151
|
Kim Y, Ko SM, Nam JM. Protein-Nanoparticle Interaction-Induced Changes in Protein Structure and Aggregation. Chem Asian J 2016; 11:1869-77. [DOI: 10.1002/asia.201600236] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Yuna Kim
- Seoul National University; Department of Chemistry; Seoul 151-742 South Korea
| | - Sung Min Ko
- Seoul National University; Department of Chemistry; Seoul 151-742 South Korea
| | - Jwa-Min Nam
- Seoul National University; Department of Chemistry; Seoul 151-742 South Korea
| |
Collapse
|
152
|
|
153
|
Xing H, Li J, Xu W, Hwang K, Wu P, Yin Q, Li Z, Cheng J, Lu Y. The Effects of Spacer Length and Composition on Aptamer-Mediated Cell-Specific Targeting with Nanoscale PEGylated Liposomal Doxorubicin. Chembiochem 2016; 17:1111-7. [PMID: 27123758 DOI: 10.1002/cbic.201600092] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Indexed: 12/27/2022]
Abstract
Aptamer-based targeted drug delivery systems have shown significant promise for clinical applications. Although much progress has been made in this area, it remains unclear how PEG coating would affect the selective binding of DNA aptamers and thus influence the overall targeting efficiency. To answer this question, we herein report a systematic investigation of the interactions between PEG and DNA aptamers on the surface of liposomes by using a series of nanoscale liposomal doxorubicin formulations with different DNA aptamer and PEG modifications. We investigated how the spatial size and composition of the spacer molecules affected the targeting ability of the liposome delivery system. We showed that a spacer of appropriate length was critical to overcome the shielding from surrounding PEG molecules in order to achieve the best targeting effect, regardless of the spacer composition. Our findings provide important guidelines for the design of aptamer-based targeted drug delivery systems.
Collapse
Affiliation(s)
- Hang Xing
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Avenue, Urbana, IL, 61801, USA
| | - Ji Li
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Avenue, Urbana, IL, 61801, USA
| | - Weidong Xu
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Kevin Hwang
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Avenue, Urbana, IL, 61801, USA
| | - Peiwen Wu
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Avenue, Urbana, IL, 61801, USA
| | - Qian Yin
- Material Science and Engineering, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Avenue, Urbana, IL, 61801, USA
| | - Zhensheng Li
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Avenue, Urbana, IL, 61801, USA
| | - Jianjun Cheng
- Material Science and Engineering, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Avenue, Urbana, IL, 61801, USA
| | - Yi Lu
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N Mathews Avenue, Urbana, IL, 61801, USA.
| |
Collapse
|
154
|
Fitzgerald KA, Rahme K, Guo J, Holmes JD, O'Driscoll CM. Anisamide-targeted gold nanoparticles for siRNA delivery in prostate cancer - synthesis, physicochemical characterisation and in vitro evaluation. J Mater Chem B 2016; 4:2242-2252. [PMID: 32263220 DOI: 10.1039/c6tb00082g] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Metastatic prostate cancer is a leading cause of cancer-related death in men and current chemotherapies are largely inadequate in terms of efficacy and toxicity. Hence improved treatments are required. The application of siRNA as a cancer therapeutic holds great promise. However, translation of siRNA into the clinic is dependent on the availability of an effective delivery system. Gold nanoparticles (AuNPs) are known to be effective and non-toxic siRNA delivery agents. In this study, a stable gold nanosphere coated with poly(ethylenimine) (PEI) was prepared to yield PEI capped AuNPs (Au-PEI). The PEI was further conjugated with the targeting ligand anisamide (AA, is known to bind to the sigma receptor overexpressed on the surface of prostate cancer cells) to produce an anisamide-targeted nanoparticle (Au-PEI-AA). The resulting untargeted and targeted nanoparticles (Au-PEI and Au-PEI-AA respectively) were positively charged and efficiently complexed siRNA. Au-PEI-AA mediated siRNA uptake into PC3 prostate cancer cells via binding to the sigma receptor. In addition, the Au-PEI-AA·siRNA complexes resulted in highly efficient knockdown of the RelA gene (∼70%) when cells were transfected in serum-free medium. In contrast, no knockdown was observed in the presence of serum, suggesting that adsorption of serum proteins inhibits the binding of the anisamide moiety to the sigma receptor. This study provides (for the first time) proof of principle that anisamide-labelled gold nanoparticles can target the sigma receptor. Further optimisation of the formulation to increase serum stability will enhance its potential to treat prostate cancer.
Collapse
|
155
|
Abstract
The convergence of nanoscience and drug delivery has prompted the formation of the field of nanomedicine, one that exploits the novel physicochemical and biological properties of nanostructures for improved medical treatments and reduced side effects. Until recently, this nanostructure-mediated strategy considered the drug to be solely a biologically active compound to be delivered, and its potential as a molecular building unit remained largely unexplored. A growing trend within nanomedicine has been the use of drug molecules to build well-defined nanostructures of various sizes and shapes. This strategy allows for the creation of self-delivering supramolecular nanomedicines containing a high and fixed drug content. Through rational design of the number and type of the drug incorporated, the resulting nanostructures can be tailored to assume various morphologies (e.g. nanospheres, rods, nanofibers, or nanotubes) for a particular mode of administration such as systemic, topical, and local delivery. This review covers the recent advances in this rapidly developing field, with the aim of providing an in-depth evaluation of the exciting opportunities that this new field could create to improve the current clinical practice of nanomedicine.
Collapse
Affiliation(s)
- Wang Ma
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Eastern Road, Zhengzhou, Henan 450052, China
| | - Andrew G. Cheetham
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA
| | - Honggang Cui
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Eastern Road, Zhengzhou, Henan 450052, China
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, Maryland 21231, USA
| |
Collapse
|
156
|
Moyano DF, Liu Y, Peer D, Rotello VM. Modulation of Immune Response Using Engineered Nanoparticle Surfaces. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:76-82. [PMID: 26618755 PMCID: PMC4749139 DOI: 10.1002/smll.201502273] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 10/10/2015] [Indexed: 05/28/2023]
Abstract
Nanoparticles (NPs) coated with a monolayer of ligands can be recognized by different components of the immune system, opening new doors for the modulation of immunological responses. By the use of different physical or chemical properties at the NP surface (such as charge, functional groups, and ligand density), NPs can be designed to have distinct cellular uptake, cytokine secretion, and immunogenicity, factors that influence the distribution and clearance of these particles. Understanding these immunological responses is critical for the development of new NP-based carriers for the delivery of therapeutic molecules, and as such several studies have been performed to understand the relationships between immune responses and NP surface functionality. In this review, we will discuss recent reports of these structure-activity relationships, and explore how these motifs can be controlled to elicit therapeutically useful immune responses.
Collapse
Affiliation(s)
- Daniel F. Moyano
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA. Tel: (+1) 413-545-2058
| | - Yuanchang Liu
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA. Tel: (+1) 413-545-2058
| | - Dan Peer
- Laboratory of Nanomedicine, Department of Cell Research and Immunology, Department of Materials Science and Engineering, Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv , 69978, Israel. Tel (+972) 3640-7925
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA. Tel: (+1) 413-545-2058
| |
Collapse
|
157
|
Juks C, Padari K, Margus H, Kriiska A, Etverk I, Arukuusk P, Koppel K, Ezzat K, Langel Ü, Pooga M. The role of endocytosis in the uptake and intracellular trafficking of PepFect14–nucleic acid nanocomplexes via class A scavenger receptors. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:3205-16. [DOI: 10.1016/j.bbamem.2015.09.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 09/02/2015] [Accepted: 09/22/2015] [Indexed: 01/24/2023]
|
158
|
Abstract
Messenger RNA (mRNA) has recently emerged with remarkable potential as an effective alternative to DNA-based therapies because of several unique advantages. mRNA does not require nuclear entry for transfection activity and has a negligible chance of integrating into the host genome which excludes the possibility of potentially detrimental genomic alternations. Chemical modification of mRNA has further enhanced its stability and decreased its activation of innate immune responses. Additionally, mRNA has been found to have rapid expression and predictable kinetics. Nevertheless, the ubiquitous application of mRNA remains challenging given its unfavorable attributes, such as large size, negative charge and susceptibility to enzymatic degradation. Further refinement of mRNA delivery modalities is therefore essential for its development as a therapeutic tool. This review provides an exclusive overview of current state-of-the-art biomaterials and nanotechnology platforms for mRNA delivery, and discusses future prospects to bring these exciting technologies into clinical practice.
Collapse
Affiliation(s)
- Mohammad Ariful Islam
- Laboratory for Nanoengineering & Drug Delivery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | |
Collapse
|
159
|
Brodin JD, Sprangers AJ, McMillan JR, Mirkin CA. DNA-Mediated Cellular Delivery of Functional Enzymes. J Am Chem Soc 2015; 137:14838-41. [PMID: 26587747 DOI: 10.1021/jacs.5b09711] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We report a strategy for creating a new class of protein transfection materials composed of a functional protein core chemically modified with a dense shell of oligonucleotides. These materials retain the native structure and catalytic ability of the hydrolytic enzyme β-galactosidase, which serves as the protein core, despite the functionalization of its surface with ∼25 DNA strands. The covalent attachment of a shell of oligonucleotides to the surface of β-galactosidase enhances its cellular uptake of by up to ∼280-fold and allows for the use of working concentrations as low as 100 pM enzyme. DNA-functionalized β-galactosidase retains its ability to catalyze the hydrolysis of β-glycosidic linkages once endocytosed, whereas equal concentrations of protein show little to no intracellular catalytic activity.
Collapse
Affiliation(s)
- Jeffrey D Brodin
- International Institute of Nanotechnology, ‡Department of Chemistry, and §Department of Biomedical Engineering, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Anthony J Sprangers
- International Institute of Nanotechnology, ‡Department of Chemistry, and §Department of Biomedical Engineering, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Janet R McMillan
- International Institute of Nanotechnology, ‡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 Chemistry, and §Department of Biomedical Engineering, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
160
|
Nicholls FJ, Rotz MW, Ghuman H, MacRenaris KW, Meade TJ, Modo M. DNA-gadolinium-gold nanoparticles for in vivo T1 MR imaging of transplanted human neural stem cells. Biomaterials 2015; 77:291-306. [PMID: 26615367 DOI: 10.1016/j.biomaterials.2015.11.021] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 10/21/2015] [Accepted: 11/10/2015] [Indexed: 01/18/2023]
Abstract
The unambiguous imaging of transplanted cells remains a major challenge to understand their biological function and therapeutic efficacy. In vivo imaging of implanted cells is reliant on tagging these to differentiate them from host tissue, such as the brain. We here characterize a gold nanoparticle conjugate that is functionalized with modified deoxythymidine oligonucleotides bearing Gd(III) chelates and a red fluorescent Cy3 moiety to visualize in vivo transplanted human neural stem cells. This DNA-Gd@Au nanoparticle (DNA-Gd@AuNP) exhibits an improved T1 relaxivity and excellent cell uptake. No significant effects of cell uptake have been found on essential cell functions. Although T1 relaxivity is attenuated within cells, it is sufficiently preserved to afford the in vivo detection of transplanted cells using an optimized voxel size. In vivo MR images were corroborated by a post-mortem histological verification of DNA-Gd@AuNPs in transplanted cells. With 70% of cells being correctly identified using the DNA-Gd-AuNPs indicates an overall reliable detection. Less than 1% of cells were false positive for DNA-Gd@AuNPs, but a significant number of 30% false negatives reveals a dramatic underestimation of transplanted cells using this approach. DNA-Gd@AuNPs therefore offer new opportunities to visualize transplanted cells unequivocally using T1 contrast and use cellular MRI as a tool to derive biologically relevant information that allows us to understand how the survival and location of implanted cells determines therapeutic efficacy.
Collapse
Affiliation(s)
- Francesca J Nicholls
- Department of Radiology, University of Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, USA; Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Matthew W Rotz
- Departments of Chemistry, Neurobiology and Radiology, Northwestern University, Evanston, IL, USA; Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Harmanvir Ghuman
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, PA, USA
| | - Keith W MacRenaris
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA; Quantitative Bio-elemental Imaging Centre, Northwestern University, Evanston, IL, USA
| | - Thomas J Meade
- Departments of Chemistry, Neurobiology and Radiology, Northwestern University, Evanston, IL, USA; Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA.
| | - Michel Modo
- Department of Radiology, University of Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, PA, USA.
| |
Collapse
|
161
|
Oligonucleotide therapeutics: chemistry, delivery and clinical progress. Future Med Chem 2015; 7:2221-42. [PMID: 26510815 DOI: 10.4155/fmc.15.144] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Oligonucleotide therapeutics have the potential to become a third pillar of drug development after small molecules and protein therapeutics. However, the three approved oligonucleotide drugs over the past 17 years have not proven to be highly successful in a commercial sense. These trailblazer drugs have nonetheless laid the foundations for entire classes of drug candidates to follow. This review will examine further advances in chemistry that are earlier in the pipeline of oligonucleotide drug candidates. Finally, we consider the possible effect of delivery systems that may provide extra footholds to improve the potency and specificity of oligonucleotide drugs. Our overview focuses on strategies to imbue antisense oligonucleotides with more drug-like properties and their applicability to other nucleic acid therapeutics.
Collapse
|
162
|
Li J, Wu N, Wu J, Wan Y, Liu C. Effect of protein adsorption on cell uptake and blood clearance of methoxy poly(ethylene glycol)-poly(caprolactone) nanoparticles. J Appl Polym Sci 2015. [DOI: 10.1002/app.42884] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jing Li
- Hubei Province Key Laboratory on Cardiovascular; Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology; Xianning Hubei 437100 People's Republic of China
| | - Ninghua Wu
- Hubei Province Key Laboratory on Cardiovascular; Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology; Xianning Hubei 437100 People's Republic of China
| | - Jiliang Wu
- Hubei Province Key Laboratory on Cardiovascular; Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology; Xianning Hubei 437100 People's Republic of China
| | - Ying Wan
- College of Life Science and Technology; Huazhong University of Science and Technology; Wuhan 430074 People's Republic of China
| | - Chao Liu
- Hubei Province Key Laboratory on Cardiovascular; Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology; Xianning Hubei 437100 People's Republic of China
| |
Collapse
|
163
|
Dai Z, Tam DY, Xu H, Chan MS, Liu LS, Bolze F, Sun XH, Lo PK. Conformational Change of Self-Assembled DNA Nanotubes Induced by Two-Photon Excitation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:4090-6. [PMID: 26011412 DOI: 10.1002/smll.201500333] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 04/20/2015] [Indexed: 05/14/2023]
Abstract
Two-photon-regulated, shape-changing DNA nanostructures are demonstrated by integrating a DNA nanotube with a two-photon photocleavable module that enables the opening of the cavities of tube, and becomes partially single-stranded in response to two-photon excitation under 800 nm fs laser pulses.
Collapse
Affiliation(s)
- Ziwen Dai
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Dick Yan Tam
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Hailiang Xu
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Miu Shan Chan
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Ling Sum Liu
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Frédéric Bolze
- Laboratoire de Conception et Application des Molécules Bioactives, UMR Université of Strasbourg-CNRS 7199, Faculté de Pharmacie, Université de Strasbourg, France
| | - Xiao Hua Sun
- School of Biological Industry, Chengdu University, Shiling Town, Chengdu City, Sichuan Province, China
| | - Pik Kwan Lo
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| |
Collapse
|
164
|
Chinen AB, Guan CM, Ferrer JR, Barnaby SN, Merkel TJ, Mirkin CA. Nanoparticle Probes for the Detection of Cancer Biomarkers, Cells, and Tissues by Fluorescence. Chem Rev 2015; 115:10530-74. [PMID: 26313138 DOI: 10.1021/acs.chemrev.5b00321] [Citation(s) in RCA: 629] [Impact Index Per Article: 69.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Alyssa B Chinen
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chenxia M Guan
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Jennifer R Ferrer
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Stacey N Barnaby
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Timothy J Merkel
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chad A Mirkin
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
165
|
Song L, Guo Y, Roebuck D, Chen C, Yang M, Yang Z, Sreedharan S, Glover C, Thomas JA, Liu D, Guo S, Chen R, Zhou D. Terminal PEGylated DNA-Gold Nanoparticle Conjugates Offering High Resistance to Nuclease Degradation and Efficient Intracellular Delivery of DNA Binding Agents. ACS APPLIED MATERIALS & INTERFACES 2015; 7:18707-16. [PMID: 26237203 PMCID: PMC4554298 DOI: 10.1021/acsami.5b05228] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/03/2015] [Indexed: 05/20/2023]
Abstract
Over the past 10 years, polyvalent DNA-gold nanoparticle (DNA-GNP) conjugate has been demonstrated as an efficient, universal nanocarrier for drug and gene delivery with high uptake by over 50 different types of primary and cancer cell lines. A barrier limiting its in vivo effectiveness is limited resistance to nuclease degradation and nonspecific interaction with blood serum contents. Herein we show that terminal PEGylation of the complementary DNA strand hybridized to a polyvalent DNA-GNP conjugate can eliminate nonspecific adsorption of serum proteins and greatly increases its resistance against DNase I-based degradation. The PEGylated DNA-GNP conjugate still retains a high cell uptake property, making it an attractive intracellular delivery nanocarrier for DNA binding reagents. We show that it can be used for successful intracellular delivery of doxorubicin, a widely used clinical cancer chemotherapeutic drug. Moreover, it can be used for efficient delivery of some cell-membrane-impermeable reagents such as propidium iodide (a DNA intercalating fluorescent dye currently limited to the use of staining dead cells only) and a diruthenium complex (a DNA groove binder), for successful staining of live cells.
Collapse
Affiliation(s)
- Lei Song
- School
of Chemistry and Astbury Structure for Molecular Biology, University of Leeds, Leeds LS2 9JT, U.K.
| | - Yuan Guo
- School
of Chemistry and Astbury Structure for Molecular Biology, University of Leeds, Leeds LS2 9JT, U.K.
| | - Deborah Roebuck
- Department
of Chemical Engineering, Imperial College
London, South Kensington
Campus, London SW7 2AZ, U.K.
| | - Chun Chen
- Department
of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Min Yang
- UCL
School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, U.K.
| | - Zhongqiang Yang
- Department
of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | | | - Caroline Glover
- Department
of Chemistry, University of Sheffield, Sheffield S3 7HF, U.K.
| | - Jim A. Thomas
- Department
of Chemistry, University of Sheffield, Sheffield S3 7HF, U.K.
| | - Dongsheng Liu
- Department
of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Shengrong Guo
- School
of Chemistry and Astbury Structure for Molecular Biology, University of Leeds, Leeds LS2 9JT, U.K.
| | - Rongjun Chen
- Department
of Chemical Engineering, Imperial College
London, South Kensington
Campus, London SW7 2AZ, U.K.
| | - Dejian Zhou
- School
of Chemistry and Astbury Structure for Molecular Biology, University of Leeds, Leeds LS2 9JT, U.K.
| |
Collapse
|
166
|
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
|
167
|
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
|
168
|
Fu B, Ren L, Liu D, Ma JZ, An TZ, Yang XQ, Ma H, Guo ZH, Zhu M, Bai J. Using a nano-flare probe to detect RNA in live donor cells prior to somatic cell nuclear transfer. Cell Biol Int 2015; 40:7-15. [PMID: 26109144 DOI: 10.1002/cbin.10504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 06/12/2015] [Indexed: 11/11/2022]
Abstract
Many transgenes are silenced in mammalian cells (donor cells used for somatic cell nuclear transfer [SCNT]). Silencing correlated with a repressed chromatin structure or suppressed promoter, and it impeded the production of transgenic animals. Gene transcription studies in live cells are challenging because of the drawbacks of reverse-transcription polymerase chain reaction and fluorescence in situ hybridization. Nano-flare probes provide an effective approach to detect RNA in living cells. We used 18S RNA, a housekeeping gene, as a reference gene. This study aimed to establish a platform to detect RNA in single living donor cells using a Nano-flare probe prior to SCNT and to verify the safety and validity of the Nano-flare probe in order to provide a technical foundation for rescuing silenced transgenes in transgenic cloned embryos. We investigated cytotoxic effect of the 18S RNA-Nano-flare probe on porcine fetal fibroblasts, characterized the distribution of the 18S RNA-Nano-flare probe in living cells and investigated the effect of the 18S RNA-Nano-flare probe on the development of cloned embryos after SCNT. The cytotoxic effect of the 18S RNA-Nano-flare probe on porcine fetal fibroblasts was dose-dependent, and 18S RNA was detected using the 18S RNA-Nano-flare probe. In addition, treating donor cells with 500 pM 18S RNA-Nano-flare probe did not have adverse effects on the development of SCNT embryos at the pre-implantation stage. In conclusion, we established a preliminary platform to detect RNA in live donor cells using a Nano-flare probe prior to SCNT.
Collapse
Affiliation(s)
- Bo Fu
- Heilongjiang Acamedy of Agricultural Sciences Postdoctoral Programme, Northeast Forestry University Postdoctoral Programme, Harbin, China.,Institute of Animal Husbandry Research, HeiLongJiang Academy of Agricultural Sciences, Harbin, 150086, China
| | - Liang Ren
- College of Animal Science, Northeast Agriculture University, Harbin, 150030, China
| | - Di Liu
- Institute of Animal Husbandry Research, HeiLongJiang Academy of Agricultural Sciences, Harbin, 150086, China.,College of Animal Science, Northeast Agriculture University, Harbin, 150030, China
| | - Jian-Zhang Ma
- Heilongjiang Acamedy of Agricultural Sciences Postdoctoral Programme, Northeast Forestry University Postdoctoral Programme, Harbin, China
| | - Tie-Zhu An
- Heilongjiang Acamedy of Agricultural Sciences Postdoctoral Programme, Northeast Forestry University Postdoctoral Programme, Harbin, China
| | - Xiu-Qin Yang
- College of Animal Science, Northeast Agriculture University, Harbin, 150030, China
| | - Hong Ma
- Institute of Animal Husbandry Research, HeiLongJiang Academy of Agricultural Sciences, Harbin, 150086, China
| | - Zhen-Hua Guo
- Institute of Animal Husbandry Research, HeiLongJiang Academy of Agricultural Sciences, Harbin, 150086, China
| | - Meng Zhu
- College of Animal Science, Northeast Agriculture University, Harbin, 150030, China
| | - Jing Bai
- Modern Education Technology and Information center, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| |
Collapse
|
169
|
Abstract
In this article, advances in designing polymeric nanoparticles for targeted cancer gene therapy are reviewed. Characterization and evaluation of biomaterials, targeting ligands, and transcriptional elements are each discussed. Advances in biomaterials have driven improvements to nanoparticle stability and tissue targeting, conjugation of ligands to the surface of polymeric nanoparticles enable binding to specific cancer cells, and the design of transcriptional elements has enabled selective DNA expression specific to the cancer cells. Together, these features have improved the performance of polymeric nanoparticles as targeted non-viral gene delivery vectors to treat cancer. As polymeric nanoparticles can be designed to be biodegradable, non-toxic, and to have reduced immunogenicity and tumorigenicity compared to viral platforms, they have significant potential for clinical use. Results of polymeric gene therapy in clinical trials and future directions for the engineering of nanoparticle systems for targeted cancer gene therapy are also presented.
Collapse
Affiliation(s)
- Jayoung Kim
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David R. Wilson
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Camila G. Zamboni
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
- Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Jordan J. Green
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
170
|
Eustaquio T, Leary JF. Nanobarcoded superparamagnetic iron oxide nanoparticles for nanomedicine: Quantitative studies of cell-nanoparticle interactions by scanning image cytometry. Cytometry A 2015; 89:207-16. [DOI: 10.1002/cyto.a.22699] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 03/07/2015] [Accepted: 05/02/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Trisha Eustaquio
- Weldon School of Biomedical Engineering; Birck Nanotechnology Center, Purdue University; West Lafayette Indiana
| | - James F. Leary
- Weldon School of Biomedical Engineering, Birck Nanotechnology Center, Bindley Bioscience Center, School of Veterinary Medicine, Department of Basic Medical Sciences, Purdue University; West Lafayette Indiana
| |
Collapse
|
171
|
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
|
172
|
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
|
173
|
White EE, Pai A, Weng Y, Suresh AK, Van Haute D, Pailevanian T, Alizadeh D, Hajimiri A, Badie B, Berlin JM. Functionalized iron oxide nanoparticles for controlling the movement of immune cells. NANOSCALE 2015; 7:7780-9. [PMID: 25848983 PMCID: PMC4409571 DOI: 10.1039/c3nr04421a] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Immunotherapy is currently being investigated for the treatment of many diseases, including cancer. The ability to control the location of immune cells during or following activation would represent a powerful new technique for this field. Targeted magnetic delivery is emerging as a technique for controlling cell movement and localization. Here we show that this technique can be extended to microglia, the primary phagocytic immune cells in the central nervous system. The magnetized microglia were generated by loading the cells with iron oxide nanoparticles functionalized with CpG oligonucleotides, serving as a proof of principle that nanoparticles can be used to both deliver an immunostimulatory cargo to cells and to control the movement of the cells. The nanoparticle-oligonucleotide conjugates are efficiently internalized, non-toxic, and immunostimulatory. We demonstrate that the in vitro migration of the adherent, loaded microglia can be controlled by an external magnetic field and that magnetically-induced migration is non-cytotoxic. In order to capture video of this magnetically-induced migration of loaded cells, a novel 3D-printed "cell box" was designed to facilitate our imaging application. Analysis of cell movement velocities clearly demonstrate increased cell velocities toward the magnet. These studies represent the initial step towards our final goal of using nanoparticles to both activate immune cells and to control their trafficking within the diseased brain.
Collapse
Affiliation(s)
- Ethan E White
- Department of Molecular Medicine, 1500 East Duarte Road, Duarte, CA, 91010, United States
- Irell & Manella Graduate School of Biological Sciences at City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, United States
| | - Alex Pai
- Department of Electrical Engineering, California Institute of Technology, 1200 E. California Blvd., Pasadena, CA 91125, United States
| | - Yiming Weng
- Department of Molecular Medicine, 1500 East Duarte Road, Duarte, CA, 91010, United States
| | - Anil K. Suresh
- Department of Molecular Medicine, 1500 East Duarte Road, Duarte, CA, 91010, United States
| | - Desiree Van Haute
- Department of Molecular Medicine, 1500 East Duarte Road, Duarte, CA, 91010, United States
- Irell & Manella Graduate School of Biological Sciences at City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, United States
| | - Torkom Pailevanian
- Department of Electrical Engineering, California Institute of Technology, 1200 E. California Blvd., Pasadena, CA 91125, United States
| | - Darya Alizadeh
- Department of Molecular Medicine, 1500 East Duarte Road, Duarte, CA, 91010, United States
- Division of Neurosurgery, Department of Surgery, Beckman Research Institute, 1500 East Duarte Road, Duarte, CA, 91010, United States
| | - Ali Hajimiri
- Department of Electrical Engineering, California Institute of Technology, 1200 E. California Blvd., Pasadena, CA 91125, United States
- Drs. Hajimiri, Badie, and Berlin, served as co-PI’s for these studies. Contact info: . Tel.: +1 626 256 4673, . Tel.: +1 626 256 4673. . Tel.: +1 626 395 2312
| | - Behnam Badie
- Division of Neurosurgery, Department of Surgery, Beckman Research Institute, 1500 East Duarte Road, Duarte, CA, 91010, United States
- Drs. Hajimiri, Badie, and Berlin, served as co-PI’s for these studies. Contact info: . Tel.: +1 626 256 4673, . Tel.: +1 626 256 4673. . Tel.: +1 626 395 2312
| | - Jacob M. Berlin
- Department of Molecular Medicine, 1500 East Duarte Road, Duarte, CA, 91010, United States
- Drs. Hajimiri, Badie, and Berlin, served as co-PI’s for these studies. Contact info: . Tel.: +1 626 256 4673, . Tel.: +1 626 256 4673. . Tel.: +1 626 395 2312
| |
Collapse
|
174
|
Li L, Feng J, Fan Y, Tang B. Simultaneous Imaging of Zn2+ and Cu2+ in Living Cells Based on DNAzyme Modified Gold Nanoparticle. Anal Chem 2015; 87:4829-35. [DOI: 10.1021/acs.analchem.5b00204] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Lu Li
- College of Chemistry,
Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry of Education,
Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, P. R. China
| | - Jie Feng
- College of Chemistry,
Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry of Education,
Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, P. R. China
| | - Yuanyuan Fan
- College of Chemistry,
Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry of Education,
Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, P. R. China
| | - Bo Tang
- College of Chemistry,
Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry of Education,
Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan 250014, P. R. China
| |
Collapse
|
175
|
Rotz MW, Culver KSB, Parigi G, MacRenaris KW, Luchinat C, Odom TW, Meade TJ. High relaxivity Gd(III)-DNA gold nanostars: investigation of shape effects on proton relaxation. ACS NANO 2015; 9:3385-96. [PMID: 25723190 PMCID: PMC4489565 DOI: 10.1021/nn5070953] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Gadolinium(III) nanoconjugate contrast agents (CAs) have distinct advantages over their small-molecule counterparts in magnetic resonance imaging. In addition to increased Gd(III) payload, a significant improvement in proton relaxation efficiency, or relaxivity (r1), is often observed. In this work, we describe the synthesis and characterization of a nanoconjugate CA created by covalent attachment of Gd(III) to thiolated DNA (Gd(III)-DNA), followed by surface conjugation onto gold nanostars (DNA-Gd@stars). These conjugates exhibit remarkable r1 with values up to 98 mM(-1) s(-1). Additionally, DNA-Gd@stars show efficient Gd(III) delivery and biocompatibility in vitro and generate significant contrast enhancement when imaged at 7 T. Using nuclear magnetic relaxation dispersion analysis, we attribute the high performance of the DNA-Gd@stars to an increased contribution of second-sphere relaxivity compared to that of spherical CA equivalents (DNA-Gd@spheres). Importantly, the surface of the gold nanostar contains Gd(III)-DNA in regions of positive, negative, and neutral curvature. We hypothesize that the proton relaxation enhancement observed results from the presence of a unique hydrophilic environment produced by Gd(III)-DNA in these regions, which allows second-sphere water molecules to remain adjacent to Gd(III) ions for up to 10 times longer than diffusion. These results establish that particle shape and second-sphere relaxivity are important considerations in the design of Gd(III) nanoconjugate CAs.
Collapse
Affiliation(s)
- Matthew W. Rotz
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Kayla S. B. Culver
- Departments of Chemistry, Materials Science and Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Giacomo Parigi
- Magnetic Resonance Center (CERM) and Department of Chemistry, University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Keith W. MacRenaris
- Quantitative Bio-elemental Imaging Center, Department of Molecular Biosciences, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Claudio Luchinat
- Magnetic Resonance Center (CERM) and Department of Chemistry, University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Teri W. Odom
- Departments of Chemistry, Materials Science and Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Thomas J. Meade
- Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
176
|
Murugan K, Choonara YE, Kumar P, Bijukumar D, du Toit LC, Pillay V. Parameters and characteristics governing cellular internalization and trans-barrier trafficking of nanostructures. Int J Nanomedicine 2015; 10:2191-206. [PMID: 25834433 PMCID: PMC4370919 DOI: 10.2147/ijn.s75615] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Cellular internalization and trans-barrier transport of nanoparticles can be manipulated on the basis of the physicochemical and mechanical characteristics of nanoparticles. Research has shown that these factors significantly influence the uptake of nanoparticles. Dictating these characteristics allows for the control of the rate and extent of cellular uptake, as well as delivering the drug-loaded nanosystem intra-cellularly, which is imperative for drugs that require a specific cellular level to exert their effects. Additionally, physicochemical characteristics of the nanoparticles should be optimal for the nanosystem to bypass the natural restricting phenomena of the body and act therapeutically at the targeted site. The factors at the focal point of emerging smart nanomedicines include nanoparticle size, surface charge, shape, hydrophobicity, surface chemistry, and even protein and ligand conjugates. Hence, this review discusses the mechanism of internalization of nanoparticles and ideal nanoparticle characteristics that allow them to evade the biological barriers in order to achieve optimal cellular uptake in different organ systems. Identifying these parameters assists with the progression of nanomedicine as an outstanding vector of pharmaceuticals.
Collapse
Affiliation(s)
- Karmani Murugan
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Yahya E Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Divya Bijukumar
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Lisa C du Toit
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Viness Pillay
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
177
|
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
|
178
|
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
|
179
|
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
|
180
|
Helmfors H, Lindberg S, Langel Ü. SCARA Involvement in the Uptake of Nanoparticles Formed by Cell-Penetrating Peptides. Methods Mol Biol 2015. [PMID: 26202269 DOI: 10.1007/978-1-4939-2806-4_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The investigation of uptake mechanisms for cell-penetrating peptides (CPPs) is and has been an ongoing project for as long as the peptides have been known, a time period that now spans over two decades. The ultimate answer is yet to be revealed and the current understanding is that no "one" mechanism will ever be found. The reason for this is that the uptake mechanism seems to be dependent on a multitude of factors that include which CPP, what cells are used, whether or not there is cargo and what the cargo is. CPPs are capable of delivering a variety of bio-macromolecules that are by themselves unable to enter into cells. Our group has reported on many different peptides in recent years, many aimed at delivering various oligonucleotide-based cargoes. These peptides have utilized the inherent positive charge of the peptides and some rationally designed modifications to non-covalently complex oligonucleotides and bring them into cells. In this chapter, we present a brief overview of the current proposals for the uptake mechanisms of CPPs and describe methods for detecting and evaluating the role of scavenger receptor class A receptors in the uptake of non-covalent cell-penetrating peptide:oligonucleotide complexes.
Collapse
Affiliation(s)
- Henrik Helmfors
- Department of Neurochemistry, Stockholm University, Stockholm, 10691, Sweden,
| | | | | |
Collapse
|
181
|
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
|
182
|
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
|
183
|
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
|
184
|
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
|
185
|
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]
|
186
|
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]
|
187
|
Cioran AM, Teixidor F, Krpetić Ž, Brust M, Viñas C. Preparation and characterization of Au nanoparticles capped with mercaptocarboranyl clusters. Dalton Trans 2014; 43:5054-61. [PMID: 24301037 DOI: 10.1039/c3dt52872c] [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
The preparation of 3-4 nm and 10 nm gold nanoparticles capped with neutral carborane-based mercaptocarboranes, via two different preparative routes, is reported. The resulting boron-enriched nanomaterials exhibit complete dispersibility in water, opening the way for the use of these monolayer protected clusters (MPCs) in medical applications, such as boron neutron capture therapy (BNCT). These newly prepared MPCs have been characterized by FTIR, (1)H and (11)B NMR spectroscopy, UV-visible, centrifugal particle sizing (CPS), and, in some cases, inductively coupled plasma atomic emission spectrometry (ICP-AES). Water dispersibility exhibited by these MPCs allowed the study of the cellular uptake by HeLa cells.
Collapse
Affiliation(s)
- Ana M Cioran
- Institut de Ciència de Materials de Barcelona, Campus U.A.B., 08193 Bellaterra, Spain.
| | | | | | | | | |
Collapse
|
188
|
Sun T, Zhang YS, Pang B, Hyun DC, Yang M, Xia Y. Engineered nanoparticles for drug delivery in cancer therapy. Angew Chem Int Ed Engl 2014; 53:12320-64. [PMID: 25294565 DOI: 10.1002/anie.201403036] [Citation(s) in RCA: 744] [Impact Index Per Article: 74.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Indexed: 12/18/2022]
Abstract
In medicine, nanotechnology has sparked a rapidly growing interest as it promises to solve a number of issues associated with conventional therapeutic agents, including their poor water solubility (at least, for most anticancer drugs), lack of targeting capability, nonspecific distribution, systemic toxicity, and low therapeutic index. Over the past several decades, remarkable progress has been made in the development and application of engineered nanoparticles to treat cancer more effectively. For example, therapeutic agents have been integrated with nanoparticles engineered with optimal sizes, shapes, and surface properties to increase their solubility, prolong their circulation half-life, improve their biodistribution, and reduce their immunogenicity. Nanoparticles and their payloads have also been favorably delivered into tumors by taking advantage of the pathophysiological conditions, such as the enhanced permeability and retention effect, and the spatial variations in the pH value. Additionally, targeting ligands (e.g., small organic molecules, peptides, antibodies, and nucleic acids) have been added to the surface of nanoparticles to specifically target cancerous cells through selective binding to the receptors overexpressed on their surface. Furthermore, it has been demonstrated that multiple types of therapeutic drugs and/or diagnostic agents (e.g., contrast agents) could be delivered through the same carrier to enable combination therapy with a potential to overcome multidrug resistance, and real-time readout on the treatment efficacy. It is anticipated that precisely engineered nanoparticles will emerge as the next-generation platform for cancer therapy and many other biomedical applications.
Collapse
Affiliation(s)
- Tianmeng Sun
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332 (USA)
| | | | | | | | | | | |
Collapse
|
189
|
Sun T, Zhang YS, Pang B, Hyun DC, Yang M, Xia Y. Maßgeschneiderte Nanopartikel für den Wirkstofftransport in der Krebstherapie. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201403036] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
190
|
Brun E, Sicard-Roselli C. Could nanoparticle corona characterization help for biological consequence prediction? Cancer Nanotechnol 2014; 5:7. [PMID: 25309635 PMCID: PMC4181791 DOI: 10.1186/s12645-014-0007-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 09/11/2014] [Indexed: 12/11/2022] Open
Abstract
As soon as they enter a biological medium (cell culture medium for in vitro, blood or plasma for in vivo studies), nanoparticles, in most cases, see their surface covered by biomolecules, especially proteins. What the cells see is thus not the ideal nanoparticle concocted by chemists, meaning the biomolecular corona could have great biological and physiological repercussions, sometimes masking the expected effects of purposely grafted molecules. In this review, we will mainly focus on gold nanoparticles. In the first part, we will discuss the fate of these particles once in a biological medium, especially in terms of size, and the protein composition of the corona. We will highlight the parameters influencing the quantity and the identity of the adsorbed proteins. In a second part, we will resume the main findings about the influence of a biomolecular corona on cellular uptake, toxicity, biodistribution and targeting ability. To be noticed is the need for standardized experiments and very precise reports of the protocols and methods used in the experimental sections to extract informative data. Given the biological consequences of this corona, we suggest that it should be taken into account in theoretical studies dealing with nanomaterials to better represent the biological environment.
Collapse
Affiliation(s)
- Emilie Brun
- Laboratoire de Chimie Physique, CNRS UMR8000, Université Paris-Sud, 91405 Orsay, Cedex France
| | - Cécile Sicard-Roselli
- Laboratoire de Chimie Physique, CNRS UMR8000, Université Paris-Sud, 91405 Orsay, Cedex France
| |
Collapse
|
191
|
Cao X, Chen C, Yu H, Wang P. Horseradish peroxidase-encapsulated chitosan nanoparticles for enzyme-prodrug cancer therapy. Biotechnol Lett 2014; 37:81-8. [DOI: 10.1007/s10529-014-1664-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 09/03/2014] [Indexed: 12/11/2022]
|
192
|
Ashby J, Pan S, Zhong W. Size and surface functionalization of iron oxide nanoparticles influence the composition and dynamic nature of their protein corona. ACS APPLIED MATERIALS & INTERFACES 2014; 6:15412-9. [PMID: 25144382 PMCID: PMC4160264 DOI: 10.1021/am503909q] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Nanoparticles (NPs) adsorb proteins when in the biological matrix, and the resulted protein corona could affect NP-cell interactions. The corona has a dynamic nature with the adsorbed proteins constantly exchanging with the free proteins in the matrix at various rates. The rapidly exchanging proteins compose the soft corona, which responds more dynamically to environment changes than the hard corona established by the ones with slow exchange rates. In the present study, the corona formed on the superparamagnetic iron oxide NPs (SPIONs) in human serum was studied by flow field-flow fractionation and ultracentrifugation, which rapidly differentiated the corona proteins based on their exchange rates. By varying the surface hydrophobicity of the SPIONs with a core size around 10 nm, we found out that, the more hydrophobic surface ligand attracted proteins with higher surface hydrophobicity and formed a more dynamic corona with a larger portion of the involved proteins with fast exchange rates. Increasing the core diameter of the SPIONs but keeping the surface ligand the same could also result in a more dynamic corona. A brief investigation of the effect on the cellular uptake of SPIONs using one selected corona protein, transferrin, was conducted. The result showed that, only the stably bound transferrin could significantly enhance cellular uptake, while transferrin bound in a dynamic nature had negligible impact. Our study has led to a better understanding of the relationship between the particle properties and the dynamic nature of the corona, which can help with design of nanomaterials with higher biocompatibility and higher efficacy in biosystems for biomedical applications.
Collapse
Affiliation(s)
- Jonathan Ashby
- Department of Chemistry and Institute for Integrative Genome Biology, University of California, Riverside, California 92521, United States
| | - Songqin Pan
- Department of Chemistry and Institute for Integrative Genome Biology, University of California, Riverside, California 92521, United States
| | - Wenwan Zhong
- Department of Chemistry and Institute for Integrative Genome Biology, University of California, Riverside, California 92521, United States
- E-mail:
| |
Collapse
|
193
|
Fleischer C, Payne CK. Nanoparticle-cell interactions: molecular structure of the protein corona and cellular outcomes. Acc Chem Res 2014; 47:2651-9. [PMID: 25014679 PMCID: PMC4139184 DOI: 10.1021/ar500190q] [Citation(s) in RCA: 380] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Indexed: 12/14/2022]
Abstract
The use of nanoparticles (NPs) in biology and medicine requires a molecular-level understanding of how NPs interact with cells in a physiological environment. A critical difference between well-controlled in vitro experiments and in vivo applications is the presence of a complex mixture of extracellular proteins. It has been established that extracellular serum proteins present in blood will adsorb onto the surface of NPs, forming a "protein corona". Our goal was to understand how this protein layer affected cellular-level events, including NP binding, internalization, and transport. A combination of microscopy, which provides spatial resolution, and spectroscopy, which provides molecular information, is necessary to probe protein-NP-cell interactions. Initial experiments used a model system composed of polystyrene NPs functionalized with either amine or carboxylate groups to provide a cationic or anionic surface, respectively. Serum proteins adsorb onto the surface of both cationic and anionic NPs, forming a net anionic protein-NP complex. Although these protein-NP complexes have similar diameters and effective surface charges, they show the exact opposite behavior in terms of cellular binding. In the presence of bovine serum albumin (BSA), the cellular binding of BSA-NP complexes formed from cationic NPs is enhanced, whereas the cellular binding of BSA-NP complexes formed from anionic NPs is inhibited. These trends are independent of NP diameter or cell type. Similar results were obtained for anionic quantum dots and colloidal gold nanospheres. Using competition assays, we determined that BSA-NP complexes formed from anionic NPs bind to albumin receptors on the cell surface. BSA-NP complexes formed from cationic NPs are redirected to scavenger receptors. The observation that similar NPs with identical protein corona compositions bind to different cellular receptors suggested that a difference in the structure of the adsorbed protein may be responsible for the differences in cellular binding of the protein-NP complexes. Circular dichroism spectroscopy, isothermal titration calorimetry, and fluorescence spectroscopy show that the structure of BSA is altered following incubation with cationic NPs, but not anionic NPs. Single-particle-tracking fluorescence microscopy was used to follow the cellular internalization and transport of protein-NP complexes. The single particle-tracking experiments show that the protein corona remains bound to the NP throughout endocytic uptake and transport. The interaction of protein-NP complexes with cells is a challenging question, as the adsorbed protein corona controls the interaction of the NP with the cell; however, the NP itself alters the structure of the adsorbed protein. A combination of microscopy and spectroscopy is necessary to understand this complex interaction, enabling the rational design of NPs for biological and medical 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
|
194
|
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
|
195
|
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
|
196
|
Meng HM, Zhang X, Lv Y, Zhao Z, Wang NN, Fu T, Fan H, Liang H, Qiu L, Zhu G, Tan W. DNA dendrimer: an efficient nanocarrier of functional nucleic acids for intracellular molecular sensing. ACS NANO 2014; 8:6171-81. [PMID: 24806614 PMCID: PMC4076030 DOI: 10.1021/nn5015962] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Functional nucleic acid (FNA)-based sensing systems have been developed for efficient detection of a wide range of biorelated analytes by employing DNAzymes or aptamers as recognition units. However, their intracellular delivery has always been a concern, mainly in delivery efficiency, kinetics, and the amount of delivered FNAs. Here we report a DNA dendrimer scaffold as an efficient nanocarrier to deliver FNAs and to conduct in situ monitoring of biological molecules in living cells. A histidine-dependent DNAzyme and an anti-ATP aptamer were chosen separately as the model FNAs to make the FNA dendrimer. The FNA-embedded DNA dendrimers maintained the catalytic activity of the DNAzyme or the aptamer recognition function toward ATP in the cellular environment, with no change in sensitivity or specificity. Moreover, these DNA dendrimeric nanocarriers show excellent biocompatibility, high intracellular delivery efficiency, and sufficient stability in a cellular environment. This FNA dendrimeric nanocarrier may find a broad spectrum of applications in biomedical diagnosis and therapy.
Collapse
Affiliation(s)
- Hong-Min Meng
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University, Changsha 410082, China
| | - Xiaobing Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University, Changsha 410082, China
- Address correspondence to ,
| | - Yifan Lv
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University, Changsha 410082, China
| | - Zilong Zhao
- Department of Chemistry and Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, Shands Cancer Center, UF Genetics Institute, University of Florida, Gainesville, FL 32611-7200, USA
| | - Nan-Nan Wang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University, Changsha 410082, China
| | - Ting Fu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University, Changsha 410082, China
- Department of Chemistry and Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, Shands Cancer Center, UF Genetics Institute, University of Florida, Gainesville, FL 32611-7200, USA
| | - Huanhuan Fan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University, Changsha 410082, China
| | - Hao Liang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University, Changsha 410082, China
| | - Liping Qiu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University, Changsha 410082, China
- Department of Chemistry and Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, Shands Cancer Center, UF Genetics Institute, University of Florida, Gainesville, FL 32611-7200, USA
| | - Guizhi Zhu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University, Changsha 410082, China
- Department of Chemistry and Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, Shands Cancer Center, UF Genetics Institute, University of Florida, Gainesville, FL 32611-7200, USA
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Collaborative Research Center of Molecular Engineering for Theranostics, Hunan University, Changsha 410082, China
- Department of Chemistry and Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, Shands Cancer Center, UF Genetics Institute, University of Florida, Gainesville, FL 32611-7200, USA
- Address correspondence to ,
| |
Collapse
|
197
|
Son S, Nam J, Kim J, Kim S, Kim WJ. i-motif-driven Au nanomachines in programmed siRNA delivery for gene-silencing and photothermal ablation. ACS NANO 2014; 8:5574-5584. [PMID: 24869928 DOI: 10.1021/nn5022567] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The present work illustrates unique design, construction and operation of an i-motif-based DNA nanomachine templated on gold nanoparticles (AuNPs), which utilizes pH-responsive dynamic motion of i-motif DNA strands and aggregational behavior of AuNPs to elicit programmed delivery of therapeutic siRNA. The pH-sensitive nucleic acids immobilized on the AuNPs consisted of three functional segments, i.e., an i-motif DNA, an overhanging linker DNA and a therapeutic siRNA. At neutral pH, the i-motif DNA is hybridized with the overhanging linker DNA segment of the therapeutic siRNA. However, in endosomal acidic pH, the i-motif DNA forms interstrand tetraplex, which could induce cluster formation of AuNPs resulting in endosomal escape of AuNP clusters, and produce a high gene silencing efficiency by releasing siRNA in the cytosol. Furthermore, the cluster formation of AuNPs accelerated photothermal ablation of cells when irradiated with laser. Precise and synchronized biomechanical motion in subcellular microenvironment is realized through judicious integration of pH-responsive behavior of the i-motif DNA and AuNPs, and meticulous designing of DNA.
Collapse
Affiliation(s)
- Sejin Son
- Center for Self-Assembly and Complexity, Institute for Basic Science (IBS) , Pohang, 790-784, Korea
| | | | | | | | | |
Collapse
|
198
|
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
|
199
|
|
200
|
Wen Y, Meng WS. Recent In Vivo Evidences of Particle-Based Delivery of Small-Interfering RNA (siRNA) into Solid Tumors. J Pharm Innov 2014; 9:158-173. [PMID: 25221632 PMCID: PMC4161233 DOI: 10.1007/s12247-014-9183-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Small-interfering RNA (siRNA) is both a powerful tool in research and a promising therapeutic platform to modulate expression of disease-related genes. Malignant tumors are attractive disease targets for nucleic acid-based therapies. siRNA directed against oncogenes, and genes driving metastases or angiogenesis have been evaluated in animal models and in some cases, in humans. The outcomes of these studies indicate that drug delivery is a significant limiting factor. This review provides perspectives on in vivo validated nanoparticle-based siRNA delivery systems. Results of recent advances in liposomes and polymeric and inorganic formulations illustrate the need for mutually optimized attributes for performance in systemic circulation, tumor interstitial space, plasma membrane, and endosomes. Physiochemical properties conducive to efficient siRNA delivery are summarized and directions for future research are discussed.
Collapse
Affiliation(s)
- Yi Wen
- Division of Pharmaceutical Sciences, Duquesne University, 600, Forbes Avenue, Pittsburgh, PA 15282, USA
| | - Wilson S. Meng
- Division of Pharmaceutical Sciences, Duquesne University, 600, Forbes Avenue, Pittsburgh, PA 15282, USA
| |
Collapse
|