Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Am Soc Mass Spectrom ; 30(9): 1779-1789, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31250320

ABSTRACT

Peptides represent a promising modality for the design of novel therapeutics that can potentially modulate traditionally non-druggable targets. Cell-penetrating peptides (CPPs) and antimicrobial peptides (AMPs) are two large families that are being explored extensively as drug delivery vehicles, imaging reagents, or therapeutic treatments for various diseases. Many CPPs and AMPs are cationic among which a significant portion is extremely basic and hydrophilic (e.g., nona-arginine). Despite their attractive therapeutic potential, it remains challenging to directly analyze and quantify these super cationic peptides from biological matrices due to their poor chromatographic behavior and MS response. Herein, we describe a generic method that combines solid phase extraction and LC-MS/MS for analysis of these peptides. As demonstrated, using a dozen strongly basic peptides, low µM concentration of perfluoropentanoic acid (PFPeA) in the mobile phase enabled excellent compound chromatographic retention, thus avoiding co-elution with solvent front ion suppressants. PFPeA also had a charge reduction effect that allowed the selection of parent/ion fragment pairs in the higher m/z region to further reduce potential low molecular weight interferences. When the method was coupled to the optimized sample extraction process, we routinely achieved low digit ng/ml sensitivity for peptides in plasma/tissue. The method allowed an efficient evaluation of plasma stability of CPPs/AMPs without fluorescence derivatization or other tagging methods. Importantly, using the widely studied HIV-TAT CPP as an example, the method enabled us to directly assess its pharmacokinetics and tissue distribution in preclinical animal models.


Subject(s)
Chromatography, Liquid/methods , Mass Spectrometry/methods , Pentanoic Acids/chemistry , Peptides/analysis , Peptides/pharmacokinetics , Animals , Antimicrobial Cationic Peptides/analysis , Antimicrobial Cationic Peptides/chemistry , Cell-Penetrating Peptides/analysis , Chemical Precipitation , Drug Stability , Fluorocarbons , Hydrophobic and Hydrophilic Interactions , Male , Peptides/chemistry , Rats, Wistar , Solid Phase Extraction , Tissue Distribution , Trichloroacetic Acid/chemistry , tat Gene Products, Human Immunodeficiency Virus/analysis , tat Gene Products, Human Immunodeficiency Virus/pharmacokinetics
2.
ACS Appl Mater Interfaces ; 8(28): 17878-84, 2016 Jul 20.
Article in English | MEDLINE | ID: mdl-27400339

ABSTRACT

Bone marrow-derived mesenchymal stem cells (BMSCs) have shown great potential for bone repair due to their strong proliferation ability and osteogenic capacity. To evaluate and improve the stem cell-based therapy, long-term tracking of stem cell differentiation into bone-forming osteoblasts is required. However, conventional fluorescent trackers such as fluorescent proteins, quantum dots, and fluorophores with aggregation-caused quenching (ACQ) characteristics have intrinsic limitations of possible interference with stem cell differentiation, heavy metal cytotoxicity, and self-quenching at a high labeling intensity. Herein, we developed aggregation-induced emission nanoparticles decorated with the Tat peptide (AIE-Tat NPs) for long-term tracking of the osteogenic differentiation of mouse BMSCs without interference of cell viability and differentiation ability. Compared with the ability of the commercial Qtracker 655 for tracking of only 6 passages of mouse BMSCs, AIE-Tat NPs have shown a much superior performance in long-term tracking for over 12 passages. Moreover, long-term tracking of the osteogenic differentiation process of mouse BMSCs was successfully conducted on the biocompatible hydroxyapatite scaffold, which is widely used in bone tissue engineering. Thus, AIE-Tat NPs have promising applications in tracking stem cell fate for bone repair.


Subject(s)
Bone Marrow Cells/cytology , Fluorescent Dyes/analysis , Mesenchymal Stem Cells/cytology , Nanoparticles/analysis , Osteogenesis/physiology , Peptide Fragments/analysis , tat Gene Products, Human Immunodeficiency Virus/analysis , Animals , Cell Differentiation/physiology , Cells, Cultured , Fluorescent Dyes/chemistry , Mice , Nanoparticles/chemistry , Peptide Fragments/chemistry , Spectrophotometry, Ultraviolet , Tissue Engineering , tat Gene Products, Human Immunodeficiency Virus/chemistry
3.
Methods Mol Biol ; 1354: 329-42, 2016.
Article in English | MEDLINE | ID: mdl-26714722

ABSTRACT

HIV-1 Tat is efficiently secreted by HIV-1-infected or Tat-transfected cells. Accordingly, Tat concentrations in the nanomolar range have been measured in the sera of HIV-1-infected patients, and this protein acts as a viral toxin on bystander cells. Nevertheless, assaying Tat concentration in media or sera is not that straightforward because extracellular Tat is unstable and particularly sensitive to oxidation. Moreover, most anti-Tat antibodies display limited affinity. Here, we describe methods to quantify extracellular Tat using a sandwich ELISA or Western blotting when Tat is secreted by suspension or adherent cells, respectively. In both cases it is important to capture exported Tat using antibodies before any Tat oxidation occurs; otherwise it will become denatured and unreactive toward antibodies.


Subject(s)
Blotting, Western/methods , Enzyme-Linked Immunosorbent Assay/methods , HIV Infections/virology , HIV-1/isolation & purification , tat Gene Products, Human Immunodeficiency Virus/analysis , Animals , Cell Culture Techniques/methods , Cell Line , Humans , Jurkat Cells , Rats
4.
Bioanalysis ; 7(8): 981-90, 2015.
Article in English | MEDLINE | ID: mdl-25633639

ABSTRACT

BACKGROUND: While HIV-1 TAT peptide-conjugation shows great promise on improving intracellular delivery of biotherapeutics in vitro and in vivo, quantification of TAT-fusion therapeutics in biological matrices represents a daunting challenge. MATERIALS & METHODS: A sensitive MS approach for accurate quantification of intact TAT-fusion protein/polypeptide in plasma was developed. i) A semi-automated 96-well ion-exchange solid phase extraction was developed; ii) a rapid LC separation on C4 was devised; iii) a TAT-fusion analog was constructed as internal standard. RESULTS: We reported that low percentage of supercharging reagents enabled a significant sensitivity improvement of MS for intact TAT-fusion protein/polypeptide analysis. We showed a proof of concept by successfully developing a sensitive LC/MRM-MS method for quantifying GAP161, a TAT-conjugating RasGAP mimics, in rat plasma. CONCLUSION: This work represents the first quantification of TAT-fusion therapeutics in biological samples by an LC-MS based method.


Subject(s)
Chromatography, Liquid/methods , Gonadotropin-Releasing Hormone/metabolism , Protein Precursors/metabolism , Tandem Mass Spectrometry/methods , ras GTPase-Activating Proteins/metabolism , tat Gene Products, Human Immunodeficiency Virus/analysis , Animals , Humans , Male , Rats , Rats, Sprague-Dawley , Solid Phase Extraction/methods
5.
Nat Nanotechnol ; 9(3): 198-203, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24561356

ABSTRACT

A detailed understanding of the cellular uptake process is essential to the development of cellular delivery strategies and to the study of viral trafficking. However, visualization of the entire process, encompassing the fast dynamics (local to the freely diffusing nanoparticle) as well the state of the larger-scale cellular environment, remains challenging. Here, we introduce a three-dimensional multi-resolution method to capture, in real time, the transient events leading to cellular binding and uptake of peptide (HIV1-Tat)-modified nanoparticles. Applying this new method to observe the landing of nanoparticles on the cellular contour in three dimensions revealed long-range deceleration of the delivery particle, possibly due to interactions with cellular receptors. Furthermore, by using the nanoparticle as a nanoscale 'dynamics pen', we discovered an unexpected correlation between small membrane terrain structures and local nanoparticle dynamics. This approach could help to reveal the hidden mechanistic steps in a variety of multiscale processes.


Subject(s)
Imaging, Three-Dimensional/methods , Nanoparticles/metabolism , Peptide Fragments/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Cell Membrane/metabolism , Cell Membrane Permeability , Mice , Microscopy/methods , Models, Molecular , NIH 3T3 Cells , Nanoparticles/analysis , Peptide Fragments/analysis , tat Gene Products, Human Immunodeficiency Virus/analysis
6.
Mol Cell Biol ; 33(15): 3036-49, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23732912

ABSTRACT

The mechanisms underlying the cellular entry of the HIV-1 Tat protein transduction domain (TatP) and the molecular information necessary to improve the transduction efficiency of TatP remain unclear due to the technical limitations for direct visualization of TatP's behavior in cells. Using confocal microscopy, total internal reflection fluorescence microscopy, and four-dimensional microscopy, we developed a single-molecule tracking assay for TatP labeled with quantum dots (QDs) to examine the kinetics of TatP initially and immediately before, at the beginning of, and immediately after entry into living cells. We report that even when the number of multivalent TatP (mTatP)-QDs bound to a cell was low, each single mTatP-QD first locally induced the cell's lateral transport machinery to move the mTatP-QD toward the center of the cell body upon cross-linking of heparan sulfate proteoglycans. The centripetal and lateral movements were linked to the integrity and flow of actomyosin and microtubules. Individual mTatP underwent lipid raft-mediated temporal confinement, followed by complete immobilization, which ultimately led to endocytotic internalization. However, bivalent TatP did not sufficiently promote either cell surface movement or internalization. Together, these findings provide clues regarding the mechanisms of TatP cell entry and indicate that increasing the valence of TatP on nanoparticles allows them to behave as cargo delivery nanomachines.


Subject(s)
Endocytosis , HIV Infections/virology , HIV-1/physiology , Host-Pathogen Interactions , Virus Internalization , tat Gene Products, Human Immunodeficiency Virus/analysis , tat Gene Products, Human Immunodeficiency Virus/metabolism , Actins/metabolism , HIV Infections/metabolism , HeLa Cells , Humans , Membrane Microdomains/metabolism , Membrane Microdomains/virology , Microscopy, Confocal/methods , Microtubules/metabolism , Microtubules/virology , Molecular Imaging/methods , Pseudopodia/metabolism , Pseudopodia/virology , Quantum Dots
7.
Biosens Bioelectron ; 40(1): 277-82, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-22975093

ABSTRACT

Diamond is a promising material for merging solid-state and biological systems owing to its chemical stability, low background current, wide potential window and biocompatibility. The effects of surface charge density on human immunodeficiency virus type 1 Trans-activator transcription (HIV-1 Tat) protein binding have been investigated on a diamond field-effect transistor (FET) using ribonucleic acid (RNA) aptamers as a sensing element on a solid surface. A change in the gate potential of 91.6 mV was observed, whereby a shift in the negative direction was observed at a source-drain current of -8 µA in the presence of HIV-1 Tat protein bound to the RNA aptamers. Moreover, the reversible change in gate potential caused by the binding and regeneration cycles was very stable throughout cyclical detections. The stable immobilization is achieved via RNA aptamers covalently bonded to the carboxyl-terminated terephtalic acids on amine sites, thereby increasing the sensitivity of the HIV-1 Tat protein sensor. The reliable use of a real sample of HIV-1 Tat protein by an aptamer-FET was demonstrated for the first time, which showed the potential of diamond biointerfaces in clinical biosensor applications.


Subject(s)
Aptamers, Nucleotide/chemistry , Biosensing Techniques/instrumentation , Conductometry/instrumentation , Diamond/chemistry , HIV-1/metabolism , Transistors, Electronic , tat Gene Products, Human Immunodeficiency Virus/analysis , Electrodes , Equipment Design , Equipment Failure Analysis , HIV-1/isolation & purification , Reproducibility of Results , Sensitivity and Specificity , tat Gene Products, Human Immunodeficiency Virus/chemistry
8.
Chem Biol Drug Des ; 79(5): 683-90, 2012 May.
Article in English | MEDLINE | ID: mdl-22188730

ABSTRACT

To understand the protein transduction domain (PTD)-mediated protein transduction behavior and to explore its potential in delivering biopharmaceutic drugs, we prepared four TAT-EGFP conjugates: TAT(+)-EGFP, TAT(-)-EGFP, EGFP-TAT(+) and EGFP-TAT(-), where TAT(+) and TAT(-) represent the original and the reversed TAT sequence, respectively. These four TAT-EGFP conjugates were incubated with HeLa and PC12 cells for in vitro study as well as injected intraperitoneally to mice for in vivo study. Flow cytometric results showed that four TAT-EGFP conjugates were able to traverse HeLa and PC12 cells with almost equal transduction efficiency. The in vivo study showed that the TAT-EGFP conjugates could be delivered into different organs of mice with different transduction capabilities. Bioinformatic analyses and CD spectroscopic data revealed that the TAT peptide has no defined secondary structure, and conjugating the TAT peptide to the EGFP cargo protein would not alter the native structure and the function of the EGFP protein. These results conclude that the sequence orientation, the spatial structure, and the relative location of the TAT peptide have much less effect on the TAT-mediated protein transduction. Thus, the TAT-fused conjugates could be constructed in more convenient and flexible formats for a wide range of biopharmaceutical applications.


Subject(s)
Fluorescent Dyes/analysis , Green Fluorescent Proteins/analysis , HIV/genetics , Transduction, Genetic , tat Gene Products, Human Immunodeficiency Virus/analysis , tat Gene Products, Human Immunodeficiency Virus/genetics , Amino Acid Sequence , Animals , Fluorescent Dyes/chemistry , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , HIV/chemistry , HeLa Cells , Humans , Mice , PC12 Cells , Protein Structure, Secondary , Rats , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , tat Gene Products, Human Immunodeficiency Virus/chemistry
9.
Nucleic Acids Res ; 40(5): 1904-15, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22067449

ABSTRACT

Microglial cells are the main HIV-1 targets in the central nervous system (CNS) and constitute an important reservoir of latently infected cells. Establishment and persistence of these reservoirs rely on the chromatin structure of the integrated proviruses. We have previously demonstrated that the cellular cofactor CTIP2 forces heterochromatin formation and HIV-1 gene silencing by recruiting HDAC and HMT activities at the integrated viral promoter. In the present work, we report that the histone demethylase LSD1 represses HIV-1 transcription and viral expression in a synergistic manner with CTIP2. We show that recruitment of LSD1 at the HIV-1 proximal promoter is associated with both H3K4me3 and H3K9me3 epigenetic marks. Finally, our data suggest that LSD1-induced H3K4 trimethylation is linked to hSET1 recruitment at the integrated provirus.


Subject(s)
Gene Silencing , HIV-1/genetics , Histone Demethylases/metabolism , Microglia/virology , Repressor Proteins/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/metabolism , Cell Line , Cell Nucleus/chemistry , Cell Nucleus/virology , Epigenesis, Genetic , HIV Long Terminal Repeat , HIV-1/physiology , Histone Demethylases/analysis , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Methylation , Promoter Regions, Genetic , Repressor Proteins/analysis , Tumor Suppressor Proteins/analysis , Virus Replication , tat Gene Products, Human Immunodeficiency Virus/analysis
10.
EMBO J ; 29(8): 1348-62, 2010 Apr 21.
Article in English | MEDLINE | ID: mdl-20224549

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) transcription relies on its transactivating Tat protein. Although devoid of a signal sequence, Tat is released by infected cells and secreted Tat can affect uninfected cells, thereby contributing to HIV-1 pathogenesis. The mechanism and the efficiency of Tat export remained to be documented. Here, we show that, in HIV-1-infected primary CD4(+) T-cells that are the main targets of the virus, Tat accumulates at the plasma membrane because of its specific binding to phosphatidylinositol-4,5-bisphosphate (PI(4,5)P(2)). This interaction is driven by a specific motif of the Tat basic domain that recognizes a single PI(4,5)P(2) molecule and is stabilized by membrane insertion of Tat tryptophan side chain. This original recognition mechanism enables binding to membrane-embedded PI(4,5)P(2) only, but with an unusually high affinity that allows Tat to perturb the PI(4,5)P(2)-mediated recruitment of cellular proteins. Tat-PI(4,5)P(2) interaction is strictly required for Tat secretion, a process that is very efficient, as approximately 2/3 of Tat are exported by HIV-1-infected cells during their lifespan. The function of extracellular Tat in HIV-1 infection might thus be more significant than earlier thought.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , HIV-1/pathogenicity , Phosphatidylinositol 4,5-Diphosphate/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Binding Sites , Cell Membrane/metabolism , Enzyme-Linked Immunosorbent Assay , HIV-1/growth & development , Humans , Jurkat Cells , Protein Binding , tat Gene Products, Human Immunodeficiency Virus/analysis
11.
Int J Mol Med ; 25(1): 41-51, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19956900

ABSTRACT

A number of cell-penetrating peptides (CPPs) have been reported, but their transduction efficiencies are too low to be used as intracellular carriers for therapeutic purposes. We conducted a comprehensive search to find novel CPPs using an in vitro virus (IVV) library, which presented random peptides consisting of 15 amino acids (diversity of the library was >10(12)). We found 9 kinds of novel CPPs with an intracellular translocation efficiency higher than that of the TAT peptide (YGRKKKRRQRRR). Interestingly, one of the novel CPPs, No. 14 (KLWMRWYSPTTRRYG), showed a dramatic improvement in translocation activity relative to the TAT peptide in CHO cells (>10-fold efficiency in 50 microM). As the intracellular translocation efficiency of No. 14 was increased by substitution Arg for Lys1 (14-1), we carried out alanine scanning on the basis of 14-1 to determine important amino acids for the intracellular translocation. The Ala substitution analysis showed that both Arg and Trp residues were important for the cell-penetrating activity and that their contribution was in the order Trp3

Subject(s)
Cell Membrane Permeability , HIV-1/genetics , Peptide Library , Peptides/analysis , Peptides/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Sequence , Amino Acids/analysis , Animals , Base Sequence , CHO Cells , Cricetinae , Cricetulus , HeLa Cells , Humans , Jurkat Cells , Molecular Sequence Data , Peptides/genetics , tat Gene Products, Human Immunodeficiency Virus/analysis , tat Gene Products, Human Immunodeficiency Virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...