1
|
Li Z, Dang Q, Wang P, Zhao F, Huang J, Wang C, Liu X, Min W. Food-Derived Peptides: Beneficial CNS Effects and Cross-BBB Transmission Strategies. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:20453-20478. [PMID: 38085598 DOI: 10.1021/acs.jafc.3c06518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Food-derived peptides, as dietary supplements, have significant effects on promoting brain health and relieving central nervous system (CNS) diseases. However, the blood-brain barrier (BBB) greatly limits their in-brain bioavailability. Thus, overcoming the BBB to target the CNS is a major challenge for bioactive peptides in the prevention and treatment of CNS diseases. This review discusses improvement in the neuroprotective function of food-derived active peptides in CNS diseases, as well as the source of BBB penetrating peptides (BBB-shuttles) and the mechanism of transmembrane transport. Notably, this review also discusses various peptide modification methods to overcome the low permeability and stability of the BBB. Lipification, glycosylation, introduction of disulfide bonds, and cyclization are effective strategies for improving the penetration efficiency of peptides through the BBB. This review provides a new prospective for improving their neuroprotective function and developing treatments to delay or even prevent CNS diseases.
Collapse
Affiliation(s)
- Zehui Li
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
- College of Food Science and Engineering, Jilin Agricultural University, ChangChun, Jilin 130118, P.R. China
| | - Qiao Dang
- College of Food Science and Engineering, Jilin Agricultural University, ChangChun, Jilin 130118, P.R. China
| | - Peng Wang
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
| | - Fanrui Zhao
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P.R. China
| | - Jianqin Huang
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P.R. China
| | - Chongchong Wang
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P.R. China
| | - Xingquan Liu
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
| | - Weihong Min
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P.R. China
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P.R. China
| |
Collapse
|
2
|
Arunmanee W, Duangkaew M, Taweecheep P, Aphicho K, Lerdvorasap P, Pitchayakorn J, Intasuk C, Jiraratmetacon R, Syamsidi A, Chanvorachote P, Chaotham C, Pornputtapong N. Resurfacing receptor binding domain of Colicin N to enhance its cytotoxic effect on human lung cancer cells. Comput Struct Biotechnol J 2021; 19:5225-5234. [PMID: 34630940 PMCID: PMC8479544 DOI: 10.1016/j.csbj.2021.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/05/2021] [Accepted: 09/08/2021] [Indexed: 11/24/2022] Open
Abstract
Colicin N (ColN) is a bacteriocin secreted by Escherichia coli (E. coli) to kill other Gram-negative bacteria by forcefully generating ion channels in the inner membrane. In addition to its bactericidal activity, ColN have been reported to selectively induce apoptosis in human lung cancer cells via the suppression of integrin modulated survival pathway. However, ColN showed mild toxicity against human lung cancer cells which could be improved for further applications. The protein resurfacing strategy was chosen to engineer ColN by extensive mutagenesis at solvent-exposed residues on ColN. The highly accessible Asp and Glu on wildtype ColN (ColNWT) were replaced by Lys to create polycationic ColN (ColN+12). Previous studies have shown that increase of positive charges on proteins leads to the enhancement of mammalian cell penetration as well as increased interaction with negatively charged surface of cancer cells. Those solvent-exposed residues of ColN were identified by Rosetta and AvNAPSA (Average number of Neighboring Atoms Per Sidechain Atom) approaches. The findings revealed that the structural features and stability of ColN+12 determined by circular dichroism were similar to ColNWT. Furthermore, the toxicity of ColN+12 was cancer selective. Human lung cancer cells, H460 and H23, were sensitive to ColN but human dermal papilla cells were not. ColN+12 also showed more potent toxicity than ColNWT in cancer cells. This confirmed that polycationic resurfacing method has enabled us to improve the anticancer activity of ColN towards human lung cancer cells.
Collapse
Affiliation(s)
- Wanatchaporn Arunmanee
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Methawee Duangkaew
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pornchanok Taweecheep
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kanokpol Aphicho
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Panuwat Lerdvorasap
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Jesada Pitchayakorn
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chayada Intasuk
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Runglada Jiraratmetacon
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Armini Syamsidi
- Department of Pharmacy, Faculty of Science, Tadulako University, Central Sulawesi 94118, Indonesia
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chatchai Chaotham
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Natapol Pornputtapong
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Corresponding author.
| |
Collapse
|
3
|
Taghizadegan N, Firoozrai M, Nassiri M, Ariannejad H. A novel strategy for engineering of a smart generation of immune ribonucleases against EGFR + cells. J Cell Physiol 2021; 236:4303-4312. [PMID: 33421131 DOI: 10.1002/jcp.30118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 10/04/2020] [Accepted: 10/06/2020] [Indexed: 12/26/2022]
Abstract
The overexpression of epidermal growth factor receptor (EGFR) could result in the development of solid tumors of prostate, breast, gastric, colorectal, ovarian, and head and neck, leading to carcinoma. Antibody therapies are ideal methods to overcome malignant diseases. However, immunoribonucleases are a new generation of antibodies in which an RNase binds to a specific antibody and shows a stronger ability to terminate cancer cells. In this study, we engineered Rana pipiens RNase to bind to the scFv of human antiepidermal growth factor receptor antibody. The molecular dynamic simulations confirmed protein stability and the ability of scFv-ranpirnase (rantoxin) to bind to epidermal growth factor receptor protein. Then, the rantoxin construct was synthesized in a pCDNA 3.1 Neo vector. CHO-K1 cells were used as expression hosts and the construct was transfected. Cells were selected by antibiotic therapies using neomycin, 120 mg/ml, and the high-yield colony was screened by real-time polymerase chain reaction (PCR) methods. Then, the recombinant protein production was confirmed using the sodium dodecyl sulfate polyacrylamide gel electrophoresis and western blot analyses. The molecular dynamic simulation (MDS) confirmed that the I467, S468, Q408, and H409 amino acids of EGFR bonded well to rantoxin. As revealed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and western blot analyses, the rantoxin production and PCR analysis showed that the T3 colony can produce rantoxin messenger RNA fourfold higher than the GAPDH gene. The immunotoxin function was assessed in A431 cancer cells and EGFR-negative HEK293 cells, and IC50 values were estimated to be 22.4 ± 3 and >620.4 ± 5 nM, respectively. The results indicated that the immunotoxins produced in this study had the potential for use as anticancer drugs.
Collapse
Affiliation(s)
- Nooshin Taghizadegan
- Department of Biochemistry, Shahrood Branch, Islamic Azad University, Shahrood, Iran
| | - Mohsen Firoozrai
- Department of Biochemistry, Shahrood Branch, Islamic Azad University, Shahrood, Iran
| | | | - Hamid Ariannejad
- Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
4
|
Taghizadegan N, Firozrai M, Nassiri M, Ariannejad H. Use of Molecular Dynamic Tools in Engineering of Onconase Enzyme to Increase Cellular Uptake and Evade RI. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-019-09881-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
5
|
Gotte G, Menegazzi M. Biological Activities of Secretory RNases: Focus on Their Oligomerization to Design Antitumor Drugs. Front Immunol 2019; 10:2626. [PMID: 31849926 PMCID: PMC6901985 DOI: 10.3389/fimmu.2019.02626] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/22/2019] [Indexed: 12/11/2022] Open
Abstract
Ribonucleases (RNases) are a large number of enzymes gathered into different bacterial or eukaryotic superfamilies. Bovine pancreatic RNase A, bovine seminal BS-RNase, human pancreatic RNase 1, angiogenin (RNase 5), and amphibian onconase belong to the pancreatic type superfamily, while binase and barnase are in the bacterial RNase N1/T1 family. In physiological conditions, most RNases secreted in the extracellular space counteract the undesired effects of extracellular RNAs and become protective against infections. Instead, if they enter the cell, RNases can digest intracellular RNAs, becoming cytotoxic and having advantageous effects against malignant cells. Their biological activities have been investigated either in vitro, toward a number of different cancer cell lines, or in some cases in vivo to test their potential therapeutic use. However, immunogenicity or other undesired effects have sometimes been associated with their action. Nevertheless, the use of RNases in therapy remains an appealing strategy against some still incurable tumors, such as mesothelioma, melanoma, or pancreatic cancer. The RNase inhibitor (RI) present inside almost all cells is the most efficacious sentry to counteract the ribonucleolytic action against intracellular RNAs because it forms a tight, irreversible and enzymatically inactive complex with many monomeric RNases. Therefore, dimerization or multimerization could represent a useful strategy for RNases to exert a remarkable cytotoxic activity by evading the interaction with RI by steric hindrance. Indeed, the majority of the mentioned RNases can hetero-dimerize with antibody derivatives, or even homo-dimerize or multimerize, spontaneously or artificially. This can occur through weak interactions or upon introducing covalent bonds. Immuno-RNases, in particular, are fusion proteins representing promising drugs by combining high target specificity with easy delivery in tumors. The results concerning the biological features of many RNases reported in the literature are described and discussed in this review. Furthermore, the activities displayed by some RNases forming oligomeric complexes, the mechanisms driving toward these supramolecular structures, and the biological rebounds connected are analyzed. These aspects are offered with the perspective to suggest possible efficacious therapeutic applications for RNases oligomeric derivatives that could contemporarily lack, or strongly reduce, immunogenicity and other undesired side-effects.
Collapse
Affiliation(s)
- Giovanni Gotte
- Biological Chemistry Section, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Marta Menegazzi
- Biological Chemistry Section, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
6
|
Raineri A, Prodomini S, Fasoli S, Gotte G, Menegazzi M. Influence of onconase in the therapeutic potential of PARP inhibitors in A375 malignant melanoma cells. Biochem Pharmacol 2019; 167:173-181. [PMID: 31185226 DOI: 10.1016/j.bcp.2019.06.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 06/06/2019] [Indexed: 12/12/2022]
Abstract
Human malignant melanoma is one of the most aggressive cancers, accompanied with poor prognosis, metastatic evolution and high mortality. Many strategies have been developed using BRAF and MEK inhibitors in spite of the classic therapy with alkylating agents, but failure related to the ability of the tumor to activate alternative proliferation pathways occurred after promising initial successes. Poly(ADP-ribose) polymerase (PARP) enzymes are well known to be crucial for DNA damage response, and PARP inhibition results in the accumulation of DNA strand breaks that induce cell injury. For this reason, PARP-inhibitors (PARPi) have become promising tools to counteract many cancer types. One of the most used by clinicians is olaparib, that, however, showed again cancer resistance in patients. Thus, new generation molecules have been designed mainly to counteract this problem. Among them, we chose to test AZD2461 on the particularly aggressive human melanoma A375 cell line. This drug is a PARPi significantly less prone than olaparib to undergo the P-glycoprotein-mediated efflux mechanism, one of those responsible for resistance, that in turn is the main adversity in melanoma therapy. Then, we analysed AZD2461 also together with the enzyme onconase (ONC) on the same A375 cells, to investigate if the combination of drugs could possibly increase the in vitro antitumor activity. ONC is a small amphibian "pancreatic-type" ribonuclease that is able to exert a remarkable antitumor activity against many cancers, either in vitro or in vivo, principally because it can evade the ubiquitous ribonuclease cytosolic inhibitor thanks to its structural determinants. Hence, ONC became relevant in the use of protein-drug strategies against incurable cancers. The studies performed in this work showed that both drugs definitely affect A375 cells viability by inducing cytostatic and pro-apoptotic effects in a time- and dose-dependent manner, either if administered alone or in combination. Although we registered low synergistic effects with the combination of the two drugs, we found that AZD2461 did not induce resistance in A375 after two months treatment with high concentration of this molecule. Moreover, we underline that A375 cells treated for a prolonged time with AZD2461 were definitely more susceptible than parental A375 cells to the pro-apoptotic action of ONC. Considering also the different inhibitory effects of the two drugs on TNF-α gene expression and NF-κB DNA-binding, the tuning of their combined delivery to the A375 tumor cell line might open a promising scenario for future therapeutic applications devoted to defeat human melanoma.
Collapse
Affiliation(s)
- Alice Raineri
- Department of Neuroscience, Biomedicine, and Movement Sciences, Biological Chemistry Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy
| | - Sara Prodomini
- Department of Neuroscience, Biomedicine, and Movement Sciences, Biological Chemistry Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy
| | - Sabrina Fasoli
- Department of Neuroscience, Biomedicine, and Movement Sciences, Biological Chemistry Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy
| | - Giovanni Gotte
- Department of Neuroscience, Biomedicine, and Movement Sciences, Biological Chemistry Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy
| | - Marta Menegazzi
- Department of Neuroscience, Biomedicine, and Movement Sciences, Biological Chemistry Section, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy.
| |
Collapse
|
7
|
Ojeda PG, Henriques ST, Pan Y, Nicolazzo JA, Craik DJ, Wang CK. Lysine to arginine mutagenesis of chlorotoxin enhances its cellular uptake. Biopolymers 2018; 108. [PMID: 28459137 DOI: 10.1002/bip.23025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/31/2017] [Accepted: 04/10/2017] [Indexed: 12/14/2022]
Abstract
Chlorotoxin (CTX), a disulfide-rich peptide from the scorpion Leiurus quinquestriatus, has several promising biopharmaceutical properties, including preferential affinity for certain cancer cells, high serum stability, and cell penetration. These properties underpin its potential for use as a drug design scaffold, especially for the treatment of cancer; indeed, several analogs of CTX have reached clinical trials. Here, we focus on its ability to internalize into cells-a trait associated with a privileged subclass of peptides called cell-penetrating peptides-and whether it can be improved through conservative substitutions. Mutants of CTX were made using solid-phase peptide synthesis and internalization into human cervical carcinoma (HeLa) cells was monitored by fluorescence and confocal microscopy. CTX_M1 (ie, [K15R/K23R]CTX) and CTX_M2 (ie, [K15R/K23R/Y29W]CTX) mutants showed at least a twofold improvement in uptake compared to CTX. We further showed that these mutants internalize into HeLa cells largely via an energy-dependent mechanism. Importantly, the mutants have high stability, remaining intact in serum for over 24 h; thus, retaining the characteristic stability of their parent peptide. Overall, we have shown that simple conservative substitutions can enhance the cellular uptake of CTX, suggesting that such type of mutations might be useful for improving uptake of other peptide toxins.
Collapse
Affiliation(s)
- Paola G Ojeda
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.,Centro de Bioinformática y Simulación Molecular (CBSM), Universidad de Talca, Talca, Chile
| | - Sónia Troeira Henriques
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Yijun Pan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - David J Craik
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Conan K Wang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
8
|
Oberbauer V, Schaefer MR. tRNA-Derived Small RNAs: Biogenesis, Modification, Function and Potential Impact on Human Disease Development. Genes (Basel) 2018; 9:genes9120607. [PMID: 30563140 PMCID: PMC6315542 DOI: 10.3390/genes9120607] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 12/11/2022] Open
Abstract
Transfer RNAs (tRNAs) are abundant small non-coding RNAs that are crucially important for decoding genetic information. Besides fulfilling canonical roles as adaptor molecules during protein synthesis, tRNAs are also the source of a heterogeneous class of small RNAs, tRNA-derived small RNAs (tsRNAs). Occurrence and the relatively high abundance of tsRNAs has been noted in many high-throughput sequencing data sets, leading to largely correlative assumptions about their potential as biologically active entities. tRNAs are also the most modified RNAs in any cell type. Mutations in tRNA biogenesis factors including tRNA modification enzymes correlate with a variety of human disease syndromes. However, whether it is the lack of tRNAs or the activity of functionally relevant tsRNAs that are causative for human disease development remains to be elucidated. Here, we review the current knowledge in regard to tsRNAs biogenesis, including the impact of RNA modifications on tRNA stability and discuss the existing experimental evidence in support for the seemingly large functional spectrum being proposed for tsRNAs. We also argue that improved methodology allowing exact quantification and specific manipulation of tsRNAs will be necessary before developing these small RNAs into diagnostic biomarkers and when aiming to harness them for therapeutic purposes.
Collapse
Affiliation(s)
- Vera Oberbauer
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University Vienna, Schwarzspanierstrasse 17, A-1090 Vienna, Austria.
| | - Matthias R Schaefer
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University Vienna, Schwarzspanierstrasse 17, A-1090 Vienna, Austria.
| |
Collapse
|
9
|
Qi J, Ye X, Li L, Bai H, Xu C. Improving the specific antitumor efficacy of ONC by fusion with N-terminal domain of transferrin. Biosci Biotechnol Biochem 2018; 82:1153-1158. [PMID: 29629632 DOI: 10.1080/09168451.2018.1456318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Onconase (ONC) as a novel anti-tumor drug has a significant killing effect on a variety of tumor cells. Drug delivery system mediated by transferrin (TF) and TF receptor (TfR), which can significantly increase the amount of drug uptake in the tumor cells, enhance the initiative target efficiency of drugs and reduce its toxic side effects. It has been widely used in drug delivery and clinical trials. In this study, the rONC-TFn was expressed in Escherichia coli by linking ONC with the N-terminal domain of TF (TFn). ELISA and competitive binding analysis demonstrated that rONC-TFn can bind to TfR. The rONC-TFn protein showed much higher cytotoxicity to the cultured HepG2 and Hela cells than rONC. These results suggested that the N-terminal domain protein of TF promoted the tumor targeting of ONC, and thus the rONC-TFn fusion protein may be further developed as a potential targeted anti-tumor drug.
Collapse
Affiliation(s)
- Jianying Qi
- a College of Life Science , Henan Normal University , Xinxiang , China.,b Key Laboratory for Cell Differentiation Regulation , Henan Normal University , Xinxiang , China
| | - Xianlong Ye
- a College of Life Science , Henan Normal University , Xinxiang , China
| | - Lingling Li
- a College of Life Science , Henan Normal University , Xinxiang , China.,b Key Laboratory for Cell Differentiation Regulation , Henan Normal University , Xinxiang , China
| | - Haijing Bai
- a College of Life Science , Henan Normal University , Xinxiang , China.,b Key Laboratory for Cell Differentiation Regulation , Henan Normal University , Xinxiang , China
| | - Cunshuan Xu
- a College of Life Science , Henan Normal University , Xinxiang , China.,b Key Laboratory for Cell Differentiation Regulation , Henan Normal University , Xinxiang , China
| |
Collapse
|
10
|
Zhao Y, Zhang YH, Zhuge Z, Tang YH, Tao JW, Chen Y. Synthesis of a Poly-l-Lysine/Black Phosphorus Hybrid for Biosensors. Anal Chem 2018; 90:3149-3155. [DOI: 10.1021/acs.analchem.7b04395] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Yun Zhao
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Ye-Hua Zhang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Zhen Zhuge
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Yi-Hong Tang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Jian-Wei Tao
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Yong Chen
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| |
Collapse
|
11
|
Chapman AM, McNaughton BR. Scratching the Surface: Resurfacing Proteins to Endow New Properties and Function. Cell Chem Biol 2017; 23:543-553. [PMID: 27203375 DOI: 10.1016/j.chembiol.2016.04.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 04/21/2016] [Accepted: 04/27/2016] [Indexed: 12/22/2022]
Abstract
Protein engineering is an emerging discipline that dovetails modern molecular biology techniques with high-throughput screening, laboratory evolution technologies, and computational approaches to modify sequence, structure, and, in some cases, function and properties of proteins. The ultimate goal is to develop new proteins with improved or designer functions for use in biotechnology, medicine, and basic research. One way to engineer proteins is to change their solvent-exposed regions through focused or random "protein resurfacing." In this review we explain what protein resurfacing is, and discuss recent examples of how this strategy is used to generate proteins with altered or broadened recognition profiles, improved stability, solubility, and expression, cell-penetrating ability, and reduced immunogenicity. Additionally we comment on how these properties can be further improved using chemical resurfacing approaches. Protein resurfacing will likely play an increasingly important role as more biologics enter clinical use, and we present some arguments to support this view.
Collapse
Affiliation(s)
- Alex M Chapman
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | - Brian R McNaughton
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA; Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
12
|
Sarapas JM, Backlund CM, deRonde BM, Minter LM, Tew GN. ROMP- and RAFT-Based Guanidinium-Containing Polymers as Scaffolds for Protein Mimic Synthesis. Chemistry 2017; 23:6858-6863. [PMID: 28370636 PMCID: PMC5551038 DOI: 10.1002/chem.201700423] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Indexed: 01/21/2023]
Abstract
Cell-penetrating peptides are an important class of molecules with promising applications in bioactive cargo delivery. A diverse series of guanidinium-containing polymeric cell-penetrating peptide mimics (CPPMs) with varying backbone chemistries was synthesized and assessed for delivery of both GFP and fluorescently tagged siRNA. Specifically, we examined CPPMs based on norbornene, methacrylate, and styrene backbones to determine how backbone structure impacted internalization of these two cargoes. Either charge content or degree of polymerization was held constant at 20, with diguanidinium norbornene molecules being polymerized to both 10 and 20 repeat units. Generally, homopolymer CPPMs delivered low amounts of siRNA into Jurkat T cells, with no apparent backbone dependence; however, by adding a short hydrophobic methyl methacrylate block to the guanidinium-rich methacrylate polymer, siRNA delivery to nearly the entire cell population was achieved. Protein internalization yielded similar results for most of the CPPMs, though the block polymer was unable to deliver proteins. In contrast, the styrene-based CPPM yielded the highest internalization for GFP (≈40 % of cells affected), showing that indeed backbone chemistry impacts protein delivery, specifically through the incorporation of an aromatic group. These results demonstrate that an understanding of how polymer structure affects cargo-dependent internalization is critical to designing new, more effective CPPMs.
Collapse
Affiliation(s)
- Joel M Sarapas
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Coralie M Backlund
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Brittany M deRonde
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Lisa M Minter
- Department of Molecular and Cellular Biology, Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Department of Veterinary and Animal Sciences, Department of Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Gregory N Tew
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Department of Molecular and Cellular Biology, Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Department of Veterinary and Animal Sciences, Department of Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| |
Collapse
|
13
|
Dissanayake S, Denny WA, Gamage S, Sarojini V. Recent developments in anticancer drug delivery using cell penetrating and tumor targeting peptides. J Control Release 2017; 250:62-76. [DOI: 10.1016/j.jconrel.2017.02.006] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 02/02/2017] [Accepted: 02/02/2017] [Indexed: 12/13/2022]
|
14
|
Crawford DW, Blakeley BD, Chen PH, Sherpa C, Le Grice SF, Laird-Offringa IA, McNaughton BR. An Evolved RNA Recognition Motif That Suppresses HIV-1 Tat/TAR-Dependent Transcription. ACS Chem Biol 2016; 11:2206-15. [PMID: 27253715 DOI: 10.1021/acschembio.6b00145] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Potent and selective recognition and modulation of disease-relevant RNAs remain a daunting challenge. We previously examined the utility of the U1A N-terminal RNA recognition motif as a scaffold for tailoring new RNA hairpin recognition and showed that as few as one or two mutations can result in moderate affinity (low μM dissociation constant) for the human immunodeficiency virus (HIV) trans-activation response element (TAR) RNA, an RNA hairpin controlling transcription of the human immunodeficiency virus (HIV) genome. Here, we use yeast display and saturation mutagenesis of established RNA-binding regions in U1A to identify new synthetic proteins that potently and selectively bind TAR RNA. Our best candidate has truly altered, not simply broadened, RNA-binding selectivity; it binds TAR with subnanomolar affinity (apparent dissociation constant of ∼0.5 nM) but does not appreciably bind the original U1A RNA target (U1hpII). It specifically recognizes the TAR RNA hairpin in the context of the HIV-1 5'-untranslated region, inhibits the interaction between TAR RNA and an HIV trans-activator of transcription (Tat)-derived peptide, and suppresses Tat/TAR-dependent transcription. Proteins described in this work are among the tightest TAR RNA-binding reagents-small molecule, nucleic acid, or protein-reported to date and thus have potential utility as therapeutics and basic research tools. Moreover, our findings demonstrate how a naturally occurring RNA recognition motif can be dramatically resurfaced through mutation, leading to potent and selective recognition-and modulation-of disease-relevant RNA.
Collapse
Affiliation(s)
| | | | - Po-Han Chen
- Department of Surgery and Department of Biochemistry & Molecular Biology, USC/Norris Comprehensive Cancer Center, Keck School of Medicine, Los Angeles, California 90033, United States
| | - Chringma Sherpa
- Basic
Research Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Stuart F.J. Le Grice
- Basic
Research Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Ite A. Laird-Offringa
- Department of Surgery and Department of Biochemistry & Molecular Biology, USC/Norris Comprehensive Cancer Center, Keck School of Medicine, Los Angeles, California 90033, United States
| | | |
Collapse
|
15
|
LaRochelle JR, Cobb GB, Steinauer A, Rhoades E, Schepartz A. Fluorescence correlation spectroscopy reveals highly efficient cytosolic delivery of certain penta-arg proteins and stapled peptides. J Am Chem Soc 2015; 137:2536-2541. [PMID: 25679876 PMCID: PMC4700819 DOI: 10.1021/ja510391n] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
We used fluorescence correlation spectroscopy (FCS) to accurately and precisely determine the relative efficiencies with which three families of "cell-penetrating peptides" traffic to the cytosol of mammalian cells. We find that certain molecules containing a "penta-arg" motif reach the cytosol, intact, with efficiencies greater than 50%. This value is at least 10-fold higher than that observed for the widely studied cationic sequence derived from HIV Tat or polyarginine Arg8, and equals that of hydrocarbon-stapled peptides that are active in cells and animals. Moreover, we show that the efficiency with which stapled peptides reach the cytosol, as determined by FCS, correlates directly with their efficacy in cell-based assays. We expect that these findings and the associated technology will aid the design of peptides, proteins, and peptide mimetics that predictably and efficiently reach the interior of mammalian cells.
Collapse
Affiliation(s)
- Jonathan R. LaRochelle
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Garrett B. Cobb
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Angela Steinauer
- Department of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Elizabeth Rhoades
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Alanna Schepartz
- Department of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520-8107, United States
| |
Collapse
|
16
|
Huang YH, Chaousis S, Cheneval O, Craik DJ, Henriques ST. Optimization of the cyclotide framework to improve cell penetration properties. Front Pharmacol 2015; 6:17. [PMID: 25709580 PMCID: PMC4321561 DOI: 10.3389/fphar.2015.00017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 01/20/2015] [Indexed: 12/11/2022] Open
Abstract
Cell penetrating peptides have been regarded as promising vectors to deliver hydrophilic molecules inside cells. Although they are great tools for research and have high potential as drug delivery systems, their application as drugs is impaired by their low stability in serum. Cyclotides, cyclic disulfide-rich peptides from plants, are ultra-stable molecules that have inspired applications in drug design as they can be used as scaffolds to stabilize linear bioactive sequences. Recently, they have also been shown to possess cell-penetrating properties. The combination of their remarkable stability and cell-penetrating properties opens new avenues for the application of peptides to bind to and inhibit intracellular proteins. Nevertheless, for a broader application of these molecules as vectors is of utmost importance to improve their cellular internalization efficiency. In this study we successfully modified MCoTI-II, one of the most widely studied cyclotide scaffolds in drug design, and improved its internalization properties. The internalization of the newly designed MCoTI-II is as efficient as the gold standard cell-penetrating peptide (CPP) TAT and maintains all the required features as a template to graft desired bioactivities.
Collapse
Affiliation(s)
- Yen-Hua Huang
- Institute for Molecular Bioscience, The University of Queensland Brisbane, QLD, Australia
| | - Stephanie Chaousis
- Institute for Molecular Bioscience, The University of Queensland Brisbane, QLD, Australia
| | - Olivier Cheneval
- Institute for Molecular Bioscience, The University of Queensland Brisbane, QLD, Australia
| | - David J Craik
- Institute for Molecular Bioscience, The University of Queensland Brisbane, QLD, Australia
| | - Sónia T Henriques
- Institute for Molecular Bioscience, The University of Queensland Brisbane, QLD, Australia
| |
Collapse
|
17
|
Vilian ATE, Mani V, Chen SM, Dinesh B, Huang ST. The Immobilization of Glucose Oxidase at Manganese Dioxide Particles-Decorated Reduced Graphene Oxide Sheets for the Fabrication of a Glucose Biosensor. Ind Eng Chem Res 2014. [DOI: 10.1021/ie502430d] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- A. T. Ezhil Vilian
- Department
of Chemical Engineering and Biotechnology, National Taipei University of Technology, No.1, Section 3, Chung-Hsiao East Road, Taipei 106, Taiwan, R.O.C
| | - Veerappan Mani
- Department
of Chemical Engineering and Biotechnology, National Taipei University of Technology, No.1, Section 3, Chung-Hsiao East Road, Taipei 106, Taiwan, R.O.C
| | - Shen-Ming Chen
- Department
of Chemical Engineering and Biotechnology, National Taipei University of Technology, No.1, Section 3, Chung-Hsiao East Road, Taipei 106, Taiwan, R.O.C
| | - Bose Dinesh
- Department
of Materials Science, School of Chemistry, Madurai Kamaraj University, Madurai, Tamilnadu 625 021, India
| | - Sheng-Tung Huang
- Department
of Chemical Engineering and Biotechnology, National Taipei University of Technology, No.1, Section 3, Chung-Hsiao East Road, Taipei 106, Taiwan, R.O.C
| |
Collapse
|
18
|
Arnold U. Stability and folding of amphibian ribonuclease A superfamily members in comparison with mammalian homologues. FEBS J 2014; 281:3559-75. [PMID: 24966023 DOI: 10.1111/febs.12891] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 06/18/2014] [Indexed: 01/05/2023]
Abstract
Comparative studies on homologous proteins can provide knowledge on how limited changes in the primary structure find their expression in large effects on catalytic activity, stability or the folding behavior. For more than half a century, members of the ribonuclease A superfamily have been the subject of a myriad of studies on protein folding and stability. Both the unfolding and refolding kinetics as well as the structure of several folding intermediates of ribonuclease A have been characterized in detail. Moreover, the RNA-degrading activity of these enzymes provides a basis for their cytotoxicity, which renders them potential tumor therapeutics. Because amphibian ribonuclease A homologues evade the human ribonuclease inhibitor, they emerged as particularly promising candidates. Interestingly, the amphibian ribonuclease A homologues investigated to date are more stable than the mammalian homologues. Nevertheless, despite the generation of numerous genetically engineered variants, knowledge of the folding of amphibian ribonuclease A homologues remains rather limited. An exception is onconase, a ribonuclease A homologue from Rana pipiens, which has been characterized in detail. This review summarizes the data on the unfolding and refolding kinetics and pathways, as well on the stability of amphibian ribonuclease A homologues compared with those of ribonuclease A, the best known member of this superfamily.
Collapse
Affiliation(s)
- Ulrich Arnold
- Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Germany
| |
Collapse
|
19
|
Sialyl-glycoconjugates in cholesterol-rich microdomains of P388 cells are the triggers for apoptosis induced by Rana catesbeiana oocyte ribonuclease. Glycoconj J 2013; 31:171-84. [PMID: 24271942 DOI: 10.1007/s10719-013-9513-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Revised: 10/21/2013] [Accepted: 11/07/2013] [Indexed: 10/26/2022]
Abstract
SBL/RC-RNase was originally isolated from frog (Rana catesbeiana) oocytes and purified as a novel sialic acid-binding lectin (SBL) that displayed strong anti-cancer activity. SBL was later shown to be identical to a ribonuclease (RC-RNase) from oocytes of the same species. The administration of SBL/RC-RNase induced apoptosis (with nuclear condensation and DNA fragmentation) in mouse leukemia P388 cells but did not kill umbilical vein endothelial or fibroblast cells derived from normal tissues. The cytotoxic activity of SBL/RC-RNase was inhibited by desialylation of P388 cells and/or the co-presence of free bovine submaxillary mucin. FACS analysis showed that SBL/RC-RNase was incorporated into cells after attachment to cholesterol-rich microdomains. Addition of the cholesterol remover methyl-β-cyclodextrin reduced SBL/RC-RNase-induced apoptosis. Apoptosis occurred through the caspase-3 pathway following activation of caspase-8 by SBL/RC-RNase. A heat shock cognate protein (Hsc70) and a heat shock protein (Hsp70) (each 70 kDa) on the cell membrane were shown to bind to SBL/RC-RNase by mass spectrometric and flow cytometric analyses. Quercetin, an inhibitor of Hsc70 and Hsp70, significantly reduced SBL/RC-RNase-induced apoptosis. Taken together, our findings suggest that sialyl-glycoconjugates present in cholesterol-rich microdomains form complexes with Hsc70 or Hsp70 that act as triggers for SBL/RC-RNase to induce apoptosis through a pathway involving the activation of caspase-3 and caspase-8.
Collapse
|
20
|
Su Y, Li S, Hong M. Cationic membrane peptides: atomic-level insight of structure-activity relationships from solid-state NMR. Amino Acids 2013; 44:821-33. [PMID: 23108593 PMCID: PMC3570695 DOI: 10.1007/s00726-012-1421-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 10/11/2012] [Indexed: 12/24/2022]
Abstract
Many membrane-active peptides, such as cationic cell-penetrating peptides (CPPs) and antimicrobial peptides (AMPs), conduct their biological functions by interacting with the cell membrane. The interactions of charged residues with lipids and water facilitate membrane insertion, translocation or disruption of these highly hydrophobic species. In this review, we will summarize high-resolution structural and dynamic findings towards the understanding of the structure-activity relationship of lipid membrane-bound CPPs and AMPs, as examples of the current development of solid-state NMR (SSNMR) techniques for studying membrane peptides. We will present the most recent atomic-resolution structure of the guanidinium-phosphate complex, as constrained from experimentally measured site-specific distances. These SSNMR results will be valuable specifically for understanding the intracellular translocation pathway of CPPs and antimicrobial mechanism of AMPs, and more generally broaden our insight into how cationic macromolecules interact with and cross the lipid membrane.
Collapse
Affiliation(s)
- Yongchao Su
- Department of Chemistry, Iowa State University, Ames, IA 50011, USA.
| | | | | |
Collapse
|
21
|
Wang J, Zhao Y, Ma FX, Wang K, Wang FB, Xia XH. Synthesis of a hydrophilic poly-l-lysine/graphene hybrid through multiple non-covalent interactions for biosensors. J Mater Chem B 2013; 1:1406-1413. [PMID: 32260780 DOI: 10.1039/c2tb00454b] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Surface modification has been proved to be one of the effective strategies for enhancing the properties of graphene sheets. When a non-covalent modification method is appropriately designed, novel opportunities for better performance of graphene nanosheets can be expected since this strategy can tailor the properties of graphene while its natural structure is retained. This paper introduces a simple route to prepare a highly biocompatible, stable and conductive graphene hybrid modified by poly-l-lysine (PLL) for biosensors using the non-covalent strategy. Results show that PLL adopts a random conformation with the nonpolar parts exposed to outside since its side chains are positively charged under neutral conditions. This conformation allows the strong adhesion of PLL to graphene surface via the hydrophobic interaction between butyl chains of PLL and graphene surface, cation-π interaction of protonated amine groups on PLL with the π electrons in graphene, and electrostatic interaction between the protonated amine groups on PLL and the negatively charged carboxyl groups remaining on graphene. All these interactions make the resultant PLL-G hybrid stable and dispersible in aqueous solutions. The resultant hybrid is then used to construct high performance biosensors. As demonstration, hemoglobin (Hb) carrying negative charges can be easily immobilized on the hybrid via electrostatic interactions with the positively charged lysine side chains of PLL modified on graphene surface, forming the Hb@PLL-G bionanocomposite. The immobilized protein retains its native structure and exhibits reversible direct electrochemistry. The Hb@PLL-G based enzymatic electrochemical biosensor shows excellent catalytic activity toward its substrate hydrogen peroxide. Its electrochemical response shows the linear dependence of hydrogen peroxide concentration in a range between 10 μM and 80 μM with a detection limit of 0.1 μM. The apparent Michaelis-Menten constant is calculated as 0.0753 mM, demonstrating the significant catalytic ability of the protein. The present strategy can be extended to modify other carbon materials and the resultant nanocomposites are promising for construction of biosensors, bioelectronics and biofuel cells.
Collapse
Affiliation(s)
- Jiong Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, China.
| | | | | | | | | | | |
Collapse
|
22
|
Diez-García F, Chakrabartty A, González C, Laurents DV. An Arg-rich putative prebiotic protein is as stable as its Lys-rich variant. Arch Biochem Biophys 2012; 528:118-26. [DOI: 10.1016/j.abb.2012.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 09/14/2012] [Accepted: 09/15/2012] [Indexed: 10/27/2022]
|
23
|
Zhao HL, Xue C, Du JL, Ren M, Xia S, Cheng YG, Liu ZM. Sustained and cancer cell targeted cytosolic delivery of Onconase results in potent antitumor effects. J Control Release 2012; 159:346-52. [DOI: 10.1016/j.jconrel.2012.02.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|