1
|
Shin HJ, Lee BK, Kang HA. Transdermal Properties of Cell-Penetrating Peptides: Applications and Skin Penetration Mechanisms. ACS APPLIED BIO MATERIALS 2024; 7:1-16. [PMID: 38079575 DOI: 10.1021/acsabm.3c00659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Cell-penetrating peptides (CPPs) consist of 5-30 amino acids with intracellular transduction abilities and diverse physicochemical properties, origins, and sequences. Although recent developments in bioinformatics have facilitated the prediction of CPP candidates with the potential for transduction into cells, the mechanisms by which CPPs penetrate cells and various tissues have not yet been elucidated at the molecular interaction level. Recently, the skin-penetrating ability of CPPs has gained wide attention and emerged as a simple and effective strategy for the delivery of macromolecules into the skin. Studies on the skin structure have suggested that the penetration potential of CPPs is based on the molecular interactions and characteristics of the lipid lamellar structure between corneocytes in the stratum corneum. This review provides a brief overview of the general properties, transduction mechanisms, applications, and safety issues of CPPs, focusing on CPPs with transdermal properties, that are currently being used to develop therapeutics and cosmetics.
Collapse
Affiliation(s)
- Hee Je Shin
- ProCell R&D Center, ProCell Therapeutics, Inc., #1009 Ace-Twin Tower II, 273, Digital-ro, Guro-gu, Seoul 08381, Republic of Korea
- Department of Life Science, College of Natural Science, Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Byung Kyu Lee
- ProCell R&D Center, ProCell Therapeutics, Inc., #1009 Ace-Twin Tower II, 273, Digital-ro, Guro-gu, Seoul 08381, Republic of Korea
| | - Hyun Ah Kang
- Department of Life Science, College of Natural Science, Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| |
Collapse
|
2
|
Efficient transdermal delivery of functional protein cargoes by a hydrophobic peptide MTD 1067. Sci Rep 2022; 12:10853. [PMID: 35760980 PMCID: PMC9237094 DOI: 10.1038/s41598-022-14463-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/07/2022] [Indexed: 11/09/2022] Open
Abstract
The skin has a protective barrier against the external environment, making the transdermal delivery of active macromolecules very difficult. Cell-penetrating peptides (CPPs) have been accepted as useful delivery tools owing to their high transduction efficiency and low cytotoxicity. In this study, we evaluated the hydrophobic peptide, macromolecule transduction domain 1067 (MTD 1067) as a CPP for the transdermal delivery of protein cargoes of various sizes, including growth hormone-releasing hexapeptide-6 (GHRP-6), a truncated form of insulin-like growth factor-I (des(1-3)IGF-I), and platelet-derived growth factor BB (PDGF-BB). The MTD 1067-conjugated GHRP-6 (MTD-GHRP-6) was chemically synthesized, whereas the MTD 1067-conjugated des(1-3)IGF-I and PDGF-BB proteins (MTD-des(1-3)IGF-I and MTD-PDGF-BB) were generated as recombinant proteins. All the MTD 1067-conjugated cargoes exhibited biological activities identical or improved when compared to those of the original cargoes. The analysis of confocal microscopy images showed that MTD-GHRP-6, MTD-des(1-3)IGF-I, and MTD-PDGF-BB were detected at 4.4-, 18.8-, and 32.9-times higher levels in the dermis, respectively, compared to the control group without MTD. Furthermore, the MTD 1067-conjugated cargoes did not show cytotoxicity. Altogether, our data demonstrate the potential of MTD 1067 conjugation in developing functional macromolecules for cosmetics and drugs with enhanced transdermal permeability.
Collapse
|
3
|
Chuang HC, Ding DS, Fan CH, Lin CH, Cheng CM. Effect of cell-permeable grouper Manganese Superoxide Dismutase on environmental stress in fish. Protein Expr Purif 2021; 187:105951. [PMID: 34358651 DOI: 10.1016/j.pep.2021.105951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 11/29/2022]
Abstract
Nitrite levels are generally high in high-density aquaculture. Nitrite is a potential stress-inducing factor and can cause oxidative stress because excessive reactive oxygen species (ROS) formation through nitrite induction cannot be scavenged by the endogenous antioxidant system, thus leading to cell damage or death. Manganese Superoxide Dismutase (MnSOD) is a highly efficient endogenous ROS scavenger that quenches mitochondrial ROS and protective against oxidative stress. To enhance the efficiency of MnSOD in removing ROS and reducing oxidative caused by nitrite, in this study, we cloned grouper MnSOD (gMnSOD) fused with a cell-penetrating peptide, TAT, to construct a TAT-gMnSOD fusion protein and assessed its potential to eliminate excess ROS induced by high nitrite concentrations and enhance the resistance of zebrafish to environmental stressors. Our results revealed that TAT-gMnSOD penetrated the grouper fin (GF-1) cells, scavenged nitrite-induced intracellular ROS, and enhanced cell viability on NaNO2 treatment. Furthermore, pretreatment of zebrafish with TAT-gMnSOD fusion protein reduced the MDA content and increased the survival rate. In addition, the TAT-gMnSOD fusion protein reduced 2-phenoxyethanol toxicity and attenuated excessive anesthesia among zebrafish. In conlusion, our cell-permeable TAT-gMnSOD fusion protein effectively counters oxidative stress, prevents environmental stress-induced damage, and increases aquaculture benefits.
Collapse
Affiliation(s)
- Hsiang-Chieh Chuang
- Department and Graduate Institute of Aquaculture, National Kaohsiung University of Science and Technology, Kaohsiung, Taiwan.
| | - De-Sing Ding
- Ph.D. Program of Aquatic Science and Technology in Industry, College of Science, National Kaohsiung University of Science and Technology, Kaohsiung, Taiwan.
| | - Chih-Hsuan Fan
- Department and Graduate Institute of Aquaculture, National Kaohsiung University of Science and Technology, Kaohsiung, Taiwan.
| | - Chia-Hua Lin
- Ph.D. Program of Aquatic Science and Technology in Industry, College of Science, National Kaohsiung University of Science and Technology, Kaohsiung, Taiwan.
| | - Chiu-Min Cheng
- Department and Graduate Institute of Aquaculture, National Kaohsiung University of Science and Technology, Kaohsiung, Taiwan.
| |
Collapse
|
4
|
The Spectrum of Design Solutions for Improving the Activity-Selectivity Product of Peptide Antibiotics against Multidrug-Resistant Bacteria and Prostate Cancer PC-3 Cells. Molecules 2020; 25:molecules25153526. [PMID: 32752241 PMCID: PMC7436000 DOI: 10.3390/molecules25153526] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/24/2022] Open
Abstract
The link between the antimicrobial and anticancer activity of peptides has long been studied, and the number of peptides identified with both activities has recently increased considerably. In this work, we hypothesized that designed peptides with a wide spectrum of selective antimicrobial activity will also have anticancer activity, and tested this hypothesis with newly designed peptides. The spectrum of peptides, used as partial or full design templates, ranged from cell-penetrating peptides and putative bacteriocin to those from the simplest animals (placozoans) and the Chordata phylum (anurans). We applied custom computational tools to predict amino acid substitutions, conferring the increased product of bacteriostatic activity and selectivity. Experiments confirmed that better overall performance was achieved with respect to that of initial templates. Nine of our synthesized helical peptides had excellent bactericidal activity against both standard and multidrug-resistant bacteria. These peptides were then compared to a known anticancer peptide polybia-MP1, for their ability to kill prostate cancer cells and dermal primary fibroblasts. The therapeutic index was higher for seven of our peptides, and anticancer activity stronger for all of them. In conclusion, the peptides that we designed for selective antimicrobial activity also have promising potential for anticancer applications.
Collapse
|
5
|
Thomas E, Dragojevic S, Price A, Raucher D. Thermally Targeted p50 Peptide Inhibits Proliferation and Induces Apoptosis of Breast Cancer Cell Lines. Macromol Biosci 2020; 20:e2000170. [PMID: 32734662 DOI: 10.1002/mabi.202000170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/06/2020] [Indexed: 12/26/2022]
Abstract
The application of rationally designed therapeutic peptides (TP) may improve outcomes in cancer treatment. These peptides hold the potential to directly target proliferative pathways and stimulate cell arrest or death pathways. Elastin-like polypeptide (ELP) is an elastin derived biopolymer that undergoes a thermally mediated phase transition. This study employs p50, a nuclear localization sequence derived peptide that inhibits the activation of NFκB and is implicated in cancer cell survival and metastasis. In order to effectively delivery p50, it is conjugated to SynB1-ELP1, a thermally responsive macromolecular carrier. By applying an external heat source, mild hyperthermic conditions (41 °C) induce aggregation and therefore can be used to specifically target ELP to solid tumors in cancer therapy. The addition of a cell penetrating peptide (CPP) to the N-terminus of the macromolecular carrier enhances the cellular uptake and directs the subcellular localization of the bioactive peptide. The novel TP, p50, inhibits proliferation and induces apoptosis of breast cancer cells by blocking the intranuclear import of NFκB. By expanding the repertoire of oncogenic targets, CPPs, and ELP carrier sizes, ELP-based polypeptides may be modulated to optimize the delivery of these novel therapies and allow for the flexibility to create individualized cancer therapies.
Collapse
Affiliation(s)
- Emily Thomas
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N State St, Jackson, MS, 39216, USA
| | - Sonja Dragojevic
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N State St, Jackson, MS, 39216, USA.,Department of Radiation Oncology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Amira Price
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N State St, Jackson, MS, 39216, USA
| | - Drazen Raucher
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N State St, Jackson, MS, 39216, USA
| |
Collapse
|
6
|
Zhang K, Cheng X, Zhao L, Huang M, Tao Y, Zhang H, Rosenholm JM, Zhuang M, Chen ZY, Chen B, Shu Y. Direct Functional Protein Delivery with a Peptide into Neonatal and Adult Mammalian Inner Ear In Vivo. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:511-519. [PMID: 32953927 PMCID: PMC7477795 DOI: 10.1016/j.omtm.2020.06.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 06/25/2020] [Indexed: 11/26/2022]
Abstract
The aim of this study was to study an antimicrobial peptide (AMP), aurein 1.2, which substantially increased protein delivery directly into multiple mammalian inner-ear cell types in vivo. Different concentrations of aurein 1.2 with superpositively charged GFP (+36 GFP) protein fused with Cre recombinase were delivered to postnatal day 1-2 (P1-2) and adult cochleae of Cre reporter transgenic mice with various delivery methods. By cochleostomy at different concentrations of aurein 1.2-+36 GFP (1 μM, 5 μM, 22.5 μM, and 50 μM, respectively), the tdTomato (tdT) expression was observed in outer hair cells (OHCs; 20.77%, 23.02%, 76.36%, and 92.47%, respectively) and inner hair cells (IHCs; 14.90%, 44.50%, 89.59%, and 96.13%, respectively) in the cochlea. The optimal concentration was 22.5 μM with the highest transfection efficiency and the lowest cytotoxicity. Wide-spread tdT signals were detected in the cochlear-supporting cells, utricular-supporting cells, auditory nerve, and spiral ligament in neonatal and adult mice. Compared to cochleostomy, injection through the round window membrane (RWM) also produced highly efficient tdT+ labeled cells with less cell loss. In summary, the peptide aurein 1.2 fused with +36 GFP dramatically expanded the target cells with increased efficiency in direct protein delivery in the inner ear. Aurein 1.2-+36 GFP has the potential to be developed as protein-based therapy in regeneration and genome editing in the mammalian inner ear.
Collapse
Affiliation(s)
- Kun Zhang
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China
| | - Xiaoting Cheng
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China
| | - Liping Zhao
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China
| | - Mingqian Huang
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA.,Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, 243 Charles St., Boston, MA 02114, USA
| | - Yong Tao
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China.,Ear Institute, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China.,Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, Shanghai 200011, China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory and Turku Bioscience Center, Åbo Akademi University, 20520 Turku, Finland
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory and Turku Bioscience Center, Åbo Akademi University, 20520 Turku, Finland
| | - Min Zhuang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zheng-Yi Chen
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA.,Eaton-Peabody Laboratory, Massachusetts Eye and Ear Infirmary, 243 Charles St., Boston, MA 02114, USA
| | - Bing Chen
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China
| | - Yilai Shu
- ENT Institute and Otorhinolaryngology Department, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.,NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China
| |
Collapse
|
7
|
Peptide-based targeted therapeutics: Focus on cancer treatment. J Control Release 2018; 292:141-162. [DOI: 10.1016/j.jconrel.2018.11.004] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/03/2018] [Accepted: 11/03/2018] [Indexed: 12/14/2022]
|
8
|
Birch D, Diedrichsen RG, Christophersen PC, Mu H, Nielsen HM. Evaluation of drug permeation under fed state conditions using mucus-covered Caco-2 cell epithelium. Eur J Pharm Sci 2018. [DOI: 10.1016/j.ejps.2018.02.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
9
|
Soleymani-Goloujeh M, Nokhodchi A, Niazi M, Najafi-Hajivar S, Shahbazi-Mojarrad J, Zarghami N, Zakeri-Milani P, Mohammadi A, Karimi M, Valizadeh H. Effects of N-terminal and C-terminal modification on cytotoxicity and cellular uptake of amphiphilic cell penetrating peptides. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:91-103. [DOI: 10.1080/21691401.2017.1414823] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Mehdi Soleymani-Goloujeh
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ali Nokhodchi
- Pharmaceutics Research Laboratory, School of Life Sciences, University of Sussex, Falmer, Brighton, UK
| | - Mehri Niazi
- Student Research Committee, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeedeh Najafi-Hajivar
- Student Research Committee, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Shahbazi-Mojarrad
- Biotechnology Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Zakeri-Milani
- Liver and Gastrointestinal Diseases Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Karimi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Valizadeh
- Drug Applied Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
10
|
Birch D, Christensen MV, Staerk D, Franzyk H, Nielsen HM. Fluorophore labeling of a cell-penetrating peptide induces differential effects on its cellular distribution and affects cell viability. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:2483-2494. [DOI: 10.1016/j.bbamem.2017.09.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 09/12/2017] [Accepted: 09/13/2017] [Indexed: 01/28/2023]
|
11
|
Holub JM. Small Scaffolds, Big Potential: Developing Miniature Proteins as Therapeutic Agents. Drug Dev Res 2017; 78:268-282. [PMID: 28799168 DOI: 10.1002/ddr.21408] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 07/26/2017] [Indexed: 12/14/2022]
Abstract
Preclinical Research Miniature proteins are a class of oligopeptide characterized by their short sequence lengths and ability to adopt well-folded, three-dimensional structures. Because of their biomimetic nature and synthetic tractability, miniature proteins have been used to study a range of biochemical processes including fast protein folding, signal transduction, catalysis and molecular transport. Recently, miniature proteins have been gaining traction as potential therapeutic agents because their small size and ability to fold into defined tertiary structures facilitates their development as protein-based drugs. This research overview discusses emerging developments involving the use of miniature proteins as scaffolds to design novel therapeutics for the treatment and study of human disease. Specifically, this review will explore strategies to: (i) stabilize miniature protein tertiary structure; (ii) optimize biomolecular recognition by grafting functional epitopes onto miniature protein scaffolds; and (iii) enhance cytosolic delivery of miniature proteins through the use of cationic motifs that facilitate endosomal escape. These objectives are discussed not only to address challenges in developing effective miniature protein-based drugs, but also to highlight the tremendous potential miniature proteins hold for combating and understanding human disease. Drug Dev Res 78 : 268-282, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Justin M Holub
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH, 45701, USA
| |
Collapse
|
12
|
Sánchez-García L, Serna N, Mattanovich M, Cazzanelli P, Sánchez-Chardi A, Conchillo-Solé O, Cortés F, Daura X, Unzueta U, Mangues R, Villaverde A, Vázquez E. The fusogenic peptide HA2 impairs selectivity of CXCR4-targeted protein nanoparticles. Chem Commun (Camb) 2017; 53:4565-4568. [DOI: 10.1039/c6cc09900a] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We demonstrate here that the genetic incorporation of the fusogenic peptide HA2 to a CXCR4-targeted protein nanoparticle dramatically reduces the specificity of the interaction between nanoparticles and cell receptor.
Collapse
|
13
|
Hu S, Wang T, Pei X, Cai H, Chen J, Zhang X, Wan Q, Wang J. Synergistic Enhancement of Antitumor Efficacy by PEGylated Multi-walled Carbon Nanotubes Modified with Cell-Penetrating Peptide TAT. NANOSCALE RESEARCH LETTERS 2016; 11:452. [PMID: 27726120 PMCID: PMC5056908 DOI: 10.1186/s11671-016-1672-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 10/05/2016] [Indexed: 06/06/2023]
Abstract
In the present study, a cell-penetrating peptide, the transactivating transcriptional factor (TAT) domain from HIV, was linked to PEGylated multi-walled carbon nanotubes (MWCNTs) to develop a highly effective antitumor drug delivery system. FITC was conjugated on MWCNTs-polyethylene glycol (PEG) and MWCNTs-PEG-TAT to provide fluorescence signal for tracing the cellular uptake of the nanocarrier. After loaded with an anticancer agent, doxorubicin (DOX) via π - π stacking interaction, the physicochemical characteristics, release profile and biological evaluation of the obtained nano-sized drug carrier were investigated. The DOX loaded MWCNTs-PEG and MWCNTs-PEG-TAT drug carriers both displayed appropriate particle size, excellent stability, high drug loading, and pH-dependent drug release profile. Nevertheless, compared with DOX-MWCNTs-PEG, DOX-MWCNTs-PEG-TAT showed improved cell internalization, intracellular distribution and potentiated anticancer efficacy due to the TAT-mediated membrane translocation, endosomal escape and nuclear targeting. Furthermore, the therapeutic efficacy of DOX was not compromised after being conjugated with MWCNTs-PEG-TAT and the proposed nanocarrier was also confirmed to have a good biocompatibility. In conclusion, our results suggested that the unique combination of TAT and MWCNTs as a multifunctional drug delivery system might be a powerful tool for improved anticancer drug development.
Collapse
Affiliation(s)
- Shanshan Hu
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Tong Wang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xibo Pei
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Prosthodontics, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - He Cai
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Junyu Chen
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xin Zhang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Qianbing Wan
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Prosthodontics, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jian Wang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China.
- Department of Prosthodontics, West China College of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
14
|
Abe S, Yamamoto K, Kurata M, Abe-Suzuki S, Horii R, Akiyama F, Kitagawa M. Targeting MCM2 function as a novel strategy for the treatment of highly malignant breast tumors. Oncotarget 2016; 6:34892-909. [PMID: 26430873 PMCID: PMC4741497 DOI: 10.18632/oncotarget.5408] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 09/18/2015] [Indexed: 12/23/2022] Open
Abstract
Highly malignant tumors express high levels of the minichromosome maintenance 2 (MCM2) protein, which is associated with advanced tumor grade, advanced stage, and poor prognosis. In a previous study, we showed that Friend leukemia virus (FLV) envelope protein gp70 bound MCM2, impaired its nuclear translocation, and enhanced DNA-damage-induced apoptosis in FLV-infected hematopoietic cells when the cells expressed high levels of MCM2. Here, we show that MCM2 is highly expressed in clinical samples of invasive carcinoma of the breast, especially triple-negative breast cancer (TNBC), and in cancer stem cell (CSC) marker-positive breast cancer cells. To generate a cancer therapy model using gp70, we introduced the gp70 protein into the cytoplasm of murine breast cancer cells that express high levels of MCM2 by conjugating the protein transduction domain (PTD) of Hph-1 to gp70 (Hph- 1-gp70). Hph-1-gp70 was successfully transduced into the cytoplasm of breast cancer cells. The transduced protein enhanced the DNA damage-induced apoptosis of cancer cells in vitro and in vivo. Therefore, an MCM2-targeted strategy using Hph-1-gp70 treatment to induce DNA damage might be a successful therapy for highly malignant breast cancers such as TNBC and for the eradication of CSC-like cells from breast cancer tissue.
Collapse
Affiliation(s)
- Shinya Abe
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kouhei Yamamoto
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Morito Kurata
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shiho Abe-Suzuki
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Rie Horii
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Futoshi Akiyama
- Department of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masanobu Kitagawa
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
15
|
Pepe D, Carvalho VF, McCall M, de Lemos DP, Lopes LB. Transportan in nanocarriers improves skin localization and antitumor activity of paclitaxel. Int J Nanomedicine 2016; 11:2009-19. [PMID: 27274232 PMCID: PMC4869655 DOI: 10.2147/ijn.s97331] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In this study, the ability of nanocarriers containing protein transduction domains (PTDs) of various classes to improve cutaneous paclitaxel delivery and efficacy in skin tumor models was evaluated. Microemulsions (MEs) were prepared by mixing a surfactant blend (polyoxyethylene 10 oleoyl ether, ethanol and propylene glycol), monocaprylin, and water. The PTD transportan (ME-T), penetratin (ME-P), or TAT (ME-TAT) was added at a concentration of 1 mM to the plain ME. All MEs displayed nanometric size (32.3–40.7 nm) and slight positive zeta potential (+4.1 mV to +6.8 mV). Skin penetration of paclitaxel from the MEs was assessed for 1–12 hours using porcine skin and Franz diffusion cells. Among the PTD-containing formulations, paclitaxel skin (stratum corneum + epidermis and dermis) penetration at 12 hours was maximized with ME-T, whereas ME-TAT provided the lowest penetration (1.6-fold less). This is consistent with the stronger ability of ME-T to increase transepidermal water loss (2.4-fold compared to water) and tissue permeability. The influence of PTD addition on the ME irritation potential was assessed by measuring interleukin-1α expression and viability of bioengineered skin equivalents. A 1.5- to 1.8-fold increase in interleukin-1α expression was induced by ME-T compared to the other formulations, but this effect was less pronounced (5.8-fold) than that mediated by the moderate irritant Triton. Because ME-T maximized paclitaxel cutaneous localization while being safer than Triton, its efficacy was assessed against basal cell carcinoma cells and a bioengineered three-dimensional melanoma model. Paclitaxel-containing ME-T reduced cells and tissue viability by twofold compared to drug solutions, suggesting the potential clinical usefulness of the formulation for the treatment of cutaneous tumors.
Collapse
Affiliation(s)
- Dominique Pepe
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| | - Vanessa Fm Carvalho
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Melissa McCall
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| | - Débora P de Lemos
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luciana B Lopes
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY, USA; Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
16
|
|
17
|
Zakrewsky M, Muraski JA, Mitragotri S. Mechanistic Analysis of Cellular Internalization of a Cell- and Skin-Penetrating Peptide. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2016. [DOI: 10.1007/s40883-016-0011-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
18
|
Li M, Tao Y, Shu Y, LaRochelle JR, Steinauer A, Thompson D, Schepartz A, Chen ZY, Liu DR. Discovery and Characterization of a Peptide That Enhances Endosomal Escape of Delivered Proteins in Vitro and in Vivo. J Am Chem Soc 2015; 137:14084-93. [DOI: 10.1021/jacs.5b05694] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Margie Li
- Department of Chemistry & Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| | - Yong Tao
- Department
of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02114, United States
| | - Yilai Shu
- Department
of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02114, United States
- Department
of Otology and Skull Base Survery, Eye, Ear, Nose and Throat Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China
- Key
Laboratory of Hearing Medicine, Ministry of Health, Shanghai, 200031, China
| | - Jonathan R. LaRochelle
- Department
of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Angela Steinauer
- Department
of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| | - David Thompson
- Department of Chemistry & Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| | - Alanna Schepartz
- Department
of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520-8107, United States
- Department
of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Zheng-Yi Chen
- Department
of Otolaryngology and Program in Neuroscience, Harvard Medical School and Eaton Peabody Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02114, United States
| | - David R. Liu
- Department of Chemistry & Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
- Howard
Hughes Medical Institute, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
19
|
Kamada H. [Development of New Biologics through Creation of a Functional Cytokine Mutant]. YAKUGAKU ZASSHI 2015; 135:843-9. [PMID: 26135083 DOI: 10.1248/yakushi.15-00007-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The clinical use of cytokines is fairly limited because of their characteristics of having significant bioactivity and low stability, although some are useful biopharmaceuticals, such as interferon. Cytokines, which are secreted from various immune cells, show many kinds of bioactivities including unexpected activities; thus it would be desirable to regulate cytokine activity. Recently, we have developed a new drug delivery system (DDS) to create structural mutant cytokines using a phage display system. This system can produce functional mutant proteins that can bind their objective targets specifically. In this study, tumor necrosis factor (TNF) was used as a model cytokine to create agonist and antagonist activities against two TNF receptors TNFR1 and TNFR2, respectively. We created a phage library expressing mutant TNF, where the amino acids in the binding interface between TNF and TNF receptors were alternately exchanged. Affinity panning was performed at the optimum condition and the bioactivities of these mutant TNFs were analyzed to obtain the objective agonists or antagonists. The pharmacological activity and toxicity of these engineered TNF mutants could indicate their potential use as novel biopharmaceutical agents.
Collapse
Affiliation(s)
- Haruhiko Kamada
- Laboratory of Biopharmaceutical Research, National Institutes of Biomedical Innovation, Health and Nutrition
| |
Collapse
|
20
|
A Heparan Sulfate-Binding Cell Penetrating Peptide for Tumor Targeting and Migration Inhibition. BIOMED RESEARCH INTERNATIONAL 2015; 2015:237969. [PMID: 26064887 PMCID: PMC4433633 DOI: 10.1155/2015/237969] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/31/2014] [Accepted: 11/14/2014] [Indexed: 11/18/2022]
Abstract
As heparan sulfate proteoglycans (HSPGs) are known as co-receptors to interact with numerous growth factors and then modulate downstream biological activities, overexpression of HS/HSPG on cell surface acts as an increasingly reliable prognostic factor in tumor progression. Cell penetrating peptides (CPPs) are short-chain peptides developed as functionalized vectors for delivery approaches of impermeable agents. On cell surface negatively charged HS provides the initial attachment of basic CPPs by electrostatic interaction, leading to multiple cellular effects. Here a functional peptide (CPPecp) has been identified from critical HS binding region in hRNase3, a unique RNase family member with in vitro antitumor activity. In this study we analyze a set of HS-binding CPPs derived from natural proteins including CPPecp. In addition to cellular binding and internalization, CPPecp demonstrated multiple functions including strong binding activity to tumor cell surface with higher HS expression, significant inhibitory effects on cancer cell migration, and suppression of angiogenesis in vitro and in vivo. Moreover, different from conventional highly basic CPPs, CPPecp facilitated magnetic nanoparticle to selectively target tumor site in vivo. Therefore, CPPecp could engage its capacity to be developed as biomaterials for diagnostic imaging agent, therapeutic supplement, or functionalized vector for drug delivery.
Collapse
|
21
|
Sharma G, Upadhyay S, Srilalitha M, Nandicoori VK, Khosla S. The interaction of mycobacterial protein Rv2966c with host chromatin is mediated through non-CpG methylation and histone H3/H4 binding. Nucleic Acids Res 2015; 43:3922-37. [PMID: 25824946 PMCID: PMC4417171 DOI: 10.1093/nar/gkv261] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 03/16/2015] [Indexed: 12/15/2022] Open
Abstract
To effectively modulate the gene expression within an infected mammalian cell, the pathogen Mycobacterium tuberculosis would need to bring about epigenetic modifications at appropriate genomic loci. Working on this hypothesis, we show in this study that the mycobacterial protein Rv2966c is a 5-methylcytosine-specific DNA methyltransferase that is secreted out from the mycobacterium and gets localized to the nucleus in addition to the cytoplasm inside the host cell. Importantly, Rv2966c binds to specific DNA sequences, methylates cytosines predominantly in a non-CpG context and its methylation activity is positively influenced by phosphorylation. Interestingly, like the mammalian DNA methyltransferase, DNMT3L, Rv2966c can also interact with histone proteins. Ours is the first study that identifies a protein from a pathogenic bacteria with potential to influence host DNA methylation in a non-canonical manner providing the pathogen with a novel mechanism to alter the host epigenetic machinery. This contention is supported by repression of host genes upon M. tuberculosis infection correlated with Rv2966c binding and non-CpG methylation.
Collapse
Affiliation(s)
- Garima Sharma
- Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad 500001, India Graduate Studies, Manipal University, Manipal 576104, India
| | | | - M Srilalitha
- Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad 500001, India
| | | | - Sanjeev Khosla
- Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad 500001, India
| |
Collapse
|
22
|
Kumar S, Zakrewsky M, Chen M, Menegatti S, Muraski JA, Mitragotri S. Peptides as skin penetration enhancers: Mechanisms of action. J Control Release 2015; 199:168-78. [DOI: 10.1016/j.jconrel.2014.12.006] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 11/22/2014] [Accepted: 12/08/2014] [Indexed: 11/28/2022]
|
23
|
Lee SH, Moroz E, Castagner B, Leroux JC. Activatable cell penetrating peptide-peptide nucleic acid conjugate via reduction of azobenzene PEG chains. J Am Chem Soc 2014; 136:12868-71. [PMID: 25185512 DOI: 10.1021/ja507547w] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The use of stimuli-responsive bioactive molecules is an attractive strategy to circumvent selectivity issues in vivo. Here, we report an activatable cell penetrating peptide (CPP) strategy ultimately aimed at delivering nucleic acid drugs to the colon mucosa using bacterial azoreductase as the local reconversion trigger. Through screening of a panel of CPPs, we identified a sequence (M918) capable of carrying a nucleic acid analogue payload. A modified M918 peptide conjugated to a peptide nucleic acid (PNA) was shown to silence luciferase in colon adenocarcinoma cells (HT-29-luc). Reversible functionalization of the conjugate's lysine residues via an azobenzene self-immolative linkage abolished transfection activity, and the free CPP-PNA was recovered after reduction of the azobenzene bond. This activatable CPP conjugate platform could find applications in the selective delivery of nucleic acid drugs to the colon mucosa, opening therapeutic avenues in colon diseases.
Collapse
Affiliation(s)
- Soo Hyeon Lee
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich (ETHZ) , Zurich 8093, Switzerland
| | | | | | | |
Collapse
|
24
|
Identification and characterization of a novel cell-penetrating peptide of 30Kc19 protein derived from Bombyx mori. Process Biochem 2014. [DOI: 10.1016/j.procbio.2014.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
25
|
Lee D, Pacheco S, Liu M. Biological effects of Tat cell-penetrating peptide: a multifunctional Trojan horse? Nanomedicine (Lond) 2014; 9:5-7. [PMID: 24354809 DOI: 10.2217/nnm.13.193] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Daiyoon Lee
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network; Institute of Medical Science, Faculty of Medicine, University of Toronto, ON, M5G 1L7, Canada
| | | | | |
Collapse
|
26
|
Favaro MTP, de Toledo MAS, Alves RF, Santos CA, Beloti LL, Janissen R, de la Torre LG, Souza AP, Azzoni AR. Development of a non-viral gene delivery vector based on the dynein light chain Rp3 and the TAT peptide. J Biotechnol 2014; 173:10-8. [PMID: 24417903 DOI: 10.1016/j.jbiotec.2014.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/20/2013] [Accepted: 01/02/2014] [Indexed: 02/02/2023]
Abstract
Gene therapy and DNA vaccination trials are limited by the lack of gene delivery vectors that combine efficiency and safety. Hence, the development of modular recombinant proteins able to mimic mechanisms used by viruses for intracellular trafficking and nuclear delivery is an important strategy. We designed a modular protein (named T-Rp3) composed of the recombinant human dynein light chain Rp3 fused to an N-terminal DNA-binding domain and a C-terminal membrane active peptide, TAT. The T-Rp3 protein was successfully expressed in Escherichia coli and interacted with the dynein intermediate chain in vitro. It was also proven to efficiently interact and condense plasmid DNA, forming a stable, small (∼100nm) and positively charged (+28.6mV) complex. Transfection of HeLa cells using T-Rp3 revealed that the vector is highly dependent on microtubule polarization, being 400 times more efficient than protamine, and only 13 times less efficient than Lipofectamine 2000™, but with a lower cytotoxicity. Confocal laser scanning microcopy studies revealed perinuclear accumulation of the vector, most likely as a result of transport via microtubules. This study contributes to the development of more efficient and less cytotoxic proteins for non-viral gene delivery.
Collapse
Affiliation(s)
- M T P Favaro
- Laboratório de Análise Genética e Molecular, Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - M A S de Toledo
- Laboratório de Análise Genética e Molecular, Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - R F Alves
- Departamento de Engenharia Química, Escola Politécnica, Universidade de São Paulo, São Paulo, SP, Brazil
| | - C A Santos
- Laboratório de Análise Genética e Molecular, Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - L L Beloti
- Laboratório de Análise Genética e Molecular, Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - R Janissen
- Instituto de Física Aplicada "Gleb Wataghin", Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - L G de la Torre
- Faculdade de Engenharia Química, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - A P Souza
- Laboratório de Análise Genética e Molecular, Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - A R Azzoni
- Departamento de Engenharia Química, Escola Politécnica, Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
27
|
Is there a future for cell-penetrating peptides in oligonucleotide delivery? Eur J Pharm Biopharm 2013; 85:5-11. [DOI: 10.1016/j.ejpb.2013.03.021] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 03/18/2013] [Accepted: 03/19/2013] [Indexed: 11/23/2022]
|
28
|
Stalmans S, Wynendaele E, Bracke N, Gevaert B, D’Hondt M, Peremans K, Burvenich C, De Spiegeleer B. Chemical-functional diversity in cell-penetrating peptides. PLoS One 2013; 8:e71752. [PMID: 23951237 PMCID: PMC3739727 DOI: 10.1371/journal.pone.0071752] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 07/03/2013] [Indexed: 12/13/2022] Open
Abstract
Cell-penetrating peptides (CPPs) are a promising tool to overcome cell membrane barriers. They have already been successfully applied as carriers for several problematic cargoes, like e.g. plasmid DNA and (si)RNA, opening doors for new therapeutics. Although several hundreds of CPPs are already described in the literature, only a few commercial applications of CPPs are currently available. Cellular uptake studies of these peptides suffer from inconsistencies in used techniques and other experimental conditions, leading to uncertainties about their uptake mechanisms and structural properties. To clarify the structural characteristics influencing the cell-penetrating properties of peptides, the chemical-functional space of peptides, already investigated for cellular uptake, was explored. For 186 peptides, a new cell-penetrating (CP)-response was proposed, based upon the scattered quantitative results for cellular influx available in the literature. Principal component analysis (PCA) and a quantitative structure-property relationship study (QSPR), using chemo-molecular descriptors and our newly defined CP-response, learned that besides typical well-known properties of CPPs, i.e. positive charge and amphipathicity, the shape, structure complexity and the 3D-pattern of constituting atoms influence the cellular uptake capacity of peptides.
Collapse
Affiliation(s)
- Sofie Stalmans
- Drug Quality and Registration (DruQuaR) Group, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Evelien Wynendaele
- Drug Quality and Registration (DruQuaR) Group, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Nathalie Bracke
- Drug Quality and Registration (DruQuaR) Group, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Bert Gevaert
- Drug Quality and Registration (DruQuaR) Group, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Matthias D’Hondt
- Drug Quality and Registration (DruQuaR) Group, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Kathelijne Peremans
- Department of Medical Imaging and Comparative Physiology and Biometrics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Christian Burvenich
- Department of Medical Imaging and Comparative Physiology and Biometrics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Bart De Spiegeleer
- Drug Quality and Registration (DruQuaR) Group, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
29
|
Duong HHP, Yung LYL. Synergistic co-delivery of doxorubicin and paclitaxel using multi-functional micelles for cancer treatment. Int J Pharm 2013; 454:486-95. [PMID: 23792465 DOI: 10.1016/j.ijpharm.2013.06.017] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 05/21/2013] [Accepted: 06/09/2013] [Indexed: 11/28/2022]
Abstract
The main purposes of this study are to demonstrate the synergistic anticancer drug systems with the combined doxorubicin (D) and paclitaxel (P) via the aid of cell penetrating and cell targeting moieties for enhancing the cancer therapeutic effect. Firstly, the synergistic effect of combined free drugs (D/P) was investigated to obtain the suitable dose combination for subsequent studies. The combination of free drugs D/P at molar ratio of 1/0.2 shows synergistic therapeutic effect compared with the treatment of a free single drug D or P. Secondly, sustainable release systems of two single drug-loaded micelles, (i) co-delivered D-FOL micelle & P-FOL micelle system and (ii) co-delivered D-TAT/FOL micelle & P-TAT/FOL micelle system, at D/P molar ratio of 1/0.2 were investigated. The results show synergistic effect with the higher efficacy of the TAT/FOL system compared to FOL only system. Finally, a dual D/P-loaded system with sustainable release rate, synergistic drug interaction, selective targeting to cancer cells and high cell penetrating ability was designed. The D/P-TAT/FOL micelles exhibit an IC50 value of 0.172 μM D/0.043 μM P, which is much lower than the IC50 values of the single drug-loaded micelles without functionalization (3.873 μM for D-micelles and 0.790 μM for P-micelles). Overall, this newly developed dual encapsulation of D and P in the multifunctional carrier would be a promising technology for cancer treatment.
Collapse
Affiliation(s)
- Hoang Hanh Phuoc Duong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117576, Singapore
| | | |
Collapse
|
30
|
Identification of novel signalling roles and targets for G(α) and G(βγ) downstream of the insulin-like growth factor 1 receptor in vascular smooth muscle cells. Biochem J 2013. [PMID: 23186281 DOI: 10.1042/bj20112158] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Vascular dysfunction is the underlying cause of nearly 80% of heart disease cases, and its initiation and progression can be exacerbated by circulating factors, such as IGF-1 (insulin-like growth factor 1). IGF-1, which is highly homologous with insulin, elicits a response via a classical tyrosine kinase receptor, the IGF-1R (IGF-1 receptor). However, it has been suggested that the IGF-1R may also be coupled to a heterotrimeric G-protein and can thus modulate cellular processes via this alternate pathway. The objective of the present study was to investigate the structural aspects of IGF-1R coupling to a heterotrimeric G-protein in VSMCs [vascular SMCs (smooth muscle cells)], as well as examine the contribution of this pathway to cellular responses that are related to vascular disease. We found that the intracellular subunit of the IGF-1R precipitates with two G-protein subunits. The G(βγ)-mediated pathway contributes to both proliferation and migration. We also show that IGF-1 specifically activates G(αi) and can directly interact with both G(αi1) and G(αi2). A phospho-screen using a novel specific G(αi)-peptide inhibitor reveals a number of potential downstream effectors of this pathway, although our results show that it is not essential for SMC proliferation or migration.
Collapse
|
31
|
Mellert K, Lamla M, Scheffzek K, Wittig R, Kaufmann D. Enhancing endosomal escape of transduced proteins by photochemical internalisation. PLoS One 2012; 7:e52473. [PMID: 23285056 PMCID: PMC3528648 DOI: 10.1371/journal.pone.0052473] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 11/14/2012] [Indexed: 12/22/2022] Open
Abstract
Induced internalisation of functional proteins into cultured cells has become an important aspect in a rising number of in vitro and in vivo assays. The endo-lysosomal entrapment of the transduced proteins remains the major problem in all transduction protocols. In this study we compared the efficiency, cytotoxicity and protein targeting of different commercially available transduction reagents by transducing a well-studied fluorescently labelled protein (Atto488-bovine serum albumin) into cultured human sarcoma cells. The amount of internalised protein and toxicity differed between the different reagents, but the percentage of transduced cells was consistently high. Furthermore, in all protocols the signals of the transduced Atto488-BSA were predominantly punctual consistent with an endosomal localisation. To overcome the endosomal entrapment, the transduction protocols were combined with a photochemical internalisation (PCI) treatment. Using this combination revealed that an endosomal disruption is highly effective in cell penetrating peptide (CPP) mediated transduction, whereas lipid-mediated transductions lead to a lower signal spreading throughout the cytosol. No change in the signal distribution could be achieved in treatments using non-lipid polymers as a transduction reagent. Therefore, the combination of protein transduction protocols based on CPPs with the endosomolytic treatment PCI can facilitate protein transduction experiments in vitro.
Collapse
Affiliation(s)
- Kevin Mellert
- Institute of Human Genetics, University of Ulm, Ulm, Germany.
| | | | | | | | | |
Collapse
|
32
|
Salomone F, Cardarelli F, Di Luca M, Boccardi C, Nifosì R, Bardi G, Di Bari L, Serresi M, Beltram F. A novel chimeric cell-penetrating peptide with membrane-disruptive properties for efficient endosomal escape. J Control Release 2012; 163:293-303. [DOI: 10.1016/j.jconrel.2012.09.019] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2012] [Revised: 09/21/2012] [Accepted: 09/24/2012] [Indexed: 11/30/2022]
|
33
|
Liou JS, Liu BR, Martin AL, Huang YW, Chiang HJ, Lee HJ. Protein transduction in human cells is enhanced by cell-penetrating peptides fused with an endosomolytic HA2 sequence. Peptides 2012; 37:273-84. [PMID: 22898256 PMCID: PMC9616647 DOI: 10.1016/j.peptides.2012.07.019] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 07/21/2012] [Accepted: 07/23/2012] [Indexed: 11/21/2022]
Abstract
Endocytosis has been proposed as one of the primary mechanisms for cellular entry of cell-penetrating peptides (CPPs) and their cargoes. However, a major limitation of endocytic pathway is entrapment of the CPP-cargo in intracellular vesicles from which the cargo must escape into the cytoplasm to exert its biological activity. Here we demonstrate that a CPP tagged with an endosomolytic fusion peptide derived from the influenza virus hemagglutinin-2 (HA2) remarkably enhances the cytosolic delivery of proteins in human A549 cells. To determine the endosome-disruptive effects, recombinant DNA plasmids containing coding sequences of HA2, CPPs and red fluorescent proteins (RFPs) were constructed. The fusion proteins were purified from plasmid-transformed Escherichia coli, and their effects on protein transduction were examined using live cell imaging and flow cytometry. Our data indicate that endocytosis is the major route for cellular internalization of CPP-HA2-tagged RFP. Mechanistic studies revealed that the fusogenic HA2 peptide dramatically facilitates CPP-mediated protein entry through the release of endocytosed RFPs from endosomes into the cytoplasm. Furthermore, incorporating the HA2 fusion peptide of the CPP-HA2 fusion protein improved cytosolic uptake without causing cytotoxicity. These findings strongly suggest that the CPP-HA2 tag could be an efficient and safe carrier that overcomes endosomal entrapment of delivered therapeutic drugs.
Collapse
Affiliation(s)
- Ji-Sing Liou
- Department of Natural Resources and Environmental Studies, National Dong Hwa University, No. 1, Sec. 2, Da-Hsueh Road, Shoufeng, Hualien 97401, Taiwan
- Graduate Institute of Biotechnology, National Dong Hwa University, No. 1, Sec. 2, Da-Hsueh Road, Shoufeng, Hualien 97401, Taiwan
| | - Betty Revon Liu
- Department of Natural Resources and Environmental Studies, National Dong Hwa University, No. 1, Sec. 2, Da-Hsueh Road, Shoufeng, Hualien 97401, Taiwan
- Graduate Institute of Biotechnology, National Dong Hwa University, No. 1, Sec. 2, Da-Hsueh Road, Shoufeng, Hualien 97401, Taiwan
| | - Adam L. Martin
- Department of Biological Sciences and the cDNA Resources Center, Missouri University of Science and Technology, 206 Schrenk Hall, 400 West 11th Street, Rolla, MO 65409-1120, USA
| | - Yue-Wern Huang
- Department of Biological Sciences, Missouri University of Science and Technology, 105 Schrenk Hall, 400 West 11th Street, Rolla, MO 65409-1120, USA
| | - Huey-Jenn Chiang
- Graduate Institute of Biotechnology, National Dong Hwa University, No. 1, Sec. 2, Da-Hsueh Road, Shoufeng, Hualien 97401, Taiwan
| | - Han-Jung Lee
- Department of Natural Resources and Environmental Studies, National Dong Hwa University, No. 1, Sec. 2, Da-Hsueh Road, Shoufeng, Hualien 97401, Taiwan
| |
Collapse
|
34
|
Jones AT, Sayers EJ. Cell entry of cell penetrating peptides: tales of tails wagging dogs. J Control Release 2012; 161:582-91. [DOI: 10.1016/j.jconrel.2012.04.003] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 04/02/2012] [Indexed: 02/02/2023]
|
35
|
Guo Q, Zhao G, Hao F, Guan Y. Effects of the TAT peptide orientation and relative location on the protein transduction efficiency. Chem Biol Drug Des 2012; 79:683-90. [PMID: 22188730 DOI: 10.1111/j.1747-0285.2011.01315.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
To understand the protein transduction domain (PTD)-mediated protein transduction behavior and to explore its potential in delivering biopharmaceutic drugs, we prepared four TAT-EGFP conjugates: TAT(+)-EGFP, TAT(-)-EGFP, EGFP-TAT(+) and EGFP-TAT(-), where TAT(+) and TAT(-) represent the original and the reversed TAT sequence, respectively. These four TAT-EGFP conjugates were incubated with HeLa and PC12 cells for in vitro study as well as injected intraperitoneally to mice for in vivo study. Flow cytometric results showed that four TAT-EGFP conjugates were able to traverse HeLa and PC12 cells with almost equal transduction efficiency. The in vivo study showed that the TAT-EGFP conjugates could be delivered into different organs of mice with different transduction capabilities. Bioinformatic analyses and CD spectroscopic data revealed that the TAT peptide has no defined secondary structure, and conjugating the TAT peptide to the EGFP cargo protein would not alter the native structure and the function of the EGFP protein. These results conclude that the sequence orientation, the spatial structure, and the relative location of the TAT peptide have much less effect on the TAT-mediated protein transduction. Thus, the TAT-fused conjugates could be constructed in more convenient and flexible formats for a wide range of biopharmaceutical applications.
Collapse
Affiliation(s)
- Qingguo Guo
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | | | | | | |
Collapse
|
36
|
Efficient protein transduction method using cationic peptides and lipids. J Biomed Biotechnol 2011; 2011:872065. [PMID: 22007148 PMCID: PMC3189636 DOI: 10.1155/2011/872065] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 07/23/2011] [Accepted: 08/10/2011] [Indexed: 11/17/2022] Open
Abstract
Cationic peptides termed protein transduction domains (PTDs) have been shown to cross biological membranes efficiently. However, proteins transduced by PTDs become entrapped within the endosomal vesicles and are not delivered into organelles. We have developed a novel protein delivery system to enhance the proton sponge effect, which results in rupture of the endosomes, by using a mixture of Wr-T transporter peptide and a commercially available cationic lipid reagent. This peptide and cationic lipid reagent mixture efficiently delivers a variety of cargo proteins into living cells by releasing them from the endosomes.
Collapse
|
37
|
Papadopoulou LC, Tsiftsoglou AS. Transduction of human recombinant proteins into mitochondria as a protein therapeutic approach for mitochondrial disorders. Pharm Res 2011; 28:2639-56. [PMID: 21874377 DOI: 10.1007/s11095-011-0546-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 07/21/2011] [Indexed: 01/10/2023]
Abstract
Protein therapy is considered an alternative approach to gene therapy for treatment of genetic-metabolic disorders. Human protein therapeutics (PTs), developed via recombinant DNA technology and used for the treatment of these illnesses, act upon membrane-bound receptors to achieve their pharmacological response. On the contrary, proteins that normally act inside the cells cannot be developed as PTs in the conventional way, since they are not able to "cross" the plasma membrane. Furthermore, in mitochondrial disorders, attributed either to depleted or malfunctioned mitochondrial proteins, PTs should also have to reach the subcellular mitochondria to exert their therapeutic potential. Nowadays, there is no effective therapy for mitochondrial disorders. The development of PTs, however, via the Protein Transduction Domain (PTD) technology offered new opportunities for the deliberate delivery of human recombinant proteins inside eukaryotic subcellular organelles. To this end, mitochondrial disorders could be clinically encountered with the delivery of human mitochondrial proteins (engineered via recombinant DNA and PTD technologies) at specific intramitochondrial sites to exert their function. Overall, PTD-mediated Protein Replacement Therapy emerges as a suitable model system for the therapeutic approach for mitochondrial disorders.
Collapse
Affiliation(s)
- Lefkothea C Papadopoulou
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki, Thessaloniki, GR54124, Macedonia, Greece.
| | | |
Collapse
|
38
|
Rayapureddi JP, Tomamichel WJ, Walton ST, Payne RM. TAT fusion protein transduction into isolated mitochondria is accelerated by sodium channel inhibitors. Biochemistry 2011; 49:9470-9. [PMID: 20925426 DOI: 10.1021/bi101057v] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Stringent control of ion and protein transport across the mitochondrial membranes is required to maintain mitochondrial function and biogenesis. In particular, the inner mitochondrial membrane is generally impermeable to proteins entering the matrix except via tightly regulated protein import mechanisms. Recently, cell penetrant peptides have been shown to move across the inner mitochondrial membrane in a manner suggesting an independent mechanism. HIV-1 transactivator of transcription (TAT) is an arginine-rich cell penetrant peptide, 47YGRKKRRQRRR57, which can transduce full-length proteins not only across the cell membrane but also into intracellular organelles. In this study, we investigated the ability of a TAT-containing protein to move into the mitochondrial matrix. Using a novel FACS assay for isolated, purified mitochondria, we show that TAT can deliver a modified fluorescent protein, mMDH-GFP, to the matrix of mitochondria and it is subsequently processed by the matrix peptidases. In addition, transduction of TAT-mMDH-GFP into mitochondria is independent of canonical protein import pathways as well as mitochondrial membrane potential. In direct contrast to published reports regarding the cell membrane where the sodium channel inhibitor, amiloride, blocks endocytosis and inhibits TAT transduction, TAT transduction into mitochondria is markedly increased by this same sodium channel inhibitor. These results confirm that the cell penetrant peptide, TAT, can readily transduce a protein cargo into the mitochondrial matrix. These results also demonstrate a novel role for mitochondrial sodium channels in mediating TAT transduction into mitochondria that is independent of endocytotic mechanisms. The mechanism of TAT transduction into mitochondria therefore is distinctly different from transduction across the cell membrane.
Collapse
|
39
|
McQueen P, Donald LJ, Vo TN, Nguyen DH, Griffiths H, Shojania S, Standing KG, O'Neil JD. Tat peptide-calmodulin binding studies and bioinformatics of HIV-1 protein-calmodulin interactions. Proteins 2011; 79:2233-46. [DOI: 10.1002/prot.23048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 03/18/2011] [Accepted: 03/22/2011] [Indexed: 01/08/2023]
|
40
|
Nakase I, Kobayashi S, Futaki S. Endosome-disruptive peptides for improving cytosolic delivery of bioactive macromolecules. Biopolymers 2011; 94:763-70. [PMID: 20564044 DOI: 10.1002/bip.21487] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Along with recent advances in therapeutic technologies based on biomacromolecules, including genes, oligonucleotides, and proteins, the development of technologies for improving the efficiency of the delivery of these therapeutic molecules into cells, more specifically into the cytosol and nucleus, is significantly required. Cell membranes are major impediments to the delivery of therapeutic macromolecules into cells. These macromolecules are usually taken up by the cells via endocytosis, and their translocation from endosomes to the cytosol is a critical step to determine their therapeutic effects. Many viruses and bacterial toxins use endocytic pathways to invade the host mammalian cells, and some of these pathogens have the ability to facilitate their endosomal escape into the cytosol by pH-induced alteration in their component proteins that leads to the disruption of the endosomal membranes and the eventual membrane fusions. To simulating these functions, endosome-disruptive peptides have been used for the intracellular delivery of biomacromolecules to accelerate their endosomal escape by sensing the endosomal acidification. In this review, current approaches for the intracellular delivery using these endosome-disruptive peptides are surveyed.
Collapse
Affiliation(s)
- Ikuhiko Nakase
- Institute for Chemical Research, Kyoto University, Kyoto 611-0011, Japan
| | | | | |
Collapse
|
41
|
Duvall CL, Convertine AJ, Benoit DSW, Hoffman AS, Stayton PS. Intracellular delivery of a proapoptotic peptide via conjugation to a RAFT synthesized endosomolytic polymer. Mol Pharm 2010; 7:468-76. [PMID: 19968323 DOI: 10.1021/mp9002267] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Peptides derived from the third B-cell lymphoma 2 (Bcl-2) homology domain (BH3) can heterodimerize with antiapoptotic Bcl-2 family members to block their activity and trigger apoptosis. Use of these peptides presents a viable anticancer approach, but delivery barriers limit the broad application of intracellular-acting peptides as clinical therapeutics. Here, a novel diblock copolymer carrier is described that confers desirable pharmaceutical properties to intracellular-acting therapeutic peptides through site-specific molecular conjugation. This polymer was prepared using reversible addition-fragmentation chain transfer (RAFT) to form a pyridyl disulfide end-functionalized, modular diblock copolymer with precisely controlled molecular weight (M(n)) and low polydispersity (PDI). The diblock polymer (M(n) 19,000 g/mol, PDI 1.27) was composed of an N-(2-hydroxypropyl) methacrylamide (HPMA) first block (M(n) 13,800 g/mol, PDI 1.13) intended to enhance water solubility and circulation time. The second polymer block was a pH-responsive composition designed to enhance endosomal escape and consisted of equimolar quantities of dimethylaminoethyl methacrylate (DMAEMA), propylacrylic acid (PAA), and butyl methacrylate (BMA). A hemolysis assay indicated that the diblock polymer undergoes a physiologically relevant pH-dependent switch from a membrane inert (1% hemolysis, pH 7.4) to a membrane disruptive (61% hemolysis, pH 5.8) conformation. Thiol-disulfide exchange reactions were found to efficiently produce reversible polymer conjugates (75 mol % peptide reactivity with polymer) with a cell-internalized proapoptotic peptide. Microscopy studies showed that peptide delivered via polymer conjugates effectively escaped endosomes and achieved diffusion into the cytosol. Peptide-polymer conjugates also produced significantly increased apoptotic activity over peptide alone in HeLa cervical carcinoma cells as found using flow cytometric measurements of mitochondrial membrane depolarization (2.5-fold increase) and cell viability tests that showed 50% cytotoxicity after 6 h of treatment with 10 muM peptide conjugate. These results indicate that this multifunctional carrier shows significant promise for proapoptotic peptide cancer therapeutics and also as a general platform for delivery of peptide drugs with intracellular targets.
Collapse
Affiliation(s)
- Craig L Duvall
- Department of Bioengineering, University of Washington, Seattle Washington 98195, USA
| | | | | | | | | |
Collapse
|
42
|
Lim KS, Won YW, Park YS, Kim YH. Preparation and functional analysis of recombinant protein transduction domain-metallothionein fusion proteins. Biochimie 2010; 92:964-70. [PMID: 20403412 DOI: 10.1016/j.biochi.2010.04.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Accepted: 04/08/2010] [Indexed: 11/16/2022]
Abstract
In order for proteins to be used as pharmaceuticals, delivery technologies need to be developed to overcome biochemical and anatomical barriers to protein drug transport, to protect proteins from systemic degradation, and to target the drug action to specific sites. Protein transduction domains (PTDs) are used for the non-specific transduction of bio-active cargo, such as proteins, genes, and particles, through cellular membranes to overcome biological barriers. Metallothionein (MT) is a low molecular weight intra-cellular protein that consists of 61 amino acids, including 20 cysteine residues, and is over-expressed under stressful conditions. Although MT has the potential to improve the viability of islet cells and cardiomyocytes by inhibiting diabetic-induced apoptosis and by removing reactive oxygen species (ROS), and thereby prevent or reduce diabetes and diabetic complications, all MT applications have been made for gene therapy or under induced over-expression of endogenous MT. To overcome the drawbacks of ineffective intra-cellular MT protein uptake, a human MT gene was cloned and fused with protein transduction domains (PTDs), such as HIV-1 Tat and undeca-arginine, in a bacterial expression vector to produce PTD-MT fusion proteins. The expression and purification of three types of proteins were optimized by adding Zn ions to maintain their stability and functionality mimicking intra-cellular stable conformation of MT as a Zn-MT cluster. The Zn-MT cluster showed better stability than MT in vitro. PTD-MT fusion proteins strongly protected Ins-1 beta cells against oxidative stress and apoptosis induced by glucolipotoxicity with or without hypoxia, and also protected H9c2 cardiomyocytes against hyperglycemia-induced apoptosis with or without hypoxia. PTD-MT recombinant fusion proteins may be useful protein therapeutics for the treatment or prevention of diabetes and diabetes-related complications.
Collapse
Affiliation(s)
- Kwang Suk Lim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seongdong-gu, Seoul 133-791, Republic of Korea
| | | | | | | |
Collapse
|
43
|
Rafat M, Cléroux CA, Fong WG, Baker AN, Leonard BC, O'Connor MD, Tsilfidis C. PEG-PLA microparticles for encapsulation and delivery of Tat-EGFP to retinal cells. Biomaterials 2010; 31:3414-21. [PMID: 20149443 DOI: 10.1016/j.biomaterials.2010.01.031] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Accepted: 01/09/2010] [Indexed: 12/14/2022]
Abstract
The efficient and controlled delivery of genes and proteins to retinal cells remains a challenge. In this study, we evaluated polyethylene glycol-polylactic acid (PEG-PLA) microparticles for encapsulation and delivery of a Transactivator of transcription-enhanced green fluorescent protein fusion (Tat-EGFP) to retinal cells. Our main objective was to develop a microparticle system that delivers Tat-EGFP with an initial rapid release (within 24 h) followed by a sustained release. We prepared four different formulations of Tat-EGFP encapsulated PEG-PLA particles to investigate the effects of protein and polymer concentrations on particle morphology and protein release, using scanning electron microscopy (SEM) and fluorometry techniques. The optimum formulation was selected based on higher protein release, and smaller particle size. The optimum formulation was then tested in vitro for cell biocompatibility and protein internalization, and in vivo for cellular toxicity following sub-retinal injections into rat eyes. The results suggest that PEG-PLA microparticles can deliver proteins in cell culture allowing protein internalization in as little as 1 h. In vivo, protein was shown to localize within the photoreceptor layer of the retina, and persist for at least 9 weeks with no observed toxicity.
Collapse
Affiliation(s)
- Mehrdad Rafat
- Ottawa Hospital Research Institute, Ottawa Hospital, General Division, Ottawa, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
44
|
Khaja K, Robbins P. Comparison of Functional Protein Transduction Domains Using the NEMO Binding Domain Peptide. Pharmaceuticals (Basel) 2010; 3:110-124. [PMID: 27713244 PMCID: PMC3991022 DOI: 10.3390/ph3010110] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2009] [Revised: 12/22/2009] [Accepted: 12/24/2009] [Indexed: 12/23/2022] Open
Abstract
Protein transduction domains (PTDs), both naturally occurring and synthetic, have been extensively utilized for intracellular delivery of biologically active molecules both in vitro and in vivo. However, most comparisons of transduction efficiency have been performed using fluorescent markers. To compare efficiency of functional protein transduction, a peptide derived from IκB kinase ß (IKKß) that prevents formation of an active IKK complex was used as a biologically active cargo. This peptide, termed NEMO Binding Domain (NBD), is able to block activation of the transcriptional factor NF-κB by IKK, but not basal NF-κB activity. Our results demonstrate that Antp and Tat PTDs were most effective for delivery of NBD for inhibition of NF-κB activation compared to other PTD-NBD in both Hela and 293 cells, however, at higher concentrations (100 µM), the Antp-NBD as well as the FGF-NBD peptide caused significant cellular toxicity. In contrast to the cell culture results, delivery of NBD using 8K (octalysine) and 6R (six arginine) were the most effect in blocking inflammation following local, footpad delivery in a KLH-induced DTH murine model of inflammatory arthritis. These results demonstrate differences between PTDs for delivery of a functional cargo between cell types.
Collapse
Affiliation(s)
- Khaleel Khaja
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Paul Robbins
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
45
|
Cell-penetrating DNA-binding protein as a safe and efficient naked DNA delivery carrier in vitro and in vivo. Biochem Biophys Res Commun 2010; 392:9-15. [DOI: 10.1016/j.bbrc.2009.12.135] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Accepted: 12/23/2009] [Indexed: 11/21/2022]
|
46
|
Kwon SJ, Han K, Jung S, Lee JE, Park S, Cheon YP, Lim HJ. Transduction of the MPG-tagged fusion protein into mammalian cells and oocytes depends on amiloride-sensitive endocytic pathway. BMC Biotechnol 2009; 9:73. [PMID: 19706197 PMCID: PMC2748071 DOI: 10.1186/1472-6750-9-73] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Accepted: 08/26/2009] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND MPG is a cell-permeable peptide with proven efficiency to deliver macromolecular cargoes into cells. In this work, we examined the efficacy of MPG as an N-terminal tag in a fusion protein to deliver a protein cargo and its mechanism of transduction. RESULTS We examined transduction of MPG-EGFP fusion protein by live imaging, flow cytometry, along with combination of cell biological and pharmacological methods. We show that MPG-EGFP fusion proteins efficiently enter various mammalian cells within a few minutes and are co-localized with FM4-64, a general marker of endosomes. The transduction of MPG-EGFP occurs rapidly and is inhibited at a low temperature. The entry of MPG-EGFP is inhibited by amiloride, but cytochalasin D and methyl-beta-cyclodextrin did not inhibit the entry, suggesting that macropinocytosis is not involved in the transduction. Overexpression of a mutant form of dynamin partially reduced the transduction of MPG-EGFP. The partial blockade of MPG-EGFP transduction by a dynamin mutant is abolished by the treatment of amiloride. MPG-EGFP transduction is also observed in the mammalian oocytes. CONCLUSION The results show that the transduction of MPG fusion protein utilizes endocytic pathway(s) which is amiloride-sensitive and partially dynamin-dependent. Collectively, the MPG fusion protein could be further developed as a novel tool of "protein therapeutics", with potentials to be used in various cell systems including mammalian oocytes.
Collapse
Affiliation(s)
- So-Jung Kwon
- Department of Biomedical Science & Technology IBST Konkuk University 1 Hwayang-dong, Kwangjin-gu, Seoul 143-701, Korea.
| | | | | | | | | | | | | |
Collapse
|
47
|
Zhang C, Qin A, Chen H, Deng X, Su Y, Qian K. MDV-1 VP22: a transporter that can selectively deliver proteins into cells. Arch Virol 2009; 154:1027-34. [PMID: 19495935 DOI: 10.1007/s00705-009-0404-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2008] [Accepted: 05/18/2009] [Indexed: 11/30/2022]
Abstract
The tegument protein VP22 of Marek's disease virus (MDV) was previously shown to be able to travel between cells. To further characterize the transport property of VP22 and assess whether it can be used for protein delivery, we investigated the subcellular localization of VP22 fused to five heterologous proteins, including green fluorescent protein, nucleoprotein of avian influenza virus, bovine IFN-gamma, F protein of Newcastle disease virus and VP2 protein of infectious bursa disease virus. The transport of these fusion proteins in monolayer cells was assayed by immunofluorescence assay. The results showed that all except VP2 could be delivered by VP22 of MDV serotype 1 (MDV-1), at different efficiencies. After being transported to surrounding cells, VP22 fused to avian influenza nucleoprotein, bovine IFN-gamma, or to F was localized in the nucleus. Our data suggest that VP22 of MDV-1 can be used as transport tool in protein delivery and that the original localization of cargo proteins may be changed after transport by MDV-1 VP22. Finally, infectious bursa disease VP2 protein could not be transported by MDV-1 VP22.
Collapse
|
48
|
Yoshikawa T, Sugita T, Mukai Y, Abe Y, Nakagawa S, Kamada H, Tsunoda SI, Tsutsumi Y. The augmentation of intracellular delivery of peptide therapeutics by artificial protein transduction domains. Biomaterials 2009; 30:3318-23. [PMID: 19304319 DOI: 10.1016/j.biomaterials.2009.02.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Accepted: 02/20/2009] [Indexed: 10/21/2022]
Abstract
Protein transduction domains (PTDs), such as HIV-derived Tat, have been successfully used as functional biomaterials for intracellular delivery of anti-cancer macromolecular drugs (protein, peptides, and oligonucleotides). Although there were therefore great expectations regarding the therapeutic potential of PTDs for the development of anti-cancer therapeutics, their clinical application so far has been extremely limited because of the relatively high concentrations required to mediate any effects on cancer cells in vitro or in vivo. In this context, improving the transduction efficiency of PTDs using phage display-based molecular evolution techniques may be useful for creating artificial PTDs with high efficiency and safety. Here, we report an evaluation of transduction efficiency and toxicity of such artificial PTDs (designated mT02 and mT03) compared with Tat. The internalization of mT02 was the most rapid and efficient by a mechanism different from the usual macropinocytosis. Furthermore, we found that artificial PTDs fused with survivin antagonistic peptide potentiate tumor cell-cytostatic activity. Thus, the results of this work provide new insights for designing new-generation peptide therapeutics for a wide variety of cancers as well as those expressing survivin.
Collapse
Affiliation(s)
- Tomoaki Yoshikawa
- Laboratory of Pharmaceutical Proteomics, National Institute of Biomedical Innovation, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Direct cell entry of gold/iron-oxide magnetic nanoparticles in adenovirus mediated gene delivery. Biomaterials 2009; 30:1809-14. [PMID: 19136151 DOI: 10.1016/j.biomaterials.2008.12.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Accepted: 12/07/2008] [Indexed: 11/21/2022]
Abstract
Gold/iron-oxide MAgnetic Nanoparticles (GoldMAN) imparts useful magnetic properties to various biomolecules. Gold nanoparticles immobilized on the surface of magnetic nanoparticles allow for the conjugation of biomolecules via an Au-S bond. Here, we present a practical application by utilizing GoldMAN and a magnetic field to induce intracellular transduction. This method has great potential for application of the adenovirus gene delivery vector (Ad), widely used for in vitro/in vivo gene transfer, to Ad-resistant cells. We demonstrated that Ad was easily immobilized on GoldMAN and the Ad/GoldMAN complex was introduced into the cell by the magnetic field, which increased gene expression over 1000 times that of Ad alone. The GoldMAN penetrated the plasma membrane directly, independent of the cell-surface virus receptors and endocytosis pathway. This mechanism will contribute to improve the gene expression efficiency of Ad. This technology is a useful tool for extending Ad tropism and enhancing transduction efficiency. GoldMAN also makes possible the effective use of various biomolecules within the cell because of its interesting cell-entry mechanism.
Collapse
|
50
|
El-Sayed A, Futaki S, Harashima H. Delivery of macromolecules using arginine-rich cell-penetrating peptides: ways to overcome endosomal entrapment. AAPS JOURNAL 2009; 11:13-22. [PMID: 19125334 DOI: 10.1208/s12248-008-9071-2] [Citation(s) in RCA: 374] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/29/2008] [Accepted: 12/07/2008] [Indexed: 11/30/2022]
Abstract
Arginine-rich cell-penetrating peptides (AR-CPPs) are very promising tools for the delivery of therapeutic macromolecules such as peptides, proteins, and nucleic acids. These peptides allow efficient internalization of the linked cargos intracellularly through the endocytic pathway. However, when linked to bulky cargos, entrapment in the endocytic vesicles is a major limitation to the application of these peptides in cytosolic delivery. Attachment of a compatible endosomal escape device is, therefore, necessary to allow cytosolic delivery of the peptide-attached cargo. This review presents different endosomal escape devices currently in application in combination with AR-CPPs. Applications of fusogenic lipids, membrane-disruptive peptides, membrane-disruptive polymers, lysosomotropic agents, and photochemical internalization to enhance the cytosolic delivery of AR-CPPs-attached cargos are presented. The properties of each system and its mechanism of action for the enhancement of endosomal escape are discussed, together with its applications for the delivery of different macromolecules in vitro and, if applicable, in vivo.
Collapse
Affiliation(s)
- Ayman El-Sayed
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 Nishi 6, Kita-Ku, Sapporo, Hokkaido, 060-0812, Japan
| | | | | |
Collapse
|