular Engineering for Theranostics, Hunan University, Changsha, Hunan 410082, China
| | - Xiaoyan Zhu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory
of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Collaborative Innovation Center of Molecular Engineering for Theranostics, Hunan University, Changsha, Hunan 410082, China
| | - Ronghua Yang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory
of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Collaborative Innovation Center of Molecular Engineering for Theranostics, Hunan University, Changsha, Hunan 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory
of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical
Engineering, College of Biology, Collaborative Innovation Center of Molecular Engineering for Theranostics, Hunan University, Changsha, Hunan 410082, China
- Center for Research at Bio/nano Interface,
Department of Chemistry and Department of Physiology and Functional
Genomics, Shands Cancer Center, UF Genetics Institute, and McKnight
Brain Institute, University of Florida, Gainesville, Florida 32611-7200, United States
| |
Collapse
|
386
|
Fan Y, Li F, Chen D. Scavenger receptor-recognized and enzyme-responsive nanoprobe for fluorescent labeling of lysosomes in live cells. Biomaterials 2014; 35:7870-80. [PMID: 24929616 DOI: 10.1016/j.biomaterials.2014.05.054] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 05/20/2014] [Indexed: 12/23/2022]
Abstract
Lysosomal imaging represents a potent tool for investigating the organization of related cellular events and their modulation via diagnostic and therapeutic approaches. However, specific labeling of the lysosome in live cells is a significant challenge. Taking advantage of the inherent lysosomal entry of nanoparticles and unique digestive inclusions in the lysosome, we developed a nanoparticle-based, enzyme-switchable fluorescence OFF-ON strategy for specific labeling of the lysosome and further imaging of extracellular acidification-induced lysosome trafficking in living cells. The nanoprobe comprised a 16 nm spherical gold nanoparticle as the core and an enzyme-responsive oligomer of fluorescein-conjugated oligo(4-vinyl-phenyl phosphate) as the shell. Due to quenching of the core gold nanoparticle, the nanoprobe was non-fluorescent. After incubation with cancer cells, the nanoprobe was rapidly internalized via scavenger receptor-mediated endocytosis and significantly shuffled into the lysosome. The nanoprobe specifically lighted up the lysosome owing to lysosome-induced fluorescence enhancement. Specifically, digestive inclusions in the lysosome hydrolyzed and released gold-quenched fluorescein molecules, leading to significant augmentation of fluorescence. On account of specific lysosomal labeling, the nanoprobe effectively facilitated imaging of a 4-6 μm anterograde trafficking event of the lysosome from the perinuclear region to the cell surface when an acidic extracellular environment developed. Our findings collectively highlight the use of nanoprobes for lysosomal imaging.
Collapse
Affiliation(s)
- Yanbin Fan
- Department of Macromolecular Science, The State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, PR China
| | - Fuyou Li
- Department of Chemistry, The State Key Laboratory of Molecular Engineering of Polymers, Institute of Biomedicine Science, Fudan University, Shanghai 200433, PR China.
| | - Daoyong Chen
- Department of Macromolecular Science, The State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, PR China.
| |
Collapse
|
387
|
Pinheiro VB, Holliger P. Towards XNA nanotechnology: new materials from synthetic genetic polymers. Trends Biotechnol 2014; 32:321-8. [PMID: 24745974 PMCID: PMC4039137 DOI: 10.1016/j.tibtech.2014.03.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 03/17/2014] [Accepted: 03/18/2014] [Indexed: 12/21/2022]
Abstract
Nucleic acids display remarkable properties beyond information storage and propagation. The well-understood base pairing rules have enabled nucleic acids to be assembled into nanostructures of ever increasing complexity. Although nanostructures can be constructed using other building blocks, including peptides and lipids, it is the capacity to evolve that sets nucleic acids apart from all other nanoscale building materials. Nonetheless, the poor chemical and biological stability of DNA and RNA constrain their applications. Recent advances in nucleic acid chemistry and polymerase engineering enable the synthesis, replication, and evolution of a range of synthetic genetic polymers (XNAs) with improved chemical and biological stability. We discuss the impact of this technology on the generation of XNA ligands, enzymes, and nanostructures with tailor-made chemistry.
Collapse
Affiliation(s)
- Vitor B Pinheiro
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Philipp Holliger
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK.
| |
Collapse
|
388
|
Wu X, Choi CHJ, Zhang C, Hao L, Mirkin CA. Intracellular fate of spherical nucleic acid nanoparticle conjugates. J Am Chem Soc 2014; 136:7726-33. [PMID: 24841494 PMCID: PMC4046773 DOI: 10.1021/ja503010a] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Indexed: 12/19/2022]
Abstract
Spherical nucleic acid (SNA) nanoparticle conjugates are a class of bionanomaterials that are extremely potent in many biomedical applications. Their unique ability to enter multiple mammalian cell types as single-entity agents arises from their novel three-dimensional architecture, which consists of a dense shell of highly oriented oligonucleotides chemically attached typically to a gold nanoparticle core. This architecture allows SNAs to engage certain cell surface receptors to facilitate entry. Here, we report studies aimed at determining the intracellular fate of SNAs and the trafficking events that occur inside C166 mouse endothelial cells after cellular entry. We show that SNAs traffic through the endocytic pathway into late endosomes and reside there for up to 24 h after incubation. Disassembly of oligonucleotides from the nanoparticle core is observed 16 h after cellular entry, most likely due to degradation by enzymes such as DNase II localized in late endosomes. Our observations point to these events being likely independent of core composition and treatment conditions, and they do not seem to be particularly dependent upon oligonucleotide sequence. Significantly and surprisingly, the SNAs do not enter the lysosomes under the conditions studied. To independently track the fate of the particle core and the fluorophore-labeled oligonucleotides that comprise its shell, we synthesized a novel class of quantum dot SNAs to determine that as the SNA structures are broken down over the 24 h time course of the experiment, the oligonucleotide fragments are recycled out of the cell while the nanoparticle core is not. This mechanistic insight points to the importance of designing and synthesizing next-generation SNAs that can bypass the degradation bottleneck imposed by their residency in late endosomes, and it also suggests that such structures might be extremely useful for endosomal signaling pathways by engaging receptors that are localized within the endosome.
Collapse
Affiliation(s)
| | | | - Chuan Zhang
- International Institute for
Nanotechnology and Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United
States
| | - Liangliang Hao
- International Institute for
Nanotechnology and Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United
States
| | - Chad A. Mirkin
- International Institute for
Nanotechnology and Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United
States
| |
Collapse
|
389
|
Rush AM, Nelles DA, Blum AP, Barnhill SA, Tatro ET, Yeo GW, Gianneschi NC. Intracellular mRNA regulation with self-assembled locked nucleic acid polymer nanoparticles. J Am Chem Soc 2014; 136:7615-8. [PMID: 24827740 PMCID: PMC4046771 DOI: 10.1021/ja503598z] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
![]()
We present an untemplated,
single-component antisense oligonucleotide
delivery system capable of regulating mRNA abundance in live human
cells. While most approaches to nucleic acid delivery rely on secondary
carriers and complex multicomponent charge-neutralizing formulations,
we demonstrate efficient delivery using a simple locked nucleic acid
(LNA)-polymer conjugate that assembles into spherical micellar nanoparticles
displaying a dense shell of nucleic acid at the surface. Cellular
uptake of soft LNA nanoparticles occurs rapidly within minutes as
evidenced by flow cytometry and fluorescence microscopy. Importantly,
these LNA nanoparticles knockdown survivin mRNA, an established target
for cancer therapy, in a sequence-specific fashion as analyzed by
RT-PCR.
Collapse
Affiliation(s)
- Anthony M Rush
- Department of Chemistry & Biochemistry, ‡Stem Cell Program and Institute for Genomic Medicine, §Department of Cellular and Molecular Medicine, ∥Department of Psychiatry, ⊥Materials Science and Engineering, University of California , San Diego, La Jolla, California 92093, United States
| | | | | | | | | | | | | |
Collapse
|
390
|
Morris W, Briley WE, Auyeung E, Cabezas MD, Mirkin CA. Nucleic Acid–Metal Organic Framework (MOF) Nanoparticle Conjugates. J Am Chem Soc 2014; 136:7261-4. [DOI: 10.1021/ja503215w] [Citation(s) in RCA: 346] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- William Morris
- Department
of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - William E. Briley
- Interdepartmental
Biological Sciences, Northwestern University, 2205 Tech Drive, Evanston, Illinois 60208, United States
| | - Evelyn Auyeung
- Department
of Materials Science and Engineering, Northwestern University, 2220 Campus
Drive, Evanston, Illinois 60208, United States
| | - Maria D. Cabezas
- Department
of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chad A. Mirkin
- Department
of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
391
|
Charoenphol P, Bermudez H. Design and application of multifunctional DNA nanocarriers for therapeutic delivery. Acta Biomater 2014; 10:1683-91. [PMID: 23896566 DOI: 10.1016/j.actbio.2013.07.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 06/17/2013] [Accepted: 07/17/2013] [Indexed: 12/31/2022]
Abstract
The unique programmability of nucleic acids offers versatility and flexibility in the creation of self-assembled DNA nanostructures. To date, many three-dimensional DNA architectures of varying sizes and shapes have been precisely formed. Their biocompatibility, biodegradability and high intrinsic stability in physiological environments emphasize their emerging use as carriers for drug and gene delivery. Furthermore, DNA nanocarriers have been shown to enter cells efficiently and without the aid of transfection reagents. A key strength of DNA nanocarriers over other delivery systems is their modularity and their ability to control the spatial distribution of cargoes and ligands. Optimizing DNA nanocarrier properties to dictate their localization, uptake and intracellular trafficking is also possible. This review presents design considerations for DNA nanocarriers and examples of their use in the context of therapeutic delivery applications. The assembly of DNA nanocarriers and approaches for loading and releasing cargo are described. The stability and safety of DNA nanocarriers are also discussed, with particular attention to the in vivo physiological environment. Mechanisms of cellular uptake and intracellular trafficking are examined, and the paper concludes with strategies to enhance the delivery efficiency of DNA nanocarriers.
Collapse
Affiliation(s)
- P Charoenphol
- Department of Polymer Science and Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - H Bermudez
- Department of Polymer Science and Engineering, University of Massachusetts, Amherst, MA 01003, USA.
| |
Collapse
|
392
|
Xia XX, Wang M, Lin Y, Xu Q, Kaplan DL. Hydrophobic drug-triggered self-assembly of nanoparticles from silk-elastin-like protein polymers for drug delivery. Biomacromolecules 2014; 15:908-14. [PMID: 24527851 PMCID: PMC3983132 DOI: 10.1021/bm4017594] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Revised: 02/11/2014] [Indexed: 11/28/2022]
Abstract
Silk-elastin-like protein polymers (SELPs) combine the mechanical and biological properties of silk and elastin. These properties have led to the development of various SELP-based materials for drug delivery. However, SELPs have rarely been developed into nanoparticles, partially due to the complicated fabrication procedures, nor assessed for potential as an anticancer drug delivery system. We have recently constructed a series of SELPs (SE8Y, S2E8Y, and S4E8Y) with various ratios of silk to elastin blocks and described their capacity to form micellar-like nanoparticles upon thermal triggering. In this study, we demonstrate that doxorubicin, a hydrophobic antitumor drug, can efficiently trigger the self-assembly of SE8Y (SELPs with silk to elastin ratio of 1:8) into uniform micellar-like nanoparticles. The drug can be loaded in the SE8Y nanoparticles with an efficiency around 6.5% (65 ng doxorubicin/μg SE8Y), S2E8Y with 6%, and S4E8Y with 4%, respectively. In vitro studies with HeLa cell lines demonstrate that the protein polymers are not cytotoxic (IC50 > 200 μg/mL), while the doxorubicin-loaded SE8Y nanoparticles showed a 1.8-fold higher cytotoxicity than the free drug. Confocal laser scanning microscopy (CLSM) and flow cytometry indicate significant uptake of the SE8Y nanoparticles by the cells and suggest internalization of the nanoparticles through endocytosis. This study provides an all-aqueous, facile method to prepare nanoscale, drug-loaded SELPs packages with potential for tumor cell treatments.
Collapse
Affiliation(s)
- Xiao-Xia Xia
- State
Key Laboratory of Microbial Metabolism, School of Life Sciences and
Biotechnology, Shanghai Jiao Tong University, 800 Dong-Chuan Road, Shanghai, 200240, China
| | - Ming Wang
- Department
of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Yinan Lin
- Department
of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Qiaobing Xu
- Department
of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - David L. Kaplan
- Department
of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| |
Collapse
|
393
|
Gold nanoparticles for nucleic acid delivery. Mol Ther 2014; 22:1075-1083. [PMID: 24599278 DOI: 10.1038/mt.2014.30] [Citation(s) in RCA: 331] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Accepted: 02/21/2014] [Indexed: 12/11/2022] Open
Abstract
Gold nanoparticles provide an attractive and applicable scaffold for delivery of nucleic acids. In this review, we focus on the use of covalent and noncovalent gold nanoparticle conjugates for applications in gene delivery and RNA-interference technologies. We also discuss challenges in nucleic acid delivery, including endosomal entrapment/escape and active delivery/presentation of nucleic acids in the cell.
Collapse
|
394
|
Alhasan AH, Patel PC, Choi CHJ, Mirkin CA. Exosome encased spherical nucleic acid gold nanoparticle conjugates as potent microRNA regulation agents. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:186-92. [PMID: 24106176 PMCID: PMC3947239 DOI: 10.1002/smll.201302143] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Indexed: 05/25/2023]
Abstract
Exosomes are a class of naturally occurring nanomaterials that play crucial roles in the protection and transport of endogenous macromolecules, such as microRNA and mRNA, over long distances. Intense effort is underway to exploit the use of exosomes to deliver synthetic therapeutics. Herein, transmission electron microscopy is used to show that when spherical nucleic acid (SNA) constructs are endocytosed into PC-3 prostate cancer cells, a small fraction of them (<1%) can be naturally sorted into exosomes. The exosome-encased SNAs are secreted into the extracellular environment from which they can be isolated and selectively re-introduced into the cell type from which they were derived. In the context of anti-miR21 experiments, the exosome-encased SNAs knockdown miR-21 target by approximately 50%. Similar knockdown of miR-21 by free SNAs requires a ≈3000-fold higher concentration.
Collapse
Affiliation(s)
- Ali H. Alhasan
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA
- Interdepartmental Biological Sciences Program, Northwestern University, 2205 Tech Drive, Evanston, IL 60208-3113, USA
| | - Pinal C. Patel
- AuraSense Therapeutics, LLC, 8045 Lamon Avenue, Suite 410, Skokie, IL 60077
| | - Chung Hang J. Choi
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA
| | - Chad A. Mirkin
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208-3113, USA
| |
Collapse
|
395
|
Juliano RL, Ming X, Carver K, Laing B. Cellular uptake and intracellular trafficking of oligonucleotides: implications for oligonucleotide pharmacology. Nucleic Acid Ther 2014; 24:101-13. [PMID: 24383421 DOI: 10.1089/nat.2013.0463] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
One of the major constraints on the therapeutic use of oligonucleotides is inefficient delivery to their sites of action in the cytosol or nucleus. Recently it has become evident that the pathways of cellular uptake and intracellular trafficking of oligonucleotides can strongly influence their pharmacological actions. Here we provide background information on the basic processes of endocytosis and trafficking and then review recent literature on targeted delivery and subcellular trafficking of oligonucleotides in that context. A variety of approaches including molecular scale ligand-oligonucleotide conjugates, ligand-targeted nanocarriers, and the use of small molecules to enhance oligonucleotide effects are discussed.
Collapse
Affiliation(s)
- R L Juliano
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina
| | | | | | | |
Collapse
|
396
|
Nanoparticles: Cellular Uptake and Cytotoxicity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 811:73-91. [DOI: 10.1007/978-94-017-8739-0_5] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
397
|
|
398
|
Heuer-Jungemann A, Harimech PK, Brown T, Kanaras AG. Gold nanoparticles and fluorescently-labelled DNA as a platform for biological sensing. NANOSCALE 2013; 5:9503-9510. [PMID: 23982570 DOI: 10.1039/c3nr03707j] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
In the past decade gold nanoparticle-nucleic acid conjugates became progressively important for biomedical applications. Fluorophores attached to nucleic acid-gold nanoparticle conjugates have opened up a new era of biological sensing. The most promising advancement in this field was the invention of the so-called 'nano-flare' systems. These systems are capable of detecting specific endocellular targets such as mRNAs, microRNAs or small molecules in real time. In this minireview, we discuss the current progress in the field of DNA-nanoparticles as sensors, their properties, stability, cellular uptake and cytotoxicity.
Collapse
Affiliation(s)
- Amelie Heuer-Jungemann
- Physics and Astronomy, Faculty of Physical Sciences and Engineering, University of Southampton, Southampton, SO17 1BJ, UK.
| | | | | | | |
Collapse
|