201
|
Cell-penetrating peptide-based non-invasive topical delivery systems. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2017. [DOI: 10.1007/s40005-017-0373-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
202
|
Learte-Aymamí S, Curado N, Rodríguez J, Vázquez ME, Mascareñas JL. Metal-Dependent DNA Recognition and Cell Internalization of Designed, Basic Peptides. J Am Chem Soc 2017; 139:16188-16193. [PMID: 29056048 PMCID: PMC5741177 DOI: 10.1021/jacs.7b07422] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Indexed: 12/18/2022]
Abstract
A fragment of the DNA basic region (br) of the GCN4 bZIP transcription factor has been modified to include two His residues at designed i and i+4 positions of its N-terminus. The resulting monomeric peptide (brHis2) does not bind to its consensus target DNA site (5'-GTCAT-3'). However, addition of Pd(en)Cl2 (en, ethylenediamine) promotes a high-affinity interaction with exquisite selectivity for this sequence. The peptide-DNA complex is disassembled by addition of a slight excess of a palladium chelator, and the interaction can be reversibly switched multiple times by playing with controlled amounts of either the metal complex or the chelator. Importantly, while the peptide brHis2 fails to translocate across cell membranes on its own, addition of the palladium reagent induces an efficient cell internalization of this peptide. In short, we report (1) a designed, short peptide that displays highly selective, major groove DNA binding, (2) a reversible, metal-dependent DNA interaction, and (3) a metal-promoted cell internalization of this basic peptide.
Collapse
Affiliation(s)
- Soraya Learte-Aymamí
- Centro Singular de Investigación
en Química Biolóxica e Materiais Moleculares (CIQUS)
and Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Natalia Curado
- Centro Singular de Investigación
en Química Biolóxica e Materiais Moleculares (CIQUS)
and Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Jéssica Rodríguez
- Centro Singular de Investigación
en Química Biolóxica e Materiais Moleculares (CIQUS)
and Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - M. Eugenio Vázquez
- Centro Singular de Investigación
en Química Biolóxica e Materiais Moleculares (CIQUS)
and Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - José L. Mascareñas
- Centro Singular de Investigación
en Química Biolóxica e Materiais Moleculares (CIQUS)
and Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
203
|
Ulasov AV, Rosenkranz AA, Sobolev AS. Transcription factors: Time to deliver. J Control Release 2017; 269:24-35. [PMID: 29113792 DOI: 10.1016/j.jconrel.2017.11.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/02/2017] [Accepted: 11/03/2017] [Indexed: 12/17/2022]
Abstract
Transcription factors (TFs) are at the center of the broad regulatory network orchestrating gene expression programs that elicit different biological responses. For a long time, TFs have been considered as potent drug targets due to their implications in the pathogenesis of a variety of diseases. At the same time, TFs, located at convergence points of cellular regulatory pathways, are powerful tools providing opportunities both for cell type change and for managing the state of cells. This task formulation requires the TF modulation problem to come to the fore. We review several ways to manage TF activity (small molecules, transfection, nanocarriers, protein-based approaches), analyzing their limitations and the possibilities to overcome them. Delivery of TFs could revolutionize the biomedical field. Whether this forecast comes true will depend on the ability to develop convenient technologies for targeted delivery of TFs.
Collapse
Affiliation(s)
- Alexey V Ulasov
- Department of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
| | - Andrey A Rosenkranz
- Department of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia; Faculty of Biology, Moscow State University, 1-12 Leninskiye Gory St., 119234 Moscow, Russia
| | - Alexander S Sobolev
- Department of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia; Faculty of Biology, Moscow State University, 1-12 Leninskiye Gory St., 119234 Moscow, Russia.
| |
Collapse
|
204
|
Verdurmen WPR, Mazlami M, Plückthun A. A quantitative comparison of cytosolic delivery via different protein uptake systems. Sci Rep 2017; 7:13194. [PMID: 29038564 PMCID: PMC5643320 DOI: 10.1038/s41598-017-13469-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 09/25/2017] [Indexed: 01/27/2023] Open
Abstract
Over many years, a variety of delivery systems have been investigated that have the capacity to shuttle macromolecular cargoes, especially proteins, into the cytosol. Due to the lack of an objective way to quantify cytosolic delivery, relative delivery efficiencies of the various transport systems have remained unclear. Here, we demonstrate the use of the biotin ligase assay for a quantitative comparison of protein transport to the cytosol via cell-penetrating peptides, supercharged proteins and bacterial toxins in four different cell lines. The data illustrate large differences in both the total cellular internalization, which denotes any intracellular location including endosomes, and in the cytosolic uptake of the transport systems, with little correlation between the two. Also, we found significant differences between the cell lines. In general, protein transport systems based on cell-penetrating peptides show a modest total uptake, and mostly do not deliver cargo to the cytosol. Systems based on bacterial toxins show a modest receptor-mediated internalization but an efficient delivery to the cytosol. Supercharged proteins, on the contrary, are not receptor-specific and lead to massive total internalization into endosomes, but only low amounts end up in the cytosol.
Collapse
Affiliation(s)
- Wouter P R Verdurmen
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057, Zurich, Switzerland.,Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud university medical center, Geert Grooteplein 28, 6525 GA, Nijmegen, The Netherlands
| | - Marigona Mazlami
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057, Zurich, Switzerland
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, 8057, Zurich, Switzerland.
| |
Collapse
|
205
|
LeCher JC, Nowak SJ, McMurry JL. Breaking in and busting out: cell-penetrating peptides and the endosomal escape problem. Biomol Concepts 2017; 8:131-141. [PMID: 28841567 PMCID: PMC5640260 DOI: 10.1515/bmc-2017-0023] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 12/27/2022] Open
Abstract
Cell-penetrating peptides (CPPs) have long held great promise for the manipulation of living cells for therapeutic and research purposes. They allow a wide array of biomolecules from large, oligomeric proteins to nucleic acids and small molecules to rapidly and efficiently traverse cytoplasmic membranes. With few exceptions, if a molecule can be associated with a CPP, it can be delivered into a cell. However, a growing realization in the field is that CPP-cargo fusions largely remain trapped in endosomes and are eventually targeted for degradation or recycling rather than released into the cytoplasm or trafficked to a desired subcellular destination. This 'endosomal escape problem' has confounded efforts to develop CPP-based delivery methods for drugs, enzymes, plasmids, etc. This review provides a brief history of CPP research and discusses current issues in the field with a primary focus on the endosomal escape problem, for which several promising potential solutions have been developed. Are we on the verge of developing technologies to deliver therapeutics such as siRNA, CRISPR/Cas complexes and others that are currently failing because of an inability to get into cells, or are we just chasing after another promising but unworkable technology? We make the case for optimism.
Collapse
Affiliation(s)
- Julia C. LeCher
- Department of Molecular & Cellular Biology, Kennesaw State University, 370 Paulding Ave NW, MD 1201, Kennesaw, GA 30144, USA
| | - Scott J. Nowak
- Department of Molecular & Cellular Biology, Kennesaw State University, 370 Paulding Ave NW, MD 1201, Kennesaw, GA 30144, USA
| | - Jonathan L. McMurry
- Department of Molecular & Cellular Biology, Kennesaw State University, 370 Paulding Ave NW, MD 1201, Kennesaw, GA 30144, USA
| |
Collapse
|
206
|
Tawiah KD, Porciani D, Burke DH. Toward the Selection of Cell Targeting Aptamers with Extended Biological Functionalities to Facilitate Endosomal Escape of Cargoes. Biomedicines 2017; 5:biomedicines5030051. [PMID: 28837119 PMCID: PMC5618309 DOI: 10.3390/biomedicines5030051] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 08/19/2017] [Accepted: 08/19/2017] [Indexed: 12/26/2022] Open
Abstract
Over the past decades there have been exciting and rapid developments of highly specific molecules to bind cancer antigens that are overexpressed on the surfaces of malignant cells. Nanomedicine aims to exploit these ligands to generate nanoscale platforms for targeted cancer therapy, and to do so with negligible off-target effects. Aptamers are structured nucleic acids that bind to defined molecular targets ranging from small molecules and proteins to whole cells or viruses. They are selected through an iterative process of amplification and enrichment called SELEX (systematic evolution of ligands by exponential enrichment), in which a combinatorial oligonucleotide library is exposed to the target of interest for several repetitive rounds. Nucleic acid ligands able to bind and internalize into malignant cells have been extensively used as tools for targeted delivery of therapeutic payloads both in vitro and in vivo. However, current cell targeting aptamer platforms suffer from limitations that have slowed their translation to the clinic. This is especially true for applications in which the cargo must reach the cytosol to exert its biological activity, as only a small percentage of the endocytosed cargo is typically able to translocate into the cytosol. Innovative technologies and selection strategies are required to enhance cytoplasmic delivery. In this review, we describe current selection methods used to generate aptamers that target cancer cells, and we highlight some of the factors that affect productive endosomal escape of cargoes. We also give an overview of the most promising strategies utilized to improve and monitor endosomal escape of therapeutic cargoes. The methods we highlight exploit tools and technologies that can potentially be incorporated in the SELEX process. Innovative selection protocols may identify aptamers with extended biological functionalities that allow effective cytosolic translocation of therapeutics. This in turn may facilitate successful translation of these platforms into clinical applications.
Collapse
Affiliation(s)
- Kwaku D Tawiah
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA.
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA.
| | - David Porciani
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA.
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA.
- Department of Molecular Microbiology & Immunology, University of Missouri, Columbia, MO 65212, USA.
| | - Donald H Burke
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA.
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA.
- Department of Molecular Microbiology & Immunology, University of Missouri, Columbia, MO 65212, USA.
- Department of Bioengineering, University of Missouri, Columbia, MO 65211, USA.
| |
Collapse
|
207
|
Bruce VJ, McNaughton BR. Inside Job: Methods for Delivering Proteins to the Interior of Mammalian Cells. Cell Chem Biol 2017; 24:924-934. [DOI: 10.1016/j.chembiol.2017.06.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 06/19/2017] [Accepted: 06/27/2017] [Indexed: 10/19/2022]
|
208
|
Targeted release of transcription factors for human cell reprogramming by ZEBRA cell-penetrating peptide. Int J Pharm 2017. [DOI: 10.1016/j.ijpharm.2017.06.073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
209
|
Breger JC, Muttenthaler M, Delehanty JB, Thompson DA, Oh E, Susumu K, Deschamps JR, Anderson GP, Field LD, Walper SA, Dawson PE, Medintz IL. Nanoparticle cellular uptake by dendritic wedge peptides: achieving single peptide facilitated delivery. NANOSCALE 2017; 9:10447-10464. [PMID: 28703833 DOI: 10.1039/c7nr03362a] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Significant efforts are being undertaken to optimize the cargo carrying capacity and especially the cellular delivery efficiency of functionalized nanoparticles for applications in biological research and pharmacological delivery. One approach to increasing nanoparticle surface cargo display capacity is to decrease the number of moieties required for mediating cellular delivery by improving their efficiency. We describe a series of multivalent cell penetrating peptide (CPP) dendrimers that facilitate rapid cellular delivery of prototypical nanoparticle-semiconductor quantum dots (QDs). The modular CPP dendrimers were assembled through an innovative convergent oxime ligation strategy between (Arg9)n motifs and a dendritic QD-coordination scaffold. Dendrimeric peptides sequentially incorporate a terminal (His)6 motif for metal-affinity QD coordination, a Pro9 spacer, a branching poly-lysine scaffold, and wedged display of (Arg9)n binding motifs with n = 1×, 2×, 4×, 8×, 16× multivalency. QD dendrimer display capacity was estimated using structural simulations and QD-(Arg9)1-16 conjugates characterized by dynamic light scattering along with surface plasmon resonance-based binding assays to heparan sulfate proteoglycan surfaces. Cellular uptake via endocytosis was confirmed and peptide delivery kinetics investigated as a function of QD-(Arg9)1-16 conjugate exposure time and QD assembly ratio where cellular viability assays reflected no overt cytotoxicity. The ability of single dendrimer conjugates to facilitate cellular uptake was confirmed for QD-(Arg9)2-16 repeats along with the ability to deliver >850 kDa of protein cargo per QD. Minimizing the number of CPPs required for cellular uptake is critical for expanding nanoparticle cargo carrying capacity and can allow for inclusion of additional sensors, therapeutics and contrast agents on their surface.
Collapse
Affiliation(s)
- Joyce C Breger
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA. and American Society for Engineering Education, Washington, DC 20036, USA
| | - Markus Muttenthaler
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA and Institute for Molecular Bioscience, The University of Queensland, 4072 St Lucia, Brisbane, Australia
| | - James B Delehanty
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA.
| | - Darren A Thompson
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA and University of Idaho, Coeur d'Alene, ID 83814, USA
| | - Eunkeu Oh
- Optical Sciences Division, Code 5600, U.S. Naval Research Laboratory, Washington, DC 20375, USA and Sotera Defense Solutions, Inc., Columbia, MD 21046, USA
| | - Kimihiro Susumu
- Optical Sciences Division, Code 5600, U.S. Naval Research Laboratory, Washington, DC 20375, USA and Sotera Defense Solutions, Inc., Columbia, MD 21046, USA
| | - Jeffrey R Deschamps
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA.
| | - George P Anderson
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA.
| | - Lauren D Field
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA. and Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Scott A Walper
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA.
| | - Philip E Dawson
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Igor L Medintz
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA.
| |
Collapse
|
210
|
Alexander-Bryant AA, Zhang H, Attaway CC, Pugh W, Eggart L, Sansevere RM, Andino LM, Dinh L, Cantini LP, Jakymiw A. Dual peptide-mediated targeted delivery of bioactive siRNAs to oral cancer cells in vivo. Oral Oncol 2017; 72:123-131. [PMID: 28797448 DOI: 10.1016/j.oraloncology.2017.07.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/19/2017] [Accepted: 07/04/2017] [Indexed: 01/27/2023]
Abstract
OBJECTIVES Despite significant advances in cancer treatment, the prognosis for oral cancer remains poor in comparison to other cancer types, including breast, skin, and prostate. As a result, more effective therapeutic modalities are needed for the treatment of oral cancer. Consequently, in the present study, we examined the feasibility of using a dual peptide carrier approach, combining an epidermal growth factor receptor (EGFR)-targeting peptide with an endosome-disruptive peptide, to mediate targeted delivery of small interfering RNAs (siRNAs) into EGFR-overexpressing oral cancer cells and induce silencing of the targeted oncogene, cancerous inhibitor of protein phosphatase 2A (CIP2A). MATERIALS AND METHODS Fluorescence microscopy, real-time PCR, Western blot analysis, and in vivo bioimaging of mice containing orthotopic xenograft tumors were used to examine the ability of the dual peptide carrier to mediate specific delivery of bioactive siRNAs into EGFR-overexpressing oral cancer cells/tissues. RESULTS Co-complexation of the EGFR-targeting peptide, GE11R9, with the endosome-disruptive 599 peptide facilitated the specific uptake of siRNAs into oral cancer cells overexpressing EGFR in vitro with optimal gene silencing observed at a 60:30:1 (GE11R9:599:siRNA) molar ratio. Furthermore, when administered systemically to mice bearing xenograft oral tumors, this dual peptide complex mediated increased targeted delivery of siRNAs into tumor tissues in comparison to the 599 peptide alone and significantly enhanced CIP2A silencing. CONCLUSION Herein we provide the first report demonstrating the clinical potential of a dual peptide strategy for siRNA-based therapeutics by synergistically mediating the effective targeting and delivery of bioactive siRNAs into EGFR-overexpressing oral cancer cells.
Collapse
Affiliation(s)
- Angela A Alexander-Bryant
- Department of Oral Health Sciences and Center for Oral Health Research, Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA; Department of Bioengineering, Clemson University, Clemson, SC 29634, USA
| | - Haiwen Zhang
- Department of Oral Health Sciences and Center for Oral Health Research, Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA
| | - Christopher C Attaway
- Department of Oral Health Sciences and Center for Oral Health Research, Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA
| | - William Pugh
- Department of Oral Health Sciences and Center for Oral Health Research, Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA
| | - Laurence Eggart
- Department of Oral Health Sciences and Center for Oral Health Research, Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA
| | - Robert M Sansevere
- Department of Oral Health Sciences and Center for Oral Health Research, Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA
| | - Lourdes M Andino
- Department of Oral Health Sciences and Center for Oral Health Research, Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA
| | - Lu Dinh
- Department of Oral Health Sciences and Center for Oral Health Research, Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA
| | - Liliana P Cantini
- Department of Oral Health Sciences and Center for Oral Health Research, Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA
| | - Andrew Jakymiw
- Department of Oral Health Sciences and Center for Oral Health Research, Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA; Department of Bioengineering, Clemson University, Clemson, SC 29634, USA.
| |
Collapse
|
211
|
Dowaidar M, Abdelhamid HN, Hällbrink M, Zou X, Langel Ü. Graphene oxide nanosheets in complex with cell penetrating peptides for oligonucleotides delivery. Biochim Biophys Acta Gen Subj 2017; 1861:2334-2341. [PMID: 28689990 DOI: 10.1016/j.bbagen.2017.07.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/27/2017] [Accepted: 07/04/2017] [Indexed: 12/15/2022]
Abstract
A new strategy for gene transfection using the nanocarrier of cell penetrating peptides (CPPs; PepFect14 (PF14) or PepFect14 (PF14) (PF221)) in complex with graphene oxide (GO) is reported. GO complexed with CPPs and plasmid (pGL3), splice correction oligonucleotides (SCO) or small interfering RNA (siRNA) are performed. Data show adsorption of CPPs and oligonucleotides on the top of the graphenic lamellar without any observed change of the particle size of GO. GO mitigates the cytotoxicity of CPPs and improves the material biocompatibility. Complexes of GO-pGL3-CPPs (CPPs; PF14 or PF221) offer 2.1-2.5 fold increase of the cell transfection compared to pGL3-CPPs (CPPs; PF14 or PF221). GO-SCO-PF14 assemblies effectively transfect the cells with an increase of >10-25 fold compared to the transfection using PF14. The concentration of GO plays a significant role in the material nanotoxicity and the transfection efficiency. The results open a new horizon in the gene treatment using CPPs and offer a simple strategy for further investigations.
Collapse
Affiliation(s)
- Moataz Dowaidar
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, Stockholm SE-10691, Sweden.
| | - Hani Nasser Abdelhamid
- Department of Materials and Environmental Chemistry, Stockholm University, Svante Arrhenius väg 16C, Stockholm SE-106 91, Sweden.
| | - Mattias Hällbrink
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, Stockholm SE-10691, Sweden
| | - Xiaodong Zou
- Department of Materials and Environmental Chemistry, Stockholm University, Svante Arrhenius väg 16C, Stockholm SE-106 91, Sweden
| | - Ülo Langel
- Department of Neurochemistry, Stockholm University, Svante Arrhenius väg 16B, Stockholm SE-10691, Sweden.
| |
Collapse
|
212
|
Kopp M, Kollenda S, Epple M. Nanoparticle-Protein Interactions: Therapeutic Approaches and Supramolecular Chemistry. Acc Chem Res 2017; 50:1383-1390. [PMID: 28480714 DOI: 10.1021/acs.accounts.7b00051] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Research on nanoparticles has evolved into a major topic in chemistry. Concerning biomedical research, nanoparticles have decisively entered the field, creating the area of nanomedicine where nanoparticles are used for drug delivery, imaging, and tumor targeting. Besides these functions, scientists have addressed the specific ways in which nanoparticles interact with biomolecules, with proteins being the most prominent example. Depending on their size, shape, charge, and surface functionality, specifically designed nanoparticles can interact with proteins in a defined way. Proteins have typical dimensions of 5-20 nm. Ultrasmall nanoparticles (size about 1-2 nm) can address specific epitopes on the surface of a protein, for example, an active center of an enzyme. Medium-sized nanoparticles (size about 5 nm) can interact with proteins on a 1:1 basis. Large nanoparticles (above 20 nm) are big in comparison to many proteins and therefore are at the borderline to a two-dimensional surface onto which a protein will adsorb. This can still lead to irreversible structural changes in a protein and a subsequent loss of function. However, as most cells readily take up nanoparticles of almost any size, it is easily possible to use nanoparticles as transporters for proteins into a cell, for example, to address an internal receptor. Much work has been dedicated to this approach, but it is constrained by two processes that can only be observed in living cells or organisms. First, nanoparticles are usually taken up by endocytosis and are delivered into an intracellular endosome. After fusion with a lysosome, a degradation or denaturation of the protein cargo by the acidic environment or by proteases may occur before it can enter the cytoplasm. Second, nanoparticles are rapidly coated with proteins upon contact with biological media like blood. This so-called protein corona influences the contact with other proteins, cells, or tissue and may prevent the desired interaction. Essentially, these effects cannot be understood in purely chemical approaches but require biological environments and systems because the underlying processes are simply too complicated to be modeled in nonbiological systems. The area of nanoparticle-protein interactions strongly relies on different approaches: Synthetic chemistry is involved to prepare, stabilize, and functionalize nanoparticles. High-end analytical chemistry is required to understand the nature of a nanoparticle surface and the steps of its interaction with proteins. Concepts from supramolecular chemistry help to understand the complex noncovalent interactions between the surfaces of proteins and nanoparticles. Protein chemistry and biophysical chemistry are required to understand the behavior of a protein in contact with a nanoparticle. Finally, all chemical concepts must live up to the "biological reality", first in cell culture experiments in vitro and finally in animal or human experiments in vivo, to open new therapies in the 21st century. This interdisciplinary approach makes the field highly exciting but also highly demanding for chemists who, however, have to learn to understand the language of other areas.
Collapse
Affiliation(s)
- Mathis Kopp
- Inorganic Chemistry and Center for Nanointegration
Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45117 Essen, Germany
| | - Sebastian Kollenda
- Inorganic Chemistry and Center for Nanointegration
Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45117 Essen, Germany
| | - Matthias Epple
- Inorganic Chemistry and Center for Nanointegration
Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45117 Essen, Germany
| |
Collapse
|
213
|
Umemoto T, Sakamoto K, Fukuda Y, Adachi Y, Tani A, Asami T. A Glutamic Acid Analog Bearing an Ethylenediamine Moiety Promotes the Cytosolic Delivery of TAT Peptides. CHEM LETT 2017. [DOI: 10.1246/cl.170097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Tadashi Umemoto
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555
| | - Kotaro Sakamoto
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555
| | - Yasunori Fukuda
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555
| | - Yusuke Adachi
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555
| | - Akiyoshi Tani
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555
| | - Taiji Asami
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa 251-8555
| |
Collapse
|
214
|
Liu Y, Xu CF, Iqbal S, Yang XZ, Wang J. Responsive Nanocarriers as an Emerging Platform for Cascaded Delivery of Nucleic Acids to Cancer. Adv Drug Deliv Rev 2017; 115:98-114. [PMID: 28396204 DOI: 10.1016/j.addr.2017.03.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 12/19/2022]
Abstract
Cascades of systemic and intracellular obstacles, including low stability in blood, little tumor accumulation, weak tumor penetration, poor cellular uptake, inefficient endosomal escape and deficient disassembly in the cytoplasm, must be overcome in order to deliver nucleic acid drugs for cancer therapy. Nanocarriers that are sensitive to a variety of physiological stimuli, such as pH, redox status, and cell enzymes, are substantially changing the landscape of nucleic acid drug delivery by helping to overcome cascaded systemic and intracellular barriers. This review discusses nucleic acid-based therapeutics, systemic and intracellular barriers to efficient nucleic acid delivery, and nanocarriers responsive to extracellular and intracellular biological stimuli to overcome individual barriers. In particular, responsive nanocarriers for the cascaded delivery of nucleic acids in vivo are highlighted. Developing novel cascaded nanocarriers that transform their physicochemical properties in response to various stimuli in a timely and spatially controlled manner for nucleic acid drug delivery holds great potential for translating the promise of nucleic acid drugs and achieving clinically successful cancer therapy.
Collapse
|
215
|
Long-term regulation of gene expression in muscle cells by systemically delivered siRNA. J Control Release 2017; 256:101-113. [DOI: 10.1016/j.jconrel.2017.04.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 04/20/2017] [Accepted: 04/25/2017] [Indexed: 12/20/2022]
|
216
|
Cheng Y, Sun C, Ou X, Liu B, Lou X, Xia F. Dual-targeted peptide-conjugated multifunctional fluorescent probe with AIEgen for efficient nucleus-specific imaging and long-term tracing of cancer cells. Chem Sci 2017; 8:4571-4578. [PMID: 28626568 PMCID: PMC5471453 DOI: 10.1039/c7sc00402h] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/19/2017] [Indexed: 12/20/2022] Open
Abstract
Precisely targeted transportation of a long-term tracing regent to a nucleus with low toxicity is one of the most challenging concerns in revealing cancer cell behaviors. Here, we report a dual-targeted peptide-conjugated multifunctional fluorescent probe (cNGR-CPP-NLS-RGD-PyTPE, TCNTP) with aggregation-induced emission (AIE) characteristic, for efficient nucleus-specific imaging and long-term and low-toxicity tracing of cancer cells. TCNTP mainly consists of two components: one is a functionalized combinatorial peptide (TCNT) containing two targeted peptides (cNGR and RGD), a cell-penetrating peptide (CPP) and a nuclear localization signal (NLS), which can specifically bind to a cell surface and effectively enter into the nucleus; the other one is an AIE-active tetraphenylethene derivative (PyTPE, a typical AIEgen) as fluorescence imaging reagent. In the presence of aminopeptidase N (CD13) and integrin αvβ3, TCNTP can specifically bind to both of them using cNGR and RGD, respectively, lighting up its yellow fluorescence. Because it contains CPP, TCNTP can be effectively integrated into the cytoplasm, and then be delivered into the nucleus with the help of NLS. TCNTP exhibited strong fluorescence in the nucleus of CD13 and integrin αvβ3 overexpression cells due to the specific targeting ability, efficient transport capacity and AIE characteristic in a more crowded space. Furthermore, TCNTP can be applied for long-term tracing in living cells, scarcely affecting normal cells with negligible toxicity in more than ten passages.
Collapse
Affiliation(s)
- Yong Cheng
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica , School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology , Wuhan 430074 , P. R. China . ;
- National Engineering Research Center for Nanomedicine , Department of Biomedical Engineering , College of Life Science and Technology , Huazhong University of Science and Technology , Wuhan 430074 , P. R. China
| | - Chunli Sun
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica , School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology , Wuhan 430074 , P. R. China . ;
| | - Xiaowen Ou
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica , School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology , Wuhan 430074 , P. R. China . ;
| | - Bifeng Liu
- National Engineering Research Center for Nanomedicine , Department of Biomedical Engineering , College of Life Science and Technology , Huazhong University of Science and Technology , Wuhan 430074 , P. R. China
| | - Xiaoding Lou
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica , School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology , Wuhan 430074 , P. R. China . ;
| | - Fan Xia
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica , School of Chemistry and Chemical Engineering , Huazhong University of Science and Technology , Wuhan 430074 , P. R. China . ;
- National Engineering Research Center for Nanomedicine , Department of Biomedical Engineering , College of Life Science and Technology , Huazhong University of Science and Technology , Wuhan 430074 , P. R. China
| |
Collapse
|
217
|
Wang TY, Libardo MDJ, Angeles-Boza AM, Pellois JP. Membrane Oxidation in Cell Delivery and Cell Killing Applications. ACS Chem Biol 2017; 12:1170-1182. [PMID: 28355059 DOI: 10.1021/acschembio.7b00237] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell delivery or cell killing processes often involve the crossing or disruption of cellular membranes. We review how, by modifying the composition and properties of membranes, membrane oxidation can be exploited to enhance the delivery of macromolecular cargoes into live human cells. We also describe how membrane oxidation can be utilized to achieve efficient killing of bacteria by antimicrobial peptides. Finally, we present recent evidence highlighting how membrane oxidation is intimately engaged in natural biological processes such as antigen delivery in dendritic cells and in the killing of bacteria by antimicrobial peptides. Overall, the insights that have been recently gained in this area should facilitate the development of more effective delivery technologies and antimicrobial therapeutic approaches.
Collapse
Affiliation(s)
- Ting-Yi Wang
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - M. Daben J. Libardo
- Department
of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Alfredo M. Angeles-Boza
- Department
of Chemistry, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Jean-Philippe Pellois
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
218
|
Peraro L, Zou Z, Makwana KM, Cummings AE, Ball HL, Yu H, Lin YS, Levine B, Kritzer JA. Diversity-Oriented Stapling Yields Intrinsically Cell-Penetrant Inducers of Autophagy. J Am Chem Soc 2017; 139:7792-7802. [PMID: 28414223 PMCID: PMC5473019 DOI: 10.1021/jacs.7b01698] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
![]()
Autophagy
is an essential pathway by which cellular and foreign
material are degraded and recycled in eukaryotic cells. Induction
of autophagy is a promising approach for treating diverse human diseases,
including neurodegenerative disorders and infectious diseases. Here,
we report the use of a diversity-oriented stapling approach to produce
autophagy-inducing peptides that are intrinsically cell-penetrant.
These peptides induce autophagy at micromolar concentrations in vitro,
have aggregate-clearing activity in a cellular model of Huntington’s
disease, and induce autophagy in vivo. Unexpectedly, the solution
structure of the most potent stapled peptide, DD5-o, revealed an α-helical
conformation in methanol, stabilized by an unusual (i,i+3) staple which cross-links two d-amino
acids. We also developed a novel assay for cell penetration that reports
exclusively on cytosolic access and used it to quantitatively compare
the cell penetration of DD5-o and other autophagy-inducing peptides.
These new, cell-penetrant autophagy inducers and their molecular details
are critical advances in the effort to understand and control autophagy.
More broadly, diversity-oriented stapling may provide a promising
alternative to polycationic sequences as a means for rendering peptides
more cell-penetrant.
Collapse
Affiliation(s)
- Leila Peraro
- Department of Chemistry, Tufts University , Medford, Massachusetts 02155, United States
| | | | - Kamlesh M Makwana
- Department of Chemistry, Tufts University , Medford, Massachusetts 02155, United States
| | - Ashleigh E Cummings
- Department of Chemistry, Tufts University , Medford, Massachusetts 02155, United States
| | | | - Hongtao Yu
- Department of Chemistry, Tufts University , Medford, Massachusetts 02155, United States
| | - Yu-Shan Lin
- Department of Chemistry, Tufts University , Medford, Massachusetts 02155, United States
| | | | - Joshua A Kritzer
- Department of Chemistry, Tufts University , Medford, Massachusetts 02155, United States
| |
Collapse
|
219
|
Gillet FX, Garcia RA, Macedo LLP, Albuquerque EVS, Silva MCM, Grossi-de-Sa MF. Investigating Engineered Ribonucleoprotein Particles to Improve Oral RNAi Delivery in Crop Insect Pests. Front Physiol 2017; 8:256. [PMID: 28503153 PMCID: PMC5408074 DOI: 10.3389/fphys.2017.00256] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/10/2017] [Indexed: 01/15/2023] Open
Abstract
Genetically modified (GM) crops producing double-stranded RNAs (dsRNAs) are being investigated largely as an RNA interference (RNAi)-based resistance strategy against crop insect pests. However, limitations of this strategy include the sensitivity of dsRNA to insect gut nucleases and its poor insect cell membrane penetration. Working with the insect pest cotton boll weevil (Anthonomus grandis), we showed that the chimeric protein PTD-DRBD (peptide transduction domain—dsRNA binding domain) combined with dsRNA forms a ribonucleoprotein particle (RNP) that improves the effectiveness of the RNAi mechanism in the insect. The RNP slows down nuclease activity, probably by masking the dsRNA. Furthermore, PTD-mediated internalization in insect gut cells is achieved within minutes after plasma membrane contact, limiting the exposure time of the RNPs to gut nucleases. Therefore, the RNP provides an approximately 2-fold increase in the efficiency of insect gene silencing upon oral delivery when compared to naked dsRNA. Taken together, these data demonstrate the role of engineered RNPs in improving dsRNA stability and cellular entry, representing a path toward the design of enhanced RNAi strategies in GM plants against crop insect pests.
Collapse
Affiliation(s)
| | - Rayssa A Garcia
- Embrapa Genetic Resources and BiotechnologyBrasília, Brazil.,Department of Cellular Biology, Brasilia Federal University (UnB)Brasília, Brazil
| | | | | | | | - Maria F Grossi-de-Sa
- Embrapa Genetic Resources and BiotechnologyBrasília, Brazil.,Graduate Program in Genomics and Biotechnology, Catholic University of BrasiliaBrasilia, Brazil
| |
Collapse
|
220
|
Effect of size and pegylation of liposomes and peptide-based synthetic lipoproteins on tumor targeting. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:1869-1878. [PMID: 28434931 DOI: 10.1016/j.nano.2017.04.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 03/28/2017] [Accepted: 04/10/2017] [Indexed: 12/31/2022]
Abstract
Synthetic high-density lipoprotein nanoparticles (sHDL) are a valuable class of nanomedicines with established animal safety profile, clinical tolerability and therapeutic efficacy for cardiovascular applications. In this study we examined how the scavenger receptor B-I-mediated (SR-BI) tumor-targeting ability of sHDL, long plasma circulation half-life, and small particle size (9.6±0.2nm) impacted sHDL accumulation in SR-BI positive colorectal carcinoma cells, 3D tumor spheroids, and in vivo xenografts. We compared tumor accumulation of sHDL with that of liposomes (LIP, 130.7±0.8nm), pegylated liposomes (PEG-LIP, 101±2nm), and pegylated sHDL (12.1±0.1nm), all prepared with the same lipid components. sHDL penetrated deep (210μm) into tumor spheroids and exhibited 12- and 3-fold higher in vivo solid tumor accumulation, compared with LIP (p<0.01) and PEG-LIP (p<0.05), respectively. These results suggest that sHDL with established human safety possess promising intrinsic tumor-targeted properties.
Collapse
|
221
|
Wang Y, Li J, Chen JJ, Gao X, Huang Z, Shen Q. Multifunctional Nanoparticles Loading with Docetaxel and GDC0941 for Reversing Multidrug Resistance Mediated by PI3K/Akt Signal Pathway. Mol Pharm 2017; 14:1120-1132. [PMID: 28291364 DOI: 10.1021/acs.molpharmaceut.6b01045] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The polylactic-co-glycolic acid polyethylene glycol conjugated with cell penetrating peptide R7 (PLGA-PEG-R7)/polysulfadimethoxine-folate nanoparticles loaded with docetaxel (DTX) and GDC0941 (R7/PSD-Fol NPs) were prepared to overcome multidrug resistance (MDR) and enhance the antitumor activity. First, polysulfadimethoxine-folate was synthesized to construct the R7/PSD-Fol NPs. The R7/PSD-Fol NPs were prepared with the abilities of effective entrapment and drug loading. Due to the pH-sensitive effect of PSD-folate, the releasing of DTX and GDC0941 from the R7/PSD-Fol NPs was lower in pH 7.4 buffer solution than that in pH 5.0 buffer solution. The half maximal inhibitory concentration (IC50) of MCF-7 and resistant to doxorubicin (MCF-7/Adr) cells illustrated the cytotoxicity of R7/PSD-Fol nanoparticles by using the MTT method. The uptake of R7/PSD-Fol NPs was visualized by using the fluorescence of Rh-123 to detect the targeting effect of folate on the surface of R7/PSD-Fol NPs. The results of the cell apoptosis and the depolarization of mitochondrial membrane potential (MMP) were adopted to show the cytotoxicity of the R7/PSD-Fol NPs on MCF-7/Adr cells. The Western blot revealed the inhibition of PI3K/Akt pathway in MCF-7/Adr cells induced by R7/PSD-Fol NPs. Finally, both in vivo distribution and in vivo antitumor showed the R7/PSD-Fol NPs displayed the better distribution at tumor site and the stronger suppression of tumor growth in the tumor bearing nude mice compared with control group. It was concluded that R7/PSD-Fol NPs loaded with DTX and GDC0941 could overcome MDR and enhance the antitumor effect further.
Collapse
Affiliation(s)
- Yiyue Wang
- School of Pharmacy, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, China
| | - Jing Li
- School of Pharmacy, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, China
| | - Jing Jing Chen
- School of Pharmacy, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, China
| | - Xuan Gao
- School of Pharmacy, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, China
| | - Zun Huang
- School of Pharmacy, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, China
| | - Qi Shen
- School of Pharmacy, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai 200240, China
| |
Collapse
|
222
|
Cellular Reprogramming Using Protein and Cell-Penetrating Peptides. Int J Mol Sci 2017; 18:ijms18030552. [PMID: 28273812 PMCID: PMC5372568 DOI: 10.3390/ijms18030552] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 02/27/2017] [Accepted: 02/28/2017] [Indexed: 12/13/2022] Open
Abstract
Recently, stem cells have been suggested as invaluable tools for cell therapy because of their self-renewal and multilineage differentiation potential. Thus, scientists have developed a variety of methods to generate pluripotent stem cells, from nuclear transfer technology to direct reprogramming using defined factors, or induced pluripotent stem cells (iPSCs). Considering the ethical issues and efficiency, iPSCs are thought to be one of the most promising stem cells for cell therapy. Induced pluripotent stem cells can be generated by transduction with a virus, plasmid, RNA, or protein. Herein, we provide an overview of the current technology for iPSC generation and describe protein-based transduction technology in detail.
Collapse
|
223
|
Hung LC, Jiang I, Chen CJ, Lu JY, Hsieh YF, Kuo PH, Hung YL, Wang LHC, Chang MDT, Sue SC. Heparin-Promoted Cellular Uptake of the Cell-Penetrating Glycosaminoglycan Binding Peptide, GBP ECP, Depends on a Single Tryptophan. ACS Chem Biol 2017; 12:398-406. [PMID: 27936565 DOI: 10.1021/acschembio.6b00864] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A 10-residue, glycosaminoglycan-binding peptide, GBPECP, derived from human eosinophil cationic protein has been recently designated as a potent cell-penetrating peptide. A model system containing peptide, glycan, and lipid was monitored by nuclear magnetic resonance (NMR) spectroscopy to determine the cell-penetrating mechanism. Heparin octasaccharide with dodecylphosphocholine (DPC) lipid micelle was titrated into the GBPECP solution. Our data revealed substantial roles for the charged residues Arg5 and Lys7 in recognizing heparin, whereas Arg3 had less effect. The aromatic residue Trp4 acted as an irreplaceable moiety for membrane insertion, as the replacement of Trp4 with Arg4 abolished cell penetration, although it significantly improved the heparin-binding ability. GBPECP bound either heparin or lipid in the presence or absence of the other ligand indicating that the peptide has two alternative binding sites: Trp4 is responsible for lipid insertion, and Arg5 and Lys7 are for GAG binding. We developed a molecular model showing that the two effects synergistically promote the penetration. The loss of either effect would abolish the penetration. GBPECP has been proven to enter cells through macropinocytosis. The GBPECP treatment inhibited A549 lung cancer cell migration and invasion, implying that the cellular microenvironment would be modulated by GBPECP internalization. The intracellular penetration of GBPECP leading to inhibition of epithelial cell migration and invasion depends on the presence of the tryptophan residue in its sequence compared with similar derivative peptides. Therefore, GBPECP shows substantial potential as a novel delivery therapeutic through rapid and effective internalization and interference with cell mobility.
Collapse
Affiliation(s)
- Li-Chun Hung
- Institute of Bioinformatics
and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ingjye Jiang
- Institute of Bioinformatics
and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chien-Jung Chen
- Institute of Molecular
and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Jia-Yin Lu
- Institute of Bioinformatics
and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yi-Fen Hsieh
- Institute of Molecular
and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ping-Hsieh Kuo
- Institute of Molecular
and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yi-Lin Hung
- Institute of Bioinformatics
and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
- Instrumentation
Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Lily Hui-Ching Wang
- Institute of Molecular
and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
- Department of Medical Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Margaret Dah-Tsyr Chang
- Institute of Molecular
and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Shih-Che Sue
- Institute of Bioinformatics
and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|
224
|
Zhao L, Chen G, Li J, Fu Y, Mavlyutov TA, Yao A, Nickells RW, Gong S, Guo LW. An intraocular drug delivery system using targeted nanocarriers attenuates retinal ganglion cell degeneration. J Control Release 2017; 247:153-166. [PMID: 28063892 PMCID: PMC5323250 DOI: 10.1016/j.jconrel.2016.12.038] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/08/2016] [Accepted: 12/29/2016] [Indexed: 02/03/2023]
Abstract
Glaucoma is a common blinding disease characterized by loss of retinal ganglion cells (RGCs). To date, there is no clinically available treatment directly targeting RGCs. We aim to develop an RGC-targeted intraocular drug delivery system using unimolecular micelle nanoparticles (unimNPs) to prevent RGC loss. The unimNPs were formed by single/individual multi-arm star amphiphilic block copolymer poly(amidoamine)-polyvalerolactone-poly(ethylene glycol) (PAMAM-PVL-PEG). While the hydrophobic PAMAM-PVL core can encapsulate hydrophobic drugs, the hydrophilic PEG shell provides excellent water dispersity. We conjugated unimNPs with the cholera toxin B domain (CTB) for RGC-targeting and with Cy5.5 for unimNP-tracing. To exploit RGC-protective sigma-1 receptor (S1R), we loaded unimNPs with an endogenous S1R agonist dehydroepiandrosterone (DHEA) as an FDA-approved model drug. These unimNPs produced a steady DHEA release in vitro for over two months at pH7.4. We then co-injected (mice, intraocular) unimNPs with the glutamate analog N-methyl-d-aspartate (NMDA), which is excito-toxic and induces RGC death. The CTB-conjugated unimNPs (i.e., targeted NPs) accumulated at the RGC layer and effectively preserved RGCs at least for 14days, whereas the unimNPs without CTB (i.e., non-targeted NPs) showed neither accumulation at nor protection of NMDA-treated RGCs. Consistent with S1R functions, targeted NPs relative to non-targeted NPs showed markedly better inhibitory effects on apoptosis and oxidative/inflammatory stresses in the RGC layer. Hence, the DHEA-loaded, CTB-conjugated unimNPs represent an RGC/S1R dual-targeted nanoplatform that generates an efficacious template for further development of a sustainable intraocular drug delivery system to protect RGCs, which may be applicable to treatments directed at glaucomatous pathology.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
| | - Guojun Chen
- Department of Materials Science and Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Jun Li
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA; Department of Ophthalmology, The First Hospital of China Medical University, Shenyang 110001, China.; Department of Ophthalmology, The 3rd People's Hospital of Dalian, Dalian 116033, China
| | - Yingmei Fu
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 Wanping Nan Road, Shanghai 200030, China
| | - Timur A Mavlyutov
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
| | - Annie Yao
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
| | - Robert W Nickells
- Department of Ophthalmology and Vision Sciences, University of Wisconsin-Madison, 1300 University Ave, Madison, WI 53706, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Shaoqin Gong
- Department of Materials Science and Engineering, Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Lian-Wang Guo
- Department of Surgery, 5151 Wisconsin Institute for Medical Research, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
225
|
Selby LI, Cortez-Jugo CM, Such GK, Johnston APR. Nanoescapology: progress toward understanding the endosomal escape of polymeric nanoparticles. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9. [PMID: 28160452 DOI: 10.1002/wnan.1452] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/07/2016] [Accepted: 12/17/2016] [Indexed: 02/06/2023]
Abstract
Using nanoparticles to deliver drugs to cells has the potential to revolutionize the treatment of many diseases, including HIV, cancer, and diabetes. One of the major challenges facing this field is controlling where the drug is trafficked once the nanoparticle is taken up into the cell. In particular, if drugs remain localized in an endosomal or lysosomal compartment, the therapeutic can be rendered completely ineffective. To ensure the design of more effective delivery systems we must first develop a better understanding of how nanoparticles and their cargo are trafficked inside cells. This needs to be combined with an understanding of what characteristics are required for nanoparticles to achieve endosomal escape, along with methods to detect endosomal escape effectively. This review is focused into three sections: first, an introduction to the mechanisms governing internalization and trafficking in cells, second, a discussion of methods to detect endosomal escape, and finally, recent advances in controlling endosomal escape from polymer- and lipid-based nanoparticles, with a focus on engineering materials to promote endosomal escape. WIREs Nanomed Nanobiotechnol 2017, 9:e1452. doi: 10.1002/wnan.1452 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Laura I Selby
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Christina M Cortez-Jugo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia.,Department of Chemical and Biomolecular Engineering, The University of Melbourne, Parkville, Victoria, Australia
| | - Georgina K Such
- Department of Chemistry, The University of Melbourne, Parkville, Victoria, Australia
| | - Angus P R Johnston
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
226
|
Tai W, Gao X. Functional peptides for siRNA delivery. Adv Drug Deliv Rev 2017; 110-111:157-168. [PMID: 27530388 PMCID: PMC5305781 DOI: 10.1016/j.addr.2016.08.004] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 06/21/2016] [Accepted: 08/05/2016] [Indexed: 11/19/2022]
Abstract
siRNA is considered as a potent therapeutic agent because of its high specificity and efficiency in suppressing genes that are overexpressed during disease development. For nearly two decades, a significant amount of efforts has been dedicated to bringing the siRNA technology into clinical uses. However, only limited success has been achieved to date, largely due to the lack of a cell type-specific, safe, and efficient delivery technology to carry siRNA into the target cells' cytosol where RNA interference takes place. Among the emerging candidate nanocarriers for siRNA delivery, peptides have gained popularity because of their structural and functional diversity. A variety of peptides have been discovered for their ability to translocate siRNA into living cells via different mechanisms such as direct penetration through the cellular membrane, endocytosis-mediated cell entry followed by endosomolysis, and receptor-mediated uptake. This review is focused on the multiple roles played by peptides in siRNA delivery, such as membrane penetration, endosome disruption, targeting, as well as the combination of these functionalities.
Collapse
Affiliation(s)
- Wanyi Tai
- Department of Bioengineering, University of Washington, William H Foege Building N561, Seattle, WA 98195, USA
| | - Xiaohu Gao
- Department of Bioengineering, University of Washington, William H Foege Building N561, Seattle, WA 98195, USA.
| |
Collapse
|
227
|
Rohovie MJ, Nagasawa M, Swartz JR. Virus-like particles: Next-generation nanoparticles for targeted therapeutic delivery. Bioeng Transl Med 2017; 2:43-57. [PMID: 29313023 PMCID: PMC5689521 DOI: 10.1002/btm2.10049] [Citation(s) in RCA: 230] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 11/23/2016] [Accepted: 11/30/2016] [Indexed: 12/12/2022] Open
Abstract
Most drug therapies distribute the agents throughout the entire body, even though the drugs are typically only needed at specific tissues. This often limits dosage and causes discomfort and harmful side‐effects. Significant research has examined nanoparticles (NPs) for use as targeted delivery vehicles for therapeutic cargo, however, major clinical success has been limited. Current work focuses mainly on liposomal and polymer‐based NPs, but emerging research is exploring the engineering of viral capsids as noninfectious protein‐based NPs—termed virus‐like particles (VLPs). This review covers the research that has been performed thus far and outlines the potential for these VLPs to become highly effective delivery vehicles that overcome the many challenges encountered for targeted delivery of therapeutic cargo.
Collapse
Affiliation(s)
- Marcus J Rohovie
- Dept. of Chemical Engineering Stanford University Stanford CA 94305
| | - Maya Nagasawa
- Dept. of Bioengineering Stanford University Stanford CA 94305
| | - James R Swartz
- Dept. of Chemical Engineering Stanford University Stanford CA 94305.,Dept. of Bioengineering Stanford University Stanford CA 94305
| |
Collapse
|
228
|
Wiedman G, Kim SY, Zapata-Mercado E, Wimley WC, Hristova K. pH-Triggered, Macromolecule-Sized Poration of Lipid Bilayers by Synthetically Evolved Peptides. J Am Chem Soc 2017; 139:937-945. [PMID: 28001058 PMCID: PMC5521809 DOI: 10.1021/jacs.6b11447] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
pH-triggered membrane-permeabilizing peptides could be exploited in a variety of applications, such as to enable cargo release from endosomes for cellular delivery, or as cancer therapeutics that selectively permeabilize the plasma membranes of malignant cells. Such peptides would be especially useful if they could enable the movement of macromolecules across membranes, a rare property in membrane-permeabilizing peptides. Here we approach this goal by using an orthogonal high-throughput screen of an iterative peptide library to identify peptide sequences that have the following two properties: (i) little synthetic lipid membrane permeabilization at physiological pH 7 at high peptide concentration and (ii) efficient formation of macromolecule-sized defects in synthetic lipid membranes at acidic pH 5 and low peptide concentration. The peptides we selected are remarkably potent macromolecular sized pore-formers at pH 5, while having little or no activity at pH 7, as intended. The action of these peptides likely relies on tight coupling between membrane partitioning, α-helix formation, and electrostatic repulsions between acidic side chains, which collectively drive a sharp pH-triggered transition between inactive and active configurations with apparent pKa values of 5.5-5.8. This work opens new doors to developing applications that utilize peptides with membrane-permeabilizing activities that are triggered by physiologically relevant decreases in pH.
Collapse
Affiliation(s)
- Gregory Wiedman
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218
| | - Sarah Y. Kim
- Graduate Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218
| | - Elmer Zapata-Mercado
- Graduate Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218
| | - William C. Wimley
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, 70112
| | - Kalina Hristova
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218
- Graduate Program in Molecular Biophysics, Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218
| |
Collapse
|
229
|
Oude Blenke E, Sleszynska M, Evers MJW, Storm G, Martin NI, Mastrobattista E. Strategies for the Activation and Release of the Membranolytic Peptide Melittin from Liposomes Using Endosomal pH as a Trigger. Bioconjug Chem 2017; 28:574-582. [DOI: 10.1021/acs.bioconjchem.6b00677] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- E. Oude Blenke
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| | - M. Sleszynska
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| | - M. J. W. Evers
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| | - G. Storm
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| | - N. I. Martin
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| | - E. Mastrobattista
- Department of Pharmaceutics and ‡Department of Chemical Biology and Drug Discovery, Utrecht Institute of Pharmaceutical Sciences (UIPS), Faculty of
Science, Utrecht University, 3512 JE Utrecht, Netherlands
| |
Collapse
|
230
|
Wang LL, Sloand JN, Gaffey AC, Venkataraman CM, Wang Z, Trubelja A, Hammer DA, Atluri P, Burdick JA. Injectable, Guest-Host Assembled Polyethylenimine Hydrogel for siRNA Delivery. Biomacromolecules 2017; 18:77-86. [PMID: 27997133 PMCID: PMC10953697 DOI: 10.1021/acs.biomac.6b01378] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
While siRNA has tremendous potential for therapeutic applications, advancement is limited by poor delivery systems. Systemically, siRNAs are rapidly degraded, may have off-target silencing, and necessitate high working concentrations. To overcome this, we developed an injectable, guest-host assembled hydrogel between polyethylenimine (PEI) and polyethylene glycol (PEG) for local siRNA delivery. Guest-host modified polymers assembled with siRNAs to form polyplexes that had improved transfection and viability compared to PEI. At higher concentrations, these polymers assembled into shear-thinning hydrogels that rapidly self-healed. With siRNA encapsulation, the assemblies eroded as polyplexes which were active and transfected cells, observed by Cy3-siRNA uptake or GFP silencing in vitro. When injected into rat myocardium, the hydrogels localized polyplex release, observed by uptake of Cy5.5-siRNA and silencing of GFP for 1 week in a GFP-expressing rat. These results illustrate the potential for this system to be applied for therapeutic siRNA delivery, such as in cardiac pathologies.
Collapse
Affiliation(s)
- Leo L. Wang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
| | - Janna N. Sloand
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
| | - Ann C. Gaffey
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, PA, 19104
| | - Chantel M. Venkataraman
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, PA, 19104
| | - Zhichun Wang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104
| | - Alen Trubelja
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, PA, 19104
| | - Daniel A. Hammer
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104
| | - Pavan Atluri
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, PA, 19104
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
| |
Collapse
|
231
|
Najjar K, Erazo-Oliveras A, Brock DJ, Wang TY, Pellois JP. An l- to d-Amino Acid Conversion in an Endosomolytic Analog of the Cell-penetrating Peptide TAT Influences Proteolytic Stability, Endocytic Uptake, and Endosomal Escape. J Biol Chem 2016; 292:847-861. [PMID: 27923812 DOI: 10.1074/jbc.m116.759837] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/05/2016] [Indexed: 12/12/2022] Open
Abstract
Cell-penetrating peptides (CPPs) are well established as delivery agents for otherwise cell-impermeable cargos. CPPs can also theoretically be used to modulate intracellular processes. However, their susceptibility to proteolytic degradation often limits their utility in these applications. Previous studies have explored the consequences for cellular uptake of converting the residues in CPPs from l- to d-stereochemistry, but conflicting results have been reported and specific steps en route to intracellular activity have not been explored. Here we use dimeric fluorescence TAT as a model CPP to explore the broader consequences of l- to d-stereochemical conversion. We show that inversion of chirality provides protease resistance without altering the overall mode of cellular entry, a process involving endocytic uptake followed by endosomal escape and cytosolic access. However, whereas inversion of chirality reduces endocytic uptake, the d-peptide, once in the endosome, is significantly more prone to escape than its l-counterpart. Moreover, the d-peptide is retained in the cytosol of cells for several days, whereas the l-peptide is degraded within hours. Notably, while the l-peptide is relatively innocuous to cells, the d-peptide exerts a prolonged anti-proliferative activity. Together, our results establish connections between chirality, protease resistance, cellular penetration, and intracellular activity that may be useful for the development of future delivery agents with improved properties.
Collapse
Affiliation(s)
- Kristina Najjar
- From the Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843
| | - Alfredo Erazo-Oliveras
- From the Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843
| | - Dakota J Brock
- From the Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843
| | - Ting-Yi Wang
- From the Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843
| | - Jean-Philippe Pellois
- From the Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843
| |
Collapse
|
232
|
Intelligent substance delivery into cells using cell-penetrating peptides. Bioorg Med Chem Lett 2016; 27:121-130. [PMID: 27956345 DOI: 10.1016/j.bmcl.2016.11.083] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 11/22/2016] [Accepted: 11/26/2016] [Indexed: 12/18/2022]
Abstract
Cell-penetrating peptides (CPPs) are oligopeptides that can permeate the cell membrane. The use of a CPP-mediated transport system could be an excellent method for delivering cell-impermeable substances such as proteins, antibodies, antisense oligonucleotides, siRNAs, plasmids, drugs, fluorescent compounds, and nanoparticles as covalently or noncovalently conjugated cargo into cells. Nonetheless, the mechanisms through which CPPs are internalized remain unclear. Endocytosis and direct translocation through the membrane are the generally accepted routes. Internalization via both pathways can occur simultaneously, depending on cellular conditions. However, the peculiar property of CPPs has attracted many researchers, especially in drug discovery or development, who intend to deliver impermeable substances into cells through the cell membrane. The delivery of drugs using CPPs may non-invasively solve the problem of drug penetration into cells with the added benefit of low cytotoxicity. Moreover, macromolecules can also be delivered by this transport system. In this review, I discuss the possibilities and advantages of substance delivery into cells using CPPs.
Collapse
|
233
|
Ullah I, Chung K, Beloor J, Kim J, Cho M, Kim N, Lee KY, Kumar P, Lee SK. Trileucine residues in a ligand-CPP-based siRNA delivery platform improve endosomal escape of siRNA. J Drug Target 2016; 25:320-329. [PMID: 27820977 DOI: 10.1080/1061186x.2016.1258566] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
siRNA entrapment within endosomes is a significant problem encountered with siRNA delivery platforms that co-opt receptor-mediated entry pathways. Attachment of a cell-penetrating peptide (CPP), such as nona-arginine (9R) to a cell receptor-binding ligand like the Rabies virus glycoprotein, RVG, allows effective siRNA delivery to the cytoplasm by non-endocytic pathways, but a significant amount of siRNA complexes also enters the cell by ligand-induced receptor endocytosis and remain localized in endosomes. Here, we report that the incorporation of trileucine (3 Leu) residues as an endo-osmolytic moiety in the peptide improves endosomal escape and intracellular delivery of siRNA. The trileucine motif did not affect early non-endosomal mechanism of cytoplasmic siRNA delivery but enhanced target gene silencing by >20% only beyond 24 h of transfection when siRNA delivery is mostly through the endocytic route and siRNA trapped in the endosomes at later stages were subject to release into cytoplasm. The mechanism may involve endosomal membrane disruption as trileucine residues lysed RBCs selectively under endosomal pH conditions. Interestingly <3 Leu or >3 Leu residues were not as effective, suggesting that 3 Leu residues are useful for enhancing cytoplasmic delivery of siRNA routed through endosomes.
Collapse
Affiliation(s)
- Irfan Ullah
- a Department of Bioengineering and Institute of Nanoscience and Technology , Hanyang University , Seoul , South Korea
| | - Kunho Chung
- a Department of Bioengineering and Institute of Nanoscience and Technology , Hanyang University , Seoul , South Korea.,b Department of Internal Medicine, Section of Infectious Diseases , Yale University School of Medicine , New Haven , CT, USA
| | - Jagadish Beloor
- b Department of Internal Medicine, Section of Infectious Diseases , Yale University School of Medicine , New Haven , CT, USA
| | - Jongkil Kim
- a Department of Bioengineering and Institute of Nanoscience and Technology , Hanyang University , Seoul , South Korea
| | - Minyoung Cho
- a Department of Bioengineering and Institute of Nanoscience and Technology , Hanyang University , Seoul , South Korea
| | - Nahyun Kim
- a Department of Bioengineering and Institute of Nanoscience and Technology , Hanyang University , Seoul , South Korea
| | - Kuen Yong Lee
- a Department of Bioengineering and Institute of Nanoscience and Technology , Hanyang University , Seoul , South Korea
| | - Priti Kumar
- b Department of Internal Medicine, Section of Infectious Diseases , Yale University School of Medicine , New Haven , CT, USA
| | - Sang-Kyung Lee
- a Department of Bioengineering and Institute of Nanoscience and Technology , Hanyang University , Seoul , South Korea
| |
Collapse
|
234
|
Ni R, Zhou J, Hossain N, Chau Y. Virus-inspired nucleic acid delivery system: Linking virus and viral mimicry. Adv Drug Deliv Rev 2016; 106:3-26. [PMID: 27473931 DOI: 10.1016/j.addr.2016.07.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 07/02/2016] [Accepted: 07/20/2016] [Indexed: 12/21/2022]
Abstract
Targeted delivery of nucleic acids into disease sites of human body has been attempted for decades, but both viral and non-viral vectors are yet to meet our expectations. Safety concerns and low delivery efficiency are the main limitations of viral and non-viral vectors, respectively. The structure of viruses is both ordered and dynamic, and is believed to be the key for effective transfection. Detailed understanding of the physical properties of viruses, their interaction with cellular components, and responses towards cellular environments leading to transfection would inspire the development of safe and effective non-viral vectors. To this goal, this review systematically summarizes distinctive features of viruses that are implied for efficient nucleic acid delivery but not yet fully explored in current non-viral vectors. The assembly and disassembly of viral structures, presentation of viral ligands, and the subcellular targeting of viruses are emphasized. Moreover, we describe the current development of cationic material-based viral mimicry (CVM) and structural viral mimicry (SVM) in these aspects. In light of the discrepancy, we identify future opportunities for rational design of viral mimics for the efficient delivery of DNA and RNA.
Collapse
Affiliation(s)
- Rong Ni
- Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Institute for Advanced Study, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Junli Zhou
- Department of Chemical and Biomolecular Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Naushad Hossain
- Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Ying Chau
- Department of Chemical and Biomolecular Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.
| |
Collapse
|
235
|
Robert MA, Lytvyn V, Deforet F, Gilbert R, Gaillet B. Virus-Like Particles Derived from HIV-1 for Delivery of Nuclear Proteins: Improvement of Production and Activity by Protein Engineering. Mol Biotechnol 2016; 59:9-23. [PMID: 27830536 DOI: 10.1007/s12033-016-9987-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Virus-like particles (VLPs) derived from retroviruses and lentiviruses can be used to deliver recombinant proteins without the fear of causing insertional mutagenesis to the host cell genome. In this study we evaluate the potential of an inducible lentiviral vector packaging cell line for VLP production. The Gag gene from HIV-1 was fused to a gene encoding a selected protein and it was transfected into the packaging cells. Three proteins served as model: the green fluorescent protein and two transcription factors-the cumate transactivator (cTA) of the inducible CR5 promoter and the human Krüppel-like factor 4 (KLF4). The sizes of the VLPs were 120-150 nm in diameter and they were resistant to freeze/thaw cycles. Protein delivery by the VLPs reached up to 100% efficacy in human cells and was well tolerated. Gag-cTA triggered up to 1100-fold gene activation of the reporter gene in comparison to the negative control. Protein engineering was required to detect Gag-KLF4 activity. Thus, insertion of the VP16 transactivation domain increased the activity of the VLPs by eightfold. An additional 2.4-fold enhancement was obtained by inserting nuclear export signal. In conclusion, our platform produced VLPs capable of efficient protein transfer, and it was shown that protein engineering can be used to improve the activity of the delivered proteins as well as VLP production.
Collapse
Affiliation(s)
- Marc-André Robert
- Département de génie chimique, Université Laval, 1065 Avenue de la Médecine, Québec, QC, G1V 0A6, Canada.,National Research Council Canada, 6100 Avenue Royalmount, Montréal, QC, H4P 2R2, Canada.,Regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, PROTEO, Québec, QC, Canada.,Réseau de thérapie cellulaire et tissulaire du FRQS, ThéCell, Québec, QC, Canada
| | - Viktoria Lytvyn
- National Research Council Canada, 6100 Avenue Royalmount, Montréal, QC, H4P 2R2, Canada
| | - Francis Deforet
- National Research Council Canada, 6100 Avenue Royalmount, Montréal, QC, H4P 2R2, Canada
| | - Rénald Gilbert
- National Research Council Canada, 6100 Avenue Royalmount, Montréal, QC, H4P 2R2, Canada.,Réseau de thérapie cellulaire et tissulaire du FRQS, ThéCell, Québec, QC, Canada
| | - Bruno Gaillet
- Département de génie chimique, Université Laval, 1065 Avenue de la Médecine, Québec, QC, G1V 0A6, Canada. .,Regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, PROTEO, Québec, QC, Canada. .,Réseau de thérapie cellulaire et tissulaire du FRQS, ThéCell, Québec, QC, Canada.
| |
Collapse
|
236
|
Liu MC, Chen CY, Chiang CH, Wang WM, Cheng RP. Effect of lysine methylation and acetylation on the RNA recognition and cellular uptake of Tat-derived peptides. Bioorg Med Chem 2016; 24:5047-5051. [DOI: 10.1016/j.bmc.2016.08.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 08/09/2016] [Accepted: 08/11/2016] [Indexed: 11/25/2022]
|
237
|
Tang R, Jiang Z, Ray M, Hou S, Rotello VM. Cytosolic delivery of large proteins using nanoparticle-stabilized nanocapsules. NANOSCALE 2016; 8:18038-18041. [PMID: 27738697 PMCID: PMC5137506 DOI: 10.1039/c6nr07162g] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
We report an effective intracellular delivery strategy for proteins of high molecular weight using AuNP stabilized capsules. This strategy provides direct delivery to the cytosol, avoiding endosomal entrapment.
Collapse
Affiliation(s)
- Rui Tang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA.
| | - Ziwen Jiang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA.
| | - Moumita Ray
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA.
| | - Singyuk Hou
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA.
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA.
| |
Collapse
|
238
|
Golan M, Feinshtein V, David A. Conjugates of HA2 with octaarginine-grafted HPMA copolymer offer effective siRNA delivery and gene silencing in cancer cells. Eur J Pharm Biopharm 2016; 109:103-112. [PMID: 27702685 DOI: 10.1016/j.ejpb.2016.09.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 09/19/2016] [Accepted: 09/28/2016] [Indexed: 01/20/2023]
Abstract
The key for successful gene silencing is to design a safe and efficient siRNA delivery system for the transfer of therapeutic nucleic acids into the target cells. Here, we describe the design of hydrophilic N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer displaying multiple copies of octaarginine (R8) and its use in promoting the effective delivery of small interfering RNA (siRNA) molecules intracellularly. Fluorescein-5-isothiocyanate (FITC)-labeled HPMA copolymer-bound R8 (P-R8-FITC) was synthesized with increasing R8 molar ratios (4-9.5mol-%) to define the optimal R8 content that allowed the polymer to serve both as a siRNA-binding domain and as an intracellular transduction moiety mediating improved cellular delivery. A subunit of the influenza virus hemagglutinin (HA2), known for its ability to disrupt endosomal membranes, was further conjugated to P-R8-FITC copolymer to promote endosomal escape. Of the different P-(R8)-FITC conjugates considered, only that polymer containing the highest mol-% of R8 (P-(R8)9.5-FITC) was able to encapsulate siRNA molecules into nano-sized polyion complexes (PICs) presenting positive surface charge, low in vitro cytotoxicity, and high serum stability. P-(R8)9.5-FITC/cy5-siRNA complexes can efficiently deliver siRNA molecules into cells, while naked siRNA or siRNA encapsulated within polymers with lower R8mol-% were unable to transfect the same cells. Conjugation of HA2 fusogenic peptide to P-(R8)-FITC significantly decreased the oncogenic RAC1 mRNA levels in cancer cells. This indicates that P-(R8)-(HA2)-FITC can deliver siRNA into target cells, and that the siRNA can reach the perinuclear region where it interacts with the RNA-induced silencing complex.
Collapse
Affiliation(s)
- Moran Golan
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Valeria Feinshtein
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Ayelet David
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel.
| |
Collapse
|
239
|
Photosensitizer enhanced disassembly of amphiphilic micelle for ROS-response targeted tumor therapy in vivo. Biomaterials 2016; 104:1-17. [DOI: 10.1016/j.biomaterials.2016.07.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/30/2016] [Accepted: 07/04/2016] [Indexed: 11/18/2022]
|
240
|
Gadok AK, Busch DJ, Ferrati S, Li B, Smyth HDC, Stachowiak JC. Connectosomes for Direct Molecular Delivery to the Cellular Cytoplasm. J Am Chem Soc 2016; 138:12833-12840. [DOI: 10.1021/jacs.6b05191] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Avinash K. Gadok
- Department
of Biomedical Engineering, ‡College of Pharmacy,
and §Institute for Cellular
and Molecular Biology, The University of Texas at Austin, Austin, Texas 78712, United States
| | - David J. Busch
- Department
of Biomedical Engineering, ‡College of Pharmacy,
and §Institute for Cellular
and Molecular Biology, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Silvia Ferrati
- Department
of Biomedical Engineering, ‡College of Pharmacy,
and §Institute for Cellular
and Molecular Biology, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Brian Li
- Department
of Biomedical Engineering, ‡College of Pharmacy,
and §Institute for Cellular
and Molecular Biology, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Hugh D. C. Smyth
- Department
of Biomedical Engineering, ‡College of Pharmacy,
and §Institute for Cellular
and Molecular Biology, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Jeanne C. Stachowiak
- Department
of Biomedical Engineering, ‡College of Pharmacy,
and §Institute for Cellular
and Molecular Biology, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
241
|
Yu LY, Su GM, Chen CK, Chiang YT, Lo CL. Specific Cancer Cytosolic Drug Delivery Triggered by Reactive Oxygen Species-Responsive Micelles. Biomacromolecules 2016; 17:3040-7. [PMID: 27536957 DOI: 10.1021/acs.biomac.6b00916] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cytosolic drug delivery, a major route in cancer therapy, is limited by the lack of efficient and safe endosomal escape techniques. Herein, we demonstrate a reactive oxygen species (ROS)-responsive micelle composed of methoxy polyethylene glycol-b-poly(diethyl sulfide) (mPEG-PS) copolymers which can induce specific endosome escape in cancer cells by changes in the hydrophobicity of copolymers. Owing to the more ROS levels in cancer cells than normal cells, the copolymers can be converted into more hydrophilic and insert into and destabilize the cancer intracellular endosome membrane after cellular uptake. More importantly, we show that acid-intolerant drugs successfully maintain their bioactivity and cause selective cytotoxicity for cancer cells over normal cells. Our results suggest that the endosomal escape induced by hydrophobic-hydrophilic exchange of copolymers has great potential to locally and efficiently deliver biological agents (e.g., proteins and genes) in the cancer cell cytosol.
Collapse
Affiliation(s)
| | | | | | - Yi-Ting Chiang
- School of Pharmacy, China Medical University , Taichung 40402, Taiwan, Republic of China
| | | |
Collapse
|
242
|
Abstract
RNA interference mediated gene silencing has tremendous applicability in fields ranging from basic biological research to clinical therapy. However, delivery of siRNA across the cell membrane into the cytoplasm, where the RNA silencing machinery is located, is a significant hurdle in most primary cells. Cell-penetrating peptides (CPPs), peptides that possess an intrinsic ability to translocate across cell membranes, have been explored as a means to achieve cellular delivery of siRNA. Approaches using CPPs by themselves or through incorporation into other siRNA delivery platforms have been investigated with the intent of improving cytoplasmic delivery. Here, we review the utilization of CPPs for siRNA delivery with a focus on strategies developed to enhance cellular uptake, endosomal escape and cytoplasmic localization of CPP/siRNA complexes.
Collapse
|
243
|
Endosomal acidic pH-induced conformational changes of a cytosol-penetrating antibody mediate endosomal escape. J Control Release 2016; 235:165-175. [DOI: 10.1016/j.jconrel.2016.05.066] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 05/13/2016] [Accepted: 05/31/2016] [Indexed: 01/11/2023]
|
244
|
Wang TY, Pellois JP. Peptide translocation through the plasma membrane of human cells: Can oxidative stress be exploited to gain better intracellular access? Commun Integr Biol 2016; 9:e1205771. [PMID: 27574543 PMCID: PMC4988429 DOI: 10.1080/19420889.2016.1205771] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 06/16/2016] [Accepted: 06/17/2016] [Indexed: 12/19/2022] Open
Abstract
Cell-penetrating peptides (CPPs) enter cells primarily by escaping from endosomal compartments or by directly translocating across the plasma membrane. Due to their capability of permeating into the cytosolic space of the cell, CPPs are utilized for the delivery of cell-impermeable molecules. However, the fundamental mechanisms and parameters associated with the penetration of CPPs and their cargos through the lipid bilayer have not been fully determined. This in turn has hampered their usage in biotechnological or therapeutic applications. We have recently reported that the cell penetration activity of poly-arginine CPPs (PACPPs) is dependent on the oxidation status of the plasma membrane of cells. Our data support a model where the positively-charged PACPP binds negatively-charged lipids exposed on the cell surface as a result of oxidative damage. The PACPP then crosses the membrane via formation of inverted micelles with these anionic lipids. This model provides a plausible explanation for the high variability in the cell delivery efficiency of a PACPP often observed in different settings. Notably, taking into account the current literature describing the effects of lipid oxidation, our data point to a highly complex and underappreciated interplay between PACPPs and oxidized membrane species. Overall, a better understanding of oxidation-dependent cell penetration might provide a fundamental basis for development of optimal cell permeable peptides (including cyclic peptides, stapled peptides, peptoids, etc…) and of robust delivery protocols.
Collapse
Affiliation(s)
- Ting-Yi Wang
- Department of Biochemistry and Biophysics, Texas A&M University , College Station, TX, USA
| | - Jean-Philippe Pellois
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, USA; Department of Chemistry, Texas A&M University, College Station, TX, USA
| |
Collapse
|
245
|
Lin YH, Chen YP, Liu TP, Chien FC, Chou CM, Chen CT, Mou CY. Approach To Deliver Two Antioxidant Enzymes with Mesoporous Silica Nanoparticles into Cells. ACS APPLIED MATERIALS & INTERFACES 2016; 8:17944-17954. [PMID: 27353012 DOI: 10.1021/acsami.6b05834] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Reactive oxygen species (ROS) are important factors in many clinical diseases. However, direct delivery of antioxidant enzymes into cells is difficult due to poor cell uptake. A proper design of delivery of enzymes by nanoparticles is very desirable for therapeutic purposes. To overcome the cell barrier problem, a designed mesoporous silica nanoparticle (MSN) system with attached TAT-fusion denatured enzyme for enhancing cell membrane penetration has been developed. Simultaneous delivery of two up-downstream antioxidant enzymes, superoxide dismutase (SOD) and glutathione peroxidase(GPx), reveals synergistic efficiency of ROS scavenging, compared to single antioxidant enzyme delivery. TAT peptide conjugation provided a facile nonendocytosis cell uptake and escape from endosome while moving and aggregating along the cytoskeleton that would allow them to be close to each other at the same time, resulting in the cellular antioxidation cascade reaction. The two-enzyme delivery shows a significant synergistic effect for protecting cells against ROS-induced cell damage and cell cycle arrest. The nanocarrier strategy for enzyme delivery demonstrates that intracellular anti-ROS cascade reactions could be regulated by multifunctional MSNs carrying image fluorophore and relevant antioxidation enzymes.
Collapse
Affiliation(s)
- Yu-Hsuan Lin
- Department of Chemistry, National Taiwan University , Taipei 106, Taiwan
| | | | - Tsang-Pai Liu
- Mackay Junior College of Medicine , Nursing and Management, Taipei 112, Taiwan
- Department of Surgery, Mackay Memorial Hospital , Taipei 104, Taiwan
| | - Fan-Ching Chien
- Department of Optics and Photonics, National Central University , Taoyuan City 320, Taiwan
| | | | | | - Chung-Yuan Mou
- Department of Chemistry, National Taiwan University , Taipei 106, Taiwan
| |
Collapse
|
246
|
Li S, Yuan H, Chen H, Wang X, Zhang P, Lv F, Liu L, Wang S. Cationic Poly(p-phenylene vinylene) Materials as a Multifunctional Platform for Light-Enhanced siRNA Delivery. Chem Asian J 2016; 11:2686-2689. [DOI: 10.1002/asia.201600447] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/10/2016] [Indexed: 11/07/2022]
Affiliation(s)
- Shengliang Li
- Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
| | - Huanxiang Yuan
- Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
| | - Hui Chen
- Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
| | - Xiaoyu Wang
- Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
| | - Pengbo Zhang
- Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
| | - Fengting Lv
- Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
| | - Libing Liu
- Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
| | - Shu Wang
- Key Laboratory of Organic Solids; Institute of Chemistry; Chinese Academy of Sciences; Beijing 100190 P. R. China
| |
Collapse
|
247
|
Toy R, Roy K. Engineering nanoparticles to overcome barriers to immunotherapy. Bioeng Transl Med 2016; 1:47-62. [PMID: 29313006 PMCID: PMC5689503 DOI: 10.1002/btm2.10005] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/14/2016] [Accepted: 04/15/2016] [Indexed: 12/14/2022] Open
Abstract
Advances in immunotherapy have led to the development of a variety of promising therapeutics, including small molecules, proteins and peptides, monoclonal antibodies, and cellular therapies. Despite this wealth of new therapeutics, the efficacy of immunotherapy has been limited by challenges in targeted delivery and controlled release, that is, spatial and temporal control on delivery. Particulate carriers, especially nanoparticles have been widely studied in drug delivery and vaccine research and are being increasingly investigated as vehicles to deliver immunotherapies. Nanoparticle-mediated drug delivery could provide several benefits, including control of biodistribution and transport kinetics, the potential for site-specific targeting, immunogenicity, tracking capability using medical imaging, and multitherapeutic loading. There are also a unique set of challenges, which include nonspecific uptake by phagocytic cells, off-target biodistribution, permeation through tissue (transport limitation), nonspecific immune-activation, and poor control over intracellular localization. This review highlights the importance of understanding the relationship between a nanoparticle's size, shape, charge, ligand density and elasticity to its vascular transport, biodistribution, cellular internalization, and immunogenicity. For the design of an effective immunotherapy, we highlight the importance of selecting a nanoparticle's physical characteristics (e.g., size, shape, elasticity) and its surface functionalization (e.g., chemical or polymer modifications, targeting or tissue-penetrating peptides) with consideration of its reactivity to the targeted microenvironment (e.g., targeted cell types, use of stimuli-sensitive biomaterials, immunogenicity). Applications of this rational nanoparticle design process in vaccine development and cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Randall Toy
- Wallace H. Coulter Dept. of Biomedical Engineering Georgia Institute of Technology, and Emory University Atlanta GA 30332
| | - Krishnendu Roy
- Wallace H. Coulter Dept. of Biomedical Engineering Georgia Institute of Technology, and Emory University Atlanta GA 30332
| |
Collapse
|
248
|
XTEN as Biological Alternative to PEGylation Allows Complete Expression of a Protease-Activatable Killin-Based Cytostatic. PLoS One 2016; 11:e0157193. [PMID: 27295081 PMCID: PMC4905650 DOI: 10.1371/journal.pone.0157193] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 05/25/2016] [Indexed: 12/31/2022] Open
Abstract
Increased effectiveness and reduced side effects are general goals in drug research, especially important in cancer therapy. The aim of this study was to design a long-circulating, activatable cytostatic drug that is completely producible in E. coli. Crucial for this goal was the novel unstructured polypeptide XTEN, which acts like polyethylene glycol (PEG) but has many important advantages. Most importantly, it can be produced in E. coli, is less immunogenic, and is biodegradable. We tested constructs containing a fragment of Killin as cytostatic/cytotoxic element, a cell-penetrating peptide, an MMP-2 cleavage site for specific activation, and XTEN for long blood circulation and deactivation of Killin. One of three sequence variants was efficiently expressed in E. coli. As typical for XTEN, it allowed efficient purification of the E. coli lysate by a heat step (10 min 75°C) and subsequent anion exchange chromatography using XTEN as purification tag. After 24 h XTEN-Killin reduced the number of viable cells of HT-1080 tumor cell line to 3.8 ±2.0% (p<0.001) compared to untreated controls. In contrast, liver derived non-tumor cells (BRL3A) did not show significant changes in viability. Our results demonstrate the feasibility of completely producing a complex protease-activatable, potentially long-circulating cytostatic/cytotoxic prodrug in E. coli—a concept that could lead to efficient production of highly multifunctional drugs in the future.
Collapse
|
249
|
Krautwald S, Dewitz C, Fändrich F, Kunzendorf U. Inhibition of regulated cell death by cell-penetrating peptides. Cell Mol Life Sci 2016; 73:2269-84. [PMID: 27048815 PMCID: PMC4887531 DOI: 10.1007/s00018-016-2200-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 03/18/2016] [Indexed: 12/18/2022]
Abstract
Development of the means to efficiently and continuously renew missing and non-functional proteins in diseased cells remains a major goal in modern molecular medicine. While gene therapy has the potential to achieve this, substantial obstacles must be overcome before clinical application can be considered. A promising alternative approach is the direct delivery of non-permeant active biomolecules, such as oligonucleotides, peptides and proteins, to the affected cells with the purpose of ameliorating an advanced disease process. In addition to receptor-mediated endocytosis, cell-penetrating peptides are widely used as vectors for rapid translocation of conjugated molecules across cell membranes into intracellular compartments and the delivery of these therapeutic molecules is generally referred to as novel prospective protein therapy. As a broad coverage of the enormous amount of published data in this field is unrewarding, this review will provide a brief, focused overview of the technology and a summary of recent studies of the most commonly used protein transduction domains and their potential as therapeutic agents for the treatment of cellular damage and the prevention of regulated cell death.
Collapse
Affiliation(s)
- Stefan Krautwald
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, 24105, Kiel, Germany.
| | - Christin Dewitz
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, 24105, Kiel, Germany
| | - Fred Fändrich
- Clinic for Applied Cellular Medicine, University Hospital Schleswig-Holstein, 24105, Kiel, Germany
| | - Ulrich Kunzendorf
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, 24105, Kiel, Germany
| |
Collapse
|
250
|
Erazo-Oliveras A, Najjar K, Truong D, Wang TY, Brock DJ, Prater AR, Pellois JP. The Late Endosome and Its Lipid BMP Act as Gateways for Efficient Cytosolic Access of the Delivery Agent dfTAT and Its Macromolecular Cargos. Cell Chem Biol 2016; 23:598-607. [PMID: 27161484 DOI: 10.1016/j.chembiol.2016.03.016] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 03/16/2016] [Accepted: 03/22/2016] [Indexed: 01/02/2023]
Abstract
Endosomal entrapment is a severely limiting bottleneck in the delivery of biologics into cells. The compound dfTAT was recently found to circumvent this problem by mediating endosomal leakage efficiently and without toxicity. Herein, we report on the mechanism of endosomal escape of this cell-penetrating peptide. By modulating the trafficking of the peptide within the endocytic pathway, we identify late endosomes as the organelles rendered leaky by dfTAT. We establish that dfTAT binds bis(monoacylglycero)phosphate (BMP), a lipid found in late endosomes, and that the peptide causes the fusion and leakage of bilayers containing BMP. Together, these data identify late endosomes as desirable gateways for cell penetration and BMP as a cellular factor that can be exploited for the development of future delivery agents.
Collapse
Affiliation(s)
- Alfredo Erazo-Oliveras
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Boulevard, College Station, TX 77843, USA
| | - Kristina Najjar
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Boulevard, College Station, TX 77843, USA
| | - Dat Truong
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Boulevard, College Station, TX 77843, USA
| | - Ting-Yi Wang
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Boulevard, College Station, TX 77843, USA
| | - Dakota J Brock
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Boulevard, College Station, TX 77843, USA
| | - Austin R Prater
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Boulevard, College Station, TX 77843, USA
| | - Jean-Philippe Pellois
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Boulevard, College Station, TX 77843, USA; Department of Chemistry, Texas A&M University, 300 Olsen Boulevard, College Station, TX 77843, USA.
| |
Collapse
|