Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Curr Protein Pept Sci ; 22(1): 74-88, 2021.
Article En | MEDLINE | ID: mdl-33208071

Breast cancer is a heterogeneous malignancy and is the second leading cause of mortality among women around the world. Increasing the resistance to anti-cancer drugs in breast cancer cells persuades researchers to search the novel therapeutic approaches for the treatment of this malignancy. Among the novel methods, therapeutic peptides that target and disrupt tumor cells have been of great interest. Therapeutic peptides are short amino acid monomer chains with high specificity to bind and modulate a protein interaction of interest. Several advantages of peptides, such as specific binding on tumor cells surface, low molecular weight, and low toxicity on normal cells, make the peptides appealing therapeutic agents against solid tumors, particularly breast cancer. Also, the National Institutes of Health (NIH) describes therapeutic peptides as a suitable candidate for the treatment of drug-resistant breast cancer. In this review, we attempt to review the different therapeutic peptides against breast cancer cells that can be used in the treatment and diagnosis of the malignancy. Meanwhile, we presented an overview of peptide vaccines that have been developed for the treatment of breast cancer.


Antineoplastic Agents/therapeutic use , BRCA1 Protein/genetics , Breast Neoplasms/drug therapy , Cancer Vaccines/therapeutic use , Cell-Penetrating Peptides/therapeutic use , Neoplasm Proteins/genetics , Antineoplastic Agents/chemical synthesis , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cancer Vaccines/biosynthesis , Cancer Vaccines/immunology , Cell Death/drug effects , Cell Death/genetics , Cell-Penetrating Peptides/biosynthesis , Cell-Penetrating Peptides/chemical synthesis , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Tumor Cells, Cultured , Vaccines, Subunit
2.
Biomolecules ; 10(11)2020 11 17.
Article En | MEDLINE | ID: mdl-33213097

Cell-penetrating peptides (CPPs) are short peptides that are able to efficiently penetrate cellular lipid bilayers. Although CPPs have been used as carriers in conjugation with certain cargos to target specific genes and pathways, how rationally designed CPPs per se affect global gene expression has not been investigated. Therefore, following time course treatments with 4 CPPs-penetratin, PepFect14, mtCPP1 and TP10, HeLa cells were transcriptionally profiled by RNA sequencing. Results from these analyses showed a time-dependent response to different CPPs, with specific sets of genes related to ribosome biogenesis, microtubule dynamics and long-noncoding RNAs being differentially expressed compared to untreated controls. By using an image-based high content phenotypic profiling platform we confirmed that differential gene expression in CPP-treated HeLa cells strongly correlates with changes in cellular phenotypes such as increased nucleolar size and dispersed microtubules, compatible with altered ribosome biogenesis and cell growth. Altogether these results suggest that cells respond to different cell penetrating peptides by alteration of specific sets of genes, which are possibly part of the common response to such stimulus.


Cell-Penetrating Peptides/biosynthesis , Microtubules/metabolism , RNA, Long Noncoding/biosynthesis , Ribosomes/metabolism , Transcription, Genetic/physiology , Cell Membrane/genetics , Cell Membrane/metabolism , Cell-Penetrating Peptides/genetics , Gene Expression , Gene Regulatory Networks/physiology , HeLa Cells , Humans , Microtubules/genetics , RNA, Long Noncoding/genetics , Ribosomes/genetics
3.
Molecules ; 24(16)2019 Aug 12.
Article En | MEDLINE | ID: mdl-31408975

Intrinsically disordered proteins play a central role in dynamic regulatory and assembly processes in the cell. Recently, a human κ-casein proteolytic fragment called lactaptin (8.6 kDa) was found to induce apoptosis of human breast adenocarcinoma MCF-7 and MDA-MB-231 cells with no cytotoxic activity toward normal cells. Earlier, we had designed some recombinant analogs of lactaptin and compared their biological activity. Among these analogs, RL2 has the highest antitumor activity, but the amino acid residues and secondary structures that are responsible for RL2's activity remain unclear. To elucidate the structure-activity relations of RL2, we studied the structural and aggregation features of this fairly large intrinsically disordered fragment of human milk κ-casein by a combination of physicochemical methods: NMR, paramagnetic relaxation enhancement (PRE), Electron Paramagnetic Resonance (EPR), circular dichroism, dynamic light scattering, atomic force microscopy, and a cytotoxic activity assay. It was found that in solution, RL2 exists as stand-alone monomeric particles and large aggregates. Whereas the disulfide-bonded homodimer turned out to be more prone to assembly into large aggregates, the monomer predominantly forms single particles. NMR relaxation analysis of spin-labeled RL2 showed that the RL2 N-terminal region, which is essential not only for multimerization of the peptide but also for its proapoptotic action on cancer cells, is more ordered than its C-terminal counterpart and contains a site with a propensity for α-helical secondary structure.


Antineoplastic Agents/chemistry , Caseins/chemistry , Cell-Penetrating Peptides/chemistry , Intrinsically Disordered Proteins/chemistry , Amino Acid Sequence , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Caseins/biosynthesis , Caseins/genetics , Caseins/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell-Penetrating Peptides/biosynthesis , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/pharmacology , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Intrinsically Disordered Proteins/biosynthesis , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/pharmacology , MCF-7 Cells , Protein Aggregates/genetics , Protein Multimerization , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Structure-Activity Relationship
4.
Curr Med Sci ; 39(3): 349-355, 2019 Jun.
Article En | MEDLINE | ID: mdl-31209802

Peptides play crucial roles in various physiological and pathological processes. Consequently, the investigation of peptide-based drugs is a highlight in the research and development of new drugs. However, natural peptides are not always ideal choices for clinical application due to their limited number and sometimes cytotoxicity to normal cells. Aiming to gain stronger or specific or novel biological effects and overcome the disadvantages of natural peptides, artificial hybrid peptides have been designed by combining the sequence of two or more different peptides with varied biological functions. Compared to natural peptides, hybrid peptides have shown better therapeutic potentials against bacteria, tumors, and metabolic diseases. In this review, design strategies, structure features and recent development of hybrid peptides are summarized; future directions for the research and development of hybrid peptide drugs are also discussed.


Antimicrobial Cationic Peptides/chemical synthesis , Antineoplastic Agents/chemical synthesis , Cell-Penetrating Peptides/biosynthesis , Hypoglycemic Agents/chemical synthesis , Protein Engineering/methods , Recombinant Fusion Proteins/biosynthesis , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/therapeutic use , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/therapeutic use , Antimicrobial Cationic Peptides/therapeutic use , Antineoplastic Agents/therapeutic use , Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/therapeutic use , Cytotoxins/chemical synthesis , Cytotoxins/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Drug Design , Humans , Hypoglycemic Agents/therapeutic use , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/pathology , Obesity/drug therapy , Obesity/metabolism , Obesity/physiopathology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use
5.
Protein Expr Purif ; 129: 128-134, 2017 01.
Article En | MEDLINE | ID: mdl-27234002

Developing a recombinant vector for noninvasively delivering biological macromolecules into the brain is important. This study constructed and purified a protein complex based on the cholera toxin (CT) molecular structure. Enhanced green fluorescent protein (EGFP)-modified A2 subunits of CT (CTA2) were used as tracer molecules for introduction of transactivator of transcription (TAT) through the A subunit into cells. The protein complex EGFP-CTA2-TAT/(CTB)5 (CTB: B subunit of CT) was obtained using an in vitro recombination method and verified by monosialoganglioside-enzyme-linked immunosorbent assay and high performance liquid chromatography assay. The protein complexes bound more strongly to monosialoganglioside (GM1) than (CTB)5 at low concentrations (0.625-1.25 µg/mL). In vitro assays revealed that the transmembrane function of TAT was also maintained. The GM1-binding activity and cell membrane-penetrating ability suggested that a CT structure-based protein complexes could be used to design a delivery carrier for intranasal administration through GM1 binding. The expression vector introduced in this study provides a feasible expression frame for constructing several new macromolecular protein drugs for effective cell penetration.


Cell-Penetrating Peptides , Cholera Toxin , Drug Carriers/pharmacology , Cell Line, Tumor , Cell-Penetrating Peptides/biosynthesis , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/isolation & purification , Cell-Penetrating Peptides/pharmacology , Escherichia coli/genetics , Escherichia coli/metabolism , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/isolation & purification , Green Fluorescent Proteins/pharmacology , Humans , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology
6.
J Pept Sci ; 22(11-12): 689-699, 2016 Nov.
Article En | MEDLINE | ID: mdl-27739168

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.


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
7.
Sci Rep ; 6: 22404, 2016 Mar 03.
Article En | MEDLINE | ID: mdl-26935651

Depression is a disturbing psychiatric disease with unsatisfied therapy. Not all patients are sensitive to anti-depressants currently in use, side-effects are unavoidable during therapy, and the cases with effectiveness are always accompanied with delayed onset of clinical efficacy. Delivering brain-derived neurotrophic factor (BDNF) to brain seems to be a promising therapy. However, a better approach to delivery is still rudimentary. The purpose of our present work is to look for a rapid-onset and long-lasting therapeutic strategy for major depressive disorder (MDD) by effectively delivering BDNF to brain. BDNF, fused with cell-penetrating peptides (TAT and HA2), was packaged in adenovirus associated virus (AAV) to construct the BDNF-HA2TAT/AAV for intranasally delivering BDNF to central nervous system (CNS) via nose-brain pathway. Intranasal administration of BDNF-HA2TAT/AAV to normal mice displayed anti-depression effect in forced swimming test when the delivery lasted relatively longer. The AAV applied to mice subjected to chronic mild stress (CMS) through intranasal administration for 10 days also alleviated depression-like behaviors. Western-blotting analysis revealed that BDNF-HA2TAT/AAV nasal administration enhanced hippocampal BDNF content. These results indicate intranasal administration of constructed BDNF-HA2TAT/AAV exerts anti-depression effect in CMS mice by increasing hippocampal BDNF, suggesting that this strategy holds a promising therapeutic potential for MDD.


Brain-Derived Neurotrophic Factor , Cell-Penetrating Peptides , Dependovirus , Depressive Disorder, Major/therapy , Genetic Therapy/methods , Recombinant Fusion Proteins , Administration, Intranasal , Animals , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , Cell-Penetrating Peptides/biosynthesis , Cell-Penetrating Peptides/genetics , Depressive Disorder, Major/genetics , Depressive Disorder, Major/metabolism , Female , Humans , Male , Mice , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics
8.
Protein Expr Purif ; 111: 9-17, 2015 Jul.
Article En | MEDLINE | ID: mdl-25797209

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.


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
9.
Appl Biochem Biotechnol ; 175(6): 3025-37, 2015 Mar.
Article En | MEDLINE | ID: mdl-25586490

Cell-penetrating peptides are molecules with the ability to cross membranes and enter cells. Attention has been put on these peptides as a tool for drug delivery research, as they are able to serve as delivery vectors for large molecules. Intracellular delivery of bioactive peptides is a very promising research area for clinical applications, since peptides are able to simulate protein regions and thus modulate key intracellular protein-protein interactions. Therefore, evaluation of different strategies for production of these peptides is necessary. In this work, an intein-mediated system was used to evaluate Escherichia coli recombinant production of p53pAnt and PNC27 anticancer cell-penetrating peptides. It was demonstrated that the pTXB1 and the pTYB11 vector systems are suitable for production of this kind of peptides. The production process involves a low-temperature induction process and an efficient on-column intein-mediated cleavage, which allowed an effective peptide recovery using a single chromatographic step.


Cell-Penetrating Peptides/biosynthesis , Escherichia coli/metabolism , Inteins , Cell-Penetrating Peptides/genetics , Cloning, Molecular , Escherichia coli/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
10.
Sheng Wu Gong Cheng Xue Bao ; 30(8): 1247-55, 2014 Aug.
Article Zh | MEDLINE | ID: mdl-25423754

he aim of this study was to obtain a cell-penetrating cytoglobin (Cygb), which combines the transmembrane function of cell-penetrating peptides TAT with the anti-aging and anti-fibrotic role of cytoglobin. The Cygb gene was complexed with TAT gene by overlapping PCR, inserted into the vector pET22b to construct the recombinant expression plasmid (pET22b-TAT-Cygb) and then transformed into Escherichia coli BL21 (DE3). The fusion protein TAT-Cygb, whose expression was induced by lactose, was purified by CM Sepharose Fast Flow Protocol and verified by Western blotting. The final TAT-Cygb had a molecular weight of 23 kDa with 95% purity, as shown by SDS-PAGE. As demonstrated by bioactivity experiments, TAT-Cygb exhibited a high specific peroxidase activity up to (422.30 ± 0.36) U/mg. Both TAT-Cygb and Cygb pretreatment group could protect Hacat cells against oxidation of H2O2, but only TAT-Cygb treatment group could remedy cells injuried by H2O2 (RGR = 98%), which was significantly different from Cygb treatment group (RGR = 79%). We successfully obtained the bioactive and cell-penetrating fusion protein TAT-Cygb that has the potential application in anti-aging, anti-fibrotic and anti-cancer.


Cell-Penetrating Peptides/biosynthesis , Gene Products, tat , Globins/biosynthesis , Blotting, Western , Cell Line , Cytoglobin , Electrophoresis, Polyacrylamide Gel , Escherichia coli/metabolism , Genetic Vectors , Humans , Hydrogen Peroxide , Recombinant Fusion Proteins/biosynthesis
11.
Sheng Wu Gong Cheng Xue Bao ; 29(11): 1644-53, 2013 Nov.
Article Zh | MEDLINE | ID: mdl-24701829

The aim of the study is to establish a platform to deliver therapeutic proteins into target cells through a polyarginine-based cell penetrating peptide. To facilitate the expression of therapeutic proteins, a pSUMO (Small Ubiquitin-like Modifier)-R9-EGFP (enhanced green fluorescence protein) prokaryotic expression vector was constructed. After induction, the fusion protein SUMO-R9-EGFP was efficiently expressed. To validate the cell penetrating ability of the fusion protein, HepG2 cells were incubated with the purified R9-EGFP or EGFP protein as control, internalization of the fluorescent proteins was examined by either flow cytometry or confocal microscopy. The result obtained by flow cytometry showed that the R9-EGFP fusion protein could efficiently penetrate into the HepG2 cells in a dose and time-dependent manner. In contrast, the fluorescence was barely detected in the HepG2 cells incubated with EGFP control. The fluorescence intensity of the R9-EGFP treated cells reached plateau phase after 1.5 h. The result obtained by confocal microscopy shows that R9-EGFP efficiently entered into the HepG2 cells and was exclusively located in the cytoplasm, whereas, no fluorescence was detected in the cells incubated with the EGFP control. The heparin inhibition experiment showed that heparin could inhibit penetrating effect of the R9-EGFP protein by about 50%, suggesting that the penetrating ability of the fusion protein is heparin-dependent. In summary, the study has established a platform to deliver therapeutic proteins into target cells through a polyarginine-based penetrating peptide.


Green Fluorescent Proteins/biosynthesis , Peptides/metabolism , Recombinant Fusion Proteins/pharmacology , Cell-Penetrating Peptides/biosynthesis , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/pharmacology , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Hep G2 Cells , Humans , Peptides/genetics , Protein Transport , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics
12.
Mol Biol Rep ; 39(6): 7175-82, 2012 Jun.
Article En | MEDLINE | ID: mdl-22311040

Generation of induced pluripotent stem cells from endangered cattle breeds makes a significant contribution to the establishment of embryonic stem cell line, conservation and utilization of genetic resources in endangered cattle. In our study, we are trying to construct recombinant proteins of transcription factors Oct-4, Nanog, Sox2 and Lin28 for reprogramming of endangered Luxi cattle fibroblast cells in induced pluripotent stem cells. To test the cell permeability and stability of the recombinant proteins, we designed and fused a pep-1 protein transduction domain to the C-terminus of enhanced green fluorescent protein, we treated Luxi cattle fibroblast cells with the purified proteins at various concentrations by adding them to the cell culture media for 2-48 h and assayed cell morphology and protein presence. We found that the purified EGFP-tagged recombinant proteins readily entered cells at a concentration of 0.12 mg/ml within 2 h and some of them could translocate into nucleus. In addition, we found that the transduced proteins appeared to be stable inside cells for up to 48 h. Then codons of Oct-4, Nanog, Sox2 and Lin28 were optimized for high level of expression in E. coli., they were synthesized using DNA oligo based, PCR gene assembling method, the reprogramming factors were designed with optimized transcription factors fused a pep-1 protein transduction domain to the C-terminus, for high level protein expression of the reprogramming factors, inducer concentration, and induction time were tested, the optimum inducer concentration for the expression of reprogramming factors Oct-4, Nanog, Sox2 and Lin28 were 0.01 mM, the optimum induction time were 10, 8, 2 and 12 h, respectively.


Cell-Penetrating Peptides/genetics , Fibroblasts/metabolism , Recombinant Fusion Proteins/genetics , Animals , Cattle , Cell Dedifferentiation , Cell-Penetrating Peptides/biosynthesis , Cell-Penetrating Peptides/metabolism , Cells, Cultured , Cloning, Molecular , Codon , Endangered Species , Escherichia coli/genetics , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Octamer Transcription Factor-3/biosynthesis , Octamer Transcription Factor-3/genetics , Permeability , Protein Stability , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics
13.
J Dent Res ; 91(1): 90-6, 2012 Jan.
Article En | MEDLINE | ID: mdl-21990606

Human dental pulp stem cells (hDPSCs) are the only mesenchymal stem cells in pulp tissue that can differentiate into osteoblasts, odontoblasts, and adipose cells. The transcriptional co-activator with PDZ-binding motif (TAZ) protein has been reported to modulate osteogenic differentiation in mouse MSCs. Therefore, we examined whether the TAZ protein plays the same role in human pulp stem cells. In this study, TAZ was applied to cells directly with low-molecular-weight protamine (LMWP) as a cell-penetrating peptide (CPP). The LMWP-TAZ fusion proteins were expressed in an E. coli system with a pET-21b vector and efficiently transferred into hDPSCs without producing toxicity in the cells. The efficient uptake of TAZ was shown by Western blot with an anti-TAZ antibody, fluorescence-activated cell sorting, and confocal microscopy in live cells. The delivered TAZ protein increased osteogenic differentiation, as confirmed by alkaline phosphatase (ALP) staining, RT-PCR, and Western blotting. In addition, TAZ also inhibited adipogenic differentiation, regulating peroxisome proliferator-activated receptor-γ (PPAR-γ), lipoprotein lipase (LPL), and adipocyte fatty acid-binding protein (aP2) mRNA levels. These in vitro studies suggest that cell-permeable TAZ may be used as a specific regulator of hard-tissue differentiation.


Cell-Penetrating Peptides/pharmacology , Dental Pulp/cytology , Intracellular Signaling Peptides and Proteins/pharmacology , Mesenchymal Stem Cells/cytology , Odontogenesis/drug effects , Recombinant Fusion Proteins/pharmacology , Tooth Calcification/drug effects , Adipogenesis/drug effects , Analysis of Variance , Cell Differentiation/drug effects , Cell-Penetrating Peptides/biosynthesis , Endpoint Determination , Escherichia coli/genetics , Humans , Odontogenesis/genetics , Osteogenesis/drug effects , Osteogenesis/genetics , Protamines/pharmacology , Protein Transport , Recombinant Fusion Proteins/biosynthesis , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transduction, Genetic
14.
Biosci Rep ; 32(3): 271-9, 2012 Jun.
Article En | MEDLINE | ID: mdl-21679157

Accumulating evidence has demonstrated that FHIT (fragile histidine triad) is a bona fide tumour suppressor gene in a large fraction of human tumours, including hepatocellular cancer. A virus-based delivery system has been developed to transfer the FHIT gene into many types of cancer cells to inhibit growth or even induce apoptosis. However, a protein-based replacement strategy for FHIT has not been performed in cancer cells. Here, we used HIV-TAT (transactivator of transcription)-derived peptide to transfer the purified FHIT protein into HCC (hepatocellular carcinoma) cells and determine the biological effect of this fusion protein in inducing apoptosis. Affinity chromatography was used to purify TAT peptide-fused human FHIT (TAT-FHIT) protein from BL21 Escherichia coli. Immunofluorescence staining and Western blot analysis were performed to identify the expression and internalization of TAT-FHIT in HCC cells compared with the purified FHIT protein. Our study showed that TAT-FHIT protein can translocate into cancer cells in 1 h after incubation at 37°C. Furthermore, the results of MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide] assay, Annexin-V staining and Western blotting demonstrated that TAT-FHIT can robustly inhibit growth and induce apoptosis of HCC cells in vitro. In addition, a mechanistic study showed that both exogenous and intrinsic apoptotic pathways were involved in TAT-FHIT-mediated apoptosis and this effect could be attenuated partially by a mitochondrial protector TAT-BH4, indicating that mitochondrion plays a critical role in TAT-FHIT-mediated pro-apoptotic effect in cancer cells. Taken together, our study suggests that TAT-FHIT is a potential pro-apoptotic molecule in HCC cells and strengthen the hypothesis of its therapeutic application against HCC.


Acid Anhydride Hydrolases/pharmacology , Apoptosis/drug effects , Cell-Penetrating Peptides/pharmacology , Neoplasm Proteins/pharmacology , Recombinant Fusion Proteins/pharmacology , tat Gene Products, Human Immunodeficiency Virus/pharmacology , Acid Anhydride Hydrolases/biosynthesis , Carcinoma, Hepatocellular , Caspase 8/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell-Penetrating Peptides/biosynthesis , Drug Stability , Hep G2 Cells , Humans , Mitochondria/metabolism , Neoplasm Proteins/biosynthesis , Permeability , Recombinant Fusion Proteins/biosynthesis , tat Gene Products, Human Immunodeficiency Virus/biosynthesis
15.
J Biomed Mater Res A ; 100(1): 211-9, 2012 Jan.
Article En | MEDLINE | ID: mdl-22042599

Macromolecules present a remarkable potential as future therapeutics. However, their translation into clinical practice has been hampered by an inherently low bioavailability. Cell-penetrating peptides (CPP) have been recently shown to significantly improve on the bioavailability of macromolecules. Yet, the high cost associated with development and production of these peptides is a major factor hindering their rapid deployment beyond the laboratory. Here, we describe a facile and robust methodology for efficient and large-scale production of low-molecular-weight protamine-a potent CPP of great clinical potential. Our methodology is based on the immobilization of thermolysin, an enzyme catalyzing digestion of native protamine, on chemically surface-modified gels produced by silica sol-gel chemistry. Thermolysin was immobilized at extremely high matrix loading of 733 mg/g matrix and exhibited good thermal and pH stability, indicating robustness with respect to processing conditions. The mechanical properties of the silica matrix further allowed utilization of the immobilized thermolysin in both batch and packed-bed reactor systems to produce the LMWP peptide in high yields. Results presented here are of high significance as this efficient and cost-effective production of high purity LMWP could enable clinical translation of many potential macromolecular drugs.


Cell-Penetrating Peptides/biosynthesis , Drug Delivery Systems/methods , Enzymes, Immobilized/metabolism , Macromolecular Substances/metabolism , Protamines/chemistry , Protamines/metabolism , Thermolysin/metabolism , Bacillus/enzymology , Bioreactors , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Chromatography, Affinity , Gels , Glutaral/chemistry , Hydrogen-Ion Concentration/drug effects , Kinetics , Microscopy, Electron, Scanning , Molecular Weight , Phase Transition/drug effects , Porosity/drug effects , Protamines/pharmacology , Silicon Dioxide/chemistry , Surface Properties/drug effects , Temperature
16.
Sheng Wu Gong Cheng Xue Bao ; 27(9): 1371-8, 2011 Sep.
Article Zh | MEDLINE | ID: mdl-22117521

Hepatitis B virus X protein (HBx) has various functions and plays a crucial role in the development of hepatocellular carcinoma (HCC). However, due to different transfection efficiency levels and experimental approaches, it is difficult to correlate the exact functions of HBx to HBV-associated HCC. In this study, we constructed two prokaryotic expression vectors, pGEX-HBx-EGFP-TLM and pGEX-EGFP-TLM, which expressed HBx-EGFP-TLM and EGFP-TLM fusion proteins respectively. Both vectors contained a coding sequence of TLM transduction motif derived from the PreS2-domain of Hepatitis B Virus surface antigens. In addition, EGFP was expressed as a reporter reflecting the transduction efficiency of TLM. The fusion protein HBx-EGFP-TLM or EGFP-TLM purified from Escherichia coli BL21(DE3) by AKTA Purifier system was incubated with AML12 and SMMC-7721 cells. Both Western blotting and laser confocal results indicated that the translocation motif TLM could lead HBx-EGFP and EGFP into the cytoplasm. Dual-Luciferase Reporter Assay revealed that the activity of mEZH2 promoter could be up-regulated by the recombinant HBx. In conclusion, we expressed a cell-permeable HBx, which could provide a new method to study the functions of HBx.


Cell-Penetrating Peptides/biosynthesis , Green Fluorescent Proteins/biosynthesis , Hepatitis B Surface Antigens/genetics , Protein Precursors/genetics , Recombinant Fusion Proteins/biosynthesis , Trans-Activators/biosynthesis , Amino Acid Motifs/genetics , Cell-Penetrating Peptides/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Trans-Activators/genetics , Viral Regulatory and Accessory Proteins
17.
BMC Biotechnol ; 10: 79, 2010 Oct 28.
Article En | MEDLINE | ID: mdl-21029412

BACKGROUND: For improved uptake of oligonucleotide-based therapy, the oligonucleotides often are coupled to peptides that facilitate entry into cells. To this end, novel cell-penetrating peptides (CPPs) were designed for mediating intracellular uptake of oligonucleotide-based therapeutics. The novel peptides were based on taking advantage of the nuclear localization properties of transcription factors in combination with a peptide that would bind putatively to cell surfaces. It was observed that adding a glutamate peptide to the N-terminus of the nuclear localization signal (NLS) of the Oct6 transcription factor resulted in a novel CPP with better uptake and better nuclear colocalization than any other peptide tested. RESULTS: Uptake of the novel peptide Glu-Oct6 by cancer cell lines was rapid (in less than 1 hr, more than 60% of DU-145 cells were positive for FITC), complete (by 4 hr, 99% of cells were positive for FITC), concentration-dependent, temperature-dependent, and inhibited by sodium azide (NaN3). Substitution of Phe, Tyr, or Asn moieties for the glutamate portion of the novel peptide resulted in abrogation of novel CPP uptake; however none of the substituted peptides inhibited uptake of the novel CPP when coincubated with cells. Live-cell imaging and analysis by imaging flow cytometry revealed that the novel CPP accumulated in nuclei. Finally, the novel CPP was coupled to a carboxyfluorescein-labeled synthetic oligonucleotide, to see if the peptide could ferry a therapeutic payload into cells. CONCLUSIONS: These studies document the creation of a novel CPP consisting of a glutamate peptide coupled to the N-terminus of the Oct6 NLS; the novel CPP exhibited nuclear colocalization as well as uptake by prostate and pancreatic cancer cell lines.


Cell-Penetrating Peptides/biosynthesis , Nuclear Localization Signals/chemistry , Pancreatic Neoplasms/metabolism , Prostatic Neoplasms/metabolism , Cell Line, Tumor , Glutamic Acid/chemistry , Humans , Male , Organic Cation Transport Proteins/chemistry , Peptide Nucleic Acids/biosynthesis
18.
Biochim Biophys Acta ; 1798(12): 2249-57, 2010 Dec.
Article En | MEDLINE | ID: mdl-20170629

Expressed protein ligation (EPL) is a useful method for the native chemical ligation of proteins with other proteins or peptides. This study assessed the practicability of EPL in the preparation of fusion proteins of enhanced green fluorescent protein (EGFP) with chemically synthesized cell-penetrating peptides (CPPs) for intracellular delivery. Using intein-mediated purification with an affinity chitin-binding tag (IMPACT) system, the thioester of EGFP (EGFP-SR) was prepared. Optimization of the ligation of EGFP-SR with arginine 12-mer (R12) produced the fusion protein in high yield. The EPL procedure also allows the preparation of EGFP-R12 containing a low level of endotoxin (ET), via the satisfactory ET removal of EGFP-SR prior to ligation with the R12 peptide. Fusion proteins of EGFP with R12 and the d-isomer of R12 prepared by EPL showed similar levels of cellular uptake compared to the fusion protein directly expressed in Escherichiacoli.


Endotoxins , Recombinant Fusion Proteins , Animals , Cell-Penetrating Peptides/biosynthesis , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/isolation & purification , Cell-Penetrating Peptides/pharmacology , Chromatography, Affinity/methods , Escherichia coli , Gene Expression , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/pharmacology , HeLa Cells , Humans , Inteins , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacology
...