Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Genes (Basel) ; 10(12)2019 12 16.
Article in English | MEDLINE | ID: mdl-31888196

ABSTRACT

This study was designed to establish a real-time quantitative polymerase chain reaction (qPCR) method to rapidly and reliably analyze the hypoglycemic polypeptide-P gene expression pattern in Momordica charantia (MC) and to examine its expression changes in different MC accessions, harvesting seasons, and tissue types. The qPCR results were further verified by using Western blotting (WB). A total of 10 MCs with different accessions were collected and tested in this study. Among the tested accessions, RU5H showed the highest expression level of the polypeptide-P gene. The expression level of the polypeptide-P gene was not only season-related (with the highest in early July) but also tissue-related (with the highest in the seed tissue). In addition, the expression characteristic of the polypeptide-P gene was maturity-related, with the highest expression level in the tender MC. The WB results show that the transcription level of this gene shows an almost similar trend to the corresponding protein expression level. In conclusion, the established qPCR method can rapidly and effectively detect the expression levels of the polypeptide-P gene in MCs with different accessions; furthermore, various factors, including the accessions, harvesting seasons, and tissue types can affect the expression level.


Subject(s)
Hypoglycemic Agents/metabolism , Momordica charantia/genetics , Peptides/genetics , Plant Proteins/genetics , Fruit/genetics , Fruit/metabolism , Gene Expression Regulation, Plant , Momordica charantia/metabolism , Peptides/metabolism , Plant Proteins/metabolism , RNA, Plant/metabolism , Real-Time Polymerase Chain Reaction/methods , Seasons , Seeds/genetics , Seeds/metabolism
2.
J Drug Target ; 26(9): 777-792, 2018 11.
Article in English | MEDLINE | ID: mdl-29303375

ABSTRACT

Cell-penetrating peptide (CPP) is used for the delivery of biomacromolecules across the cell membrane and is limited in cancer therapy due to the lack of cell selectivity. Epidermal growth factor receptor (EGFR) has been widely used in clinical targeted therapy for tumours. Here, we reported a novel tumour targeting cell-penetrating peptide (TCPP), EHB (ELBD-C6H) with 20-fold and 3000-fold greater transmembrane ability and tumour cell selectivity than our previously reported S3-HBD and classic CPP TAT, respectively. In this new TCPP, a specific alpha helix structure was inserted into a repeated amino acid (AA) sequence formed by tandem multiple selected key AA residues of vaccinia growth factor (VGF), and this sequence was then fused to a tailored heparin binding domain sequence (C6H) derived from heparin-binding epidermal growth factor-like growth factor to intensify its targeting delivery ability. EHB could carry anticancer proteins such as MAP30 (Momordica Antiviral Protein 30 kDa) into EGFR-overexpressing cancer cell and inhibit cell growth, but it had a greatly reduced interaction with normal cells. These results indicated that EHB, as a novel efficient TCPP for the selective delivery of drug molecules into cancer cells, would help to improve the efficacy and safety of anti-tumour drugs.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Drug Delivery Systems/methods , Cell Line, Tumor , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/metabolism , Humans , Protein Conformation
3.
Biotechnol Lett ; 39(1): 71-78, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27714558

ABSTRACT

OBJECTIVE: To evaluate the anti-tumor effects of trichosanthin after fusion with a cell penetrating peptide, heparin-binding peptide (HBP), derived from human heparin-binding EGF-like growth factor (HB-EGF). RESULTS: The fusion protein of trichosanthin-HBP was expressed in Escherichia coli BL21 and purified by Ni-NTA affinity chromatography. The HBP domain had no influence on the topological inactivation activity and N-glycosidase activity of trichosanthin. Trichosanthin-HBP significantly inhibited the growth of tested cancer cells which are impervious to trichosanthin. Tumor cell apoptosis and both the mitochondrial- and death receptor-mediated apoptotic signaling pathways induced by trichosanthin-HBP were more significant than those induced by trichosanthin in HeLa cells. CONCLUSION: HBP is an efficient intracellular delivery vehicle for trichosanthin and makes trichosanthin-HBP become a promising agent for cancer therapy.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Heparin-binding EGF-like Growth Factor/chemistry , Peptides/metabolism , Peptides/pharmacology , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Trichosanthin/metabolism , Trichosanthin/pharmacology , Apoptosis/drug effects , HeLa Cells , Humans , Peptides/genetics , Recombinant Fusion Proteins/genetics , Trichosanthin/genetics
4.
J Pept Sci ; 22(11-12): 689-699, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27739168

ABSTRACT

Cell-penetrating peptides (CPPs) have been shown to be potential drug carriers for cancer therapy. The inherently low immunogenicity and cytotoxicity of human-derived CPPs make them more suitable for intracellular drug delivery compared to other delivery vehicles. In this work, the protein transduction ability of a novel CPP (termed HBP) derived from the heparin-binding domain of HB-EGF was evaluated. Our data shows, for the first time, that HBP possesses similar properties to typical CPPs and is a potent drug delivery vector for improving the antitumor activity of impermeable MAP30. The intrinsic bioactivities of recombinant MAP30-HBP were well preserved compared to those of free MAP30. Furthermore, HBP conjugated to the C-terminus of MAP30 promoted the cellular uptake of recombinant MAP30-HBP. Moreover, the fusion of HBP to MAP30 gave rise to significantly enhanced cytotoxic effects in all of the tumor cell lines tested. In HeLa cells, this cytotoxicity was mainly caused by the induction of cell apoptosis. Further investigation revealed that HBP enhanced MAP30-induced apoptosis through the activation of the mitochondrial- and death receptor-mediated signaling pathways. In addition, the MAP30-HBP fusion protein caused more HeLa cells to become arrested in S phase compared to MAP30 alone. These results highlight the MAP30-HBP fusion protein as a promising drug candidate for cancer therapy and demonstrate HBP, a novel CPP derived from human HB-EGF, as a new potential vector for antitumor drug delivery. Copyright © 2016 European Peptide Society and John Wiley & Sons, Ltd.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Drug Carriers/pharmacology , Heparin-binding EGF-like Growth Factor/pharmacology , Recombinant Fusion Proteins/pharmacology , Ribosome Inactivating Proteins, Type 2/pharmacology , Amino Acid Sequence , Apoptosis/drug effects , Cell Line, Tumor , Cell-Penetrating Peptides/biosynthesis , Cell-Penetrating Peptides/genetics , Cloning, Molecular , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Drug Carriers/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HeLa Cells , Heparin/chemistry , Heparin/metabolism , Heparin-binding EGF-like Growth Factor/biosynthesis , Heparin-binding EGF-like Growth Factor/genetics , Humans , Momordica charantia/chemistry , Protein Binding , Protein Domains , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Ribosome Inactivating Proteins, Type 2/biosynthesis , Ribosome Inactivating Proteins, Type 2/genetics , S Phase/drug effects , Signal Transduction
5.
Protein Expr Purif ; 111: 9-17, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25797209

ABSTRACT

MAP30 (Momordica Antiviral Protein 30 Kd), a single-stranded type-I ribosome inactivating protein, possesses versatile biological activities including anti-tumor abilities. However, the low efficiency penetrating into tumor cells hampers the tumoricidal effect of MAP30. This paper describes MAP30 fused with a human-derived cell penetrating peptide HBD which overcome the low uptake efficiency by tumor cells and exhibits higher anti-tumor bioactivity. MAP30 gene was cloned from the genomic DNA of Momordica charantia and the recombinant plasmid pET28b-MAP30-HBD was established and transferred into Escherichia coli BL21 (DE3). The recombinant MAP30-HBD protein (rMAP30-HBD) was expressed in a soluble form after being induced by 0.5mM IPTG for 14h at 15°C. The recombinant protein was purified to greater than 95% purity with Ni-NTA affinity chromatography. The rMAP30-HBD protein not only has topological inactivation and protein translation inhibition activity but also showed significant improvements in cytotoxic activity compared to that of the rMAP30 protein without HBD in the tested tumor cell lines, and induced higher apoptosis rates in HeLa cells analyzed by Annexin V-FITC with FACS. This paper demonstrated a new method for improving MAP30 protein anti-tumor activity and might have potential applications in cancer therapy area.


Subject(s)
Antineoplastic Agents , Cell-Penetrating Peptides , Neoplasms/drug therapy , Ribosome Inactivating Proteins, Type 2 , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Cell-Penetrating Peptides/biosynthesis , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/isolation & purification , Cell-Penetrating Peptides/pharmacology , HeLa Cells , Humans , Neoplasms/metabolism , Neoplasms/pathology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacology , Ribosome Inactivating Proteins, Type 2/biosynthesis , Ribosome Inactivating Proteins, Type 2/chemistry , Ribosome Inactivating Proteins, Type 2/genetics , Ribosome Inactivating Proteins, Type 2/isolation & purification , Ribosome Inactivating Proteins, Type 2/pharmacology
6.
Amino Acids ; 47(5): 997-1006, 2015 May.
Article in English | MEDLINE | ID: mdl-25655386

ABSTRACT

Cell-penetrating peptides (CPPs) are well known as intracellular delivery vectors. However, unsatisfactory delivery efficiency and poor specificity are challenging barriers to CPP applications at the clinical trial stage. Here, we showed that S3, an EGFR-binding domain derived from vaccinia virus growth factor, when fused to a CPP such as HBD or TAT can substantially enhance its internalization efficiency and tumor selectivity. The uptake of S3-HBD (S3H) recombinant molecule by tumor cells was nearly 80 folds increased compared to HBD alone. By contrast, the uptake of S3H by non-neoplastic cells still remained at a low level. The specific recognition between S3 and its receptor, EGFR, as well as between HBD and heparan sulfate proteoglycans on the cell surface was essential for these improvements, suggesting a syngeneic effect between the two functional domains in conjugation. This syngeneic effect is likely similar to that of the heparin-binding epidermal growth factor, which is highly abundant particularly in metastatic tumors. The process that S3H entered cells was dependent on time, dosage, and energy, via macropinocytosis pathway. With excellent cell-penetrating efficacy and a novel tumor-targeting ability, S3H appears as a promising candidate vector for targeted anti-cancer drug delivery.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Blood Proteins/metabolism , Carrier Proteins/metabolism , Cell-Penetrating Peptides/metabolism , Drug Delivery Systems/methods , ErbB Receptors/metabolism , Peptides/metabolism , Recombinant Fusion Proteins/metabolism , Amino Acid Sequence , Antimicrobial Cationic Peptides/genetics , Blood Proteins/genetics , Carrier Proteins/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/pharmacology , Cloning, Molecular , Dose-Response Relationship, Drug , ErbB Receptors/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HEK293 Cells , Heparan Sulfate Proteoglycans/chemistry , Heparan Sulfate Proteoglycans/metabolism , Humans , Intercellular Signaling Peptides and Proteins , Molecular Sequence Data , Molecular Targeted Therapy , Organ Specificity , Peptides/genetics , Pinocytosis , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Time Factors , Vaccinia virus/chemistry
7.
J Drug Target ; 23(5): 444-52, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25609361

ABSTRACT

Human-derived cell penetrating peptides (CPPs) have attracted much more attentions than other CPPs which are limited by their potential toxicity and immunogenicity. Previously, we identified a novel human-originated CPP (named heparin-binding domain (HBD) in this article), which derived from the C-terminus of human extracellular superoxide dismutase, and demonstrated HBD is an efficient vector for delivering exogenous drug molecules such as apoptin into HeLa cells. In this study, we found this novel CPP showed differentiated efficiency in several tested cell lines. Heparin competitive inhibition experiment and heparanase pre-incubation experiment showed cell surface polysaccharides play an important role for the transmembrane transport. The results of endocytosis inhibitors suggested that HBD penetrates the cell membrane via a direct translocation, which is different from that of TAT, a classical clathrin-mediated endocytosis. HBD could deliver up to 90 kD protein cargoes into cells. Different conjugated modes with cargo molecules greatly affect their translocation efficiency. HBD also showed significant nuclear transport capacity when it was incubated with HeLa cells. Furthermore, the core region for HBD possessing membrane-penetrating ability was identified by deletion analyses. These results would be helpful for developing HBD as a new nuclear delivery tool for therapeutic biomolecules.


Subject(s)
Cell-Penetrating Peptides/metabolism , Drug Carriers/metabolism , Drug Delivery Systems , Animals , Biological Transport/physiology , Cell Line , Cell Membrane/metabolism , Fluorescent Dyes/metabolism , Glucuronidase/metabolism , HeLa Cells , Heparin/metabolism , Humans , Polysaccharides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...