Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
ACS Appl Mater Interfaces ; 11(35): 31638-31648, 2019 Sep 04.
Article in English | MEDLINE | ID: mdl-31389684

ABSTRACT

Responsive delivery of anticancer proteins into cells is an emerging field in biological therapeutics. Currently, the delivery of proteins is highly compromised by multiple successive physiological barriers that reduce the therapeutic efficacy. Hence, there is a need to design a robust and sustainable nanocarrier to provide suitable protection of proteins and overcome the physiological barriers for better cellular accumulation. In this work, polyethylenimine (PEI) cross-linked by oxaliplatin(IV) prodrug (oxliPt(IV)) was used to fabricate a redox-responsive nanocomplex (PEI-oxliPt(IV)@RNBC/GOD) for the delivery of a reactive oxygen species-cleavable, reversibly caged RNase A protein (i.e., RNase A nitrophenylboronic conjugate, RNBC) and glucose oxidase (GOD) in order to realize efficient cancer treatment. The generation of hydrogen peroxide by GOD can uncage and restore the enzymatic activity of RNBC. On account of the responsiveness of the nanocomplex to highly reducing cellular environment, it would dissociate and release the protein and active oxaliplatin drug, causing cell death by both catalyzing RNA degradation and inhibiting DNA synthesis. As assessed by the RNA degradation assay, the activity of the encapsulated RNBC was recovered by the catalytic production of hydrogen peroxide from GOD and glucose substrate overexpressed in cancer cells. Monitoring of the changes in nanoparticle size confirmed that the nanocomplex could dissociate in the reducing environment, with the release of active oxaliplatin drug and protein. Confocal laser scanning microscopy (CLSM) and flow cytometry analysis revealed highly efficient accumulation of the nanocomplex as compared to free native proteins. In vitro cytotoxicity experiments using 4T1 cancer cells showed ∼80% cell killing efficacy, with highly efficient apoptosis induction. Assisted by the cationic polymeric carrier, it was evident from CLSM images that intracellular delivery of the therapeutic protein significantly depleted the RNA level. Thus, this work provides a promising platform for the delivery of therapeutic proteins and chemotherapeutic drugs for efficient cancer treatment.


Subject(s)
Neoplasms , Oxaliplatin , Prodrugs , Ribonuclease, Pancreatic , Antineoplastic Agents , Cell Line, Tumor , DNA, Neoplasm/biosynthesis , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Humans , Hydrogen Peroxide/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Oxaliplatin/chemistry , Oxaliplatin/pharmacokinetics , Oxaliplatin/pharmacology , Oxidation-Reduction , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , RNA Stability/drug effects , Ribonuclease, Pancreatic/chemistry , Ribonuclease, Pancreatic/pharmacokinetics , Ribonuclease, Pancreatic/pharmacology
2.
Nano Lett ; 18(4): 2294-2303, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29547698

ABSTRACT

Protein therapeutics hold increasing interest with the promise of revolutionizing the cancer treatment by virtue of a potent specific activity and reduced adverse effects. Nonetheless, the therapeutic efficacy of anticancer proteins is highly compromised by multiple successive physiological barriers to protein delivery. In addition, concurrent elimination of bulk tumor cells and highly tumorigenic cancer stem-like cells (CSCs) as a promising strategy has been evidenced to significantly improve cancer therapy. Here we show that a hierarchically assembled nanocomposite can self-adaptively transform its particulate property in response to endogenous tumor-associated signals to overcome the sequential barriers and achieve an enhanced antitumor efficacy by killing CSCs and bulk tumor cells synchronously. The nanoassemblies preferentially accumulate in tumors and dissociate under tumor microenvironmental acidity accompanied by the extracellular release of small-sized ribonuclease A (RNase A)-encapsulated nanocapsule (R-rNC) and small-molecule anti-CSC doxycycline (Doc), which exhibit increased tumor penetration and intracellular accumulation. The endocytosed R-rNC rapidly releases RNase A within both CSCs and tumor cells at intracellular reductive conditions, causing cell death by catalyzing RNA degradation, while Doc eradicates CSCs by inhibiting the mitochondrial biogenesis. The hierarchical assemblies show enhanced cytotoxicity on the CSC-enriched MDA-MB-231 mammospheres and an enhanced antitumor efficacy on the xenograft tumor mouse model.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Breast Neoplasms/drug therapy , Delayed-Action Preparations/chemistry , Doxycycline/administration & dosage , Nanostructures/chemistry , Neoplastic Stem Cells/drug effects , Ribonuclease, Pancreatic/administration & dosage , Animals , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Doxycycline/pharmacokinetics , Doxycycline/pharmacology , Doxycycline/therapeutic use , Drug Delivery Systems , Female , Humans , Mice , Ribonuclease, Pancreatic/pharmacokinetics , Ribonuclease, Pancreatic/pharmacology , Ribonuclease, Pancreatic/therapeutic use
3.
PLoS One ; 13(2): e0193302, 2018.
Article in English | MEDLINE | ID: mdl-29486010

ABSTRACT

Angiogenin (ANG), a member of the RNase superfamily (also known as RNase 5) has neurotrophic, neuroprotective and angiogenic activities. Recently it has also been shown to be important in stem cell homeostasis. Mutations in ANG are associated with neurodegenerative diseases such as Amyotrophic Lateral Sclerosis (ALS) and Fronto-temporal dementia (FTD). ANG is a secreted protein which is taken up by cells and translocated to the nucleus. However, the import pathway/s through which ANG is taken up is/are still largely unclear. We have characterised the uptake of ANG in neuronal, astrocytic and microglial cell lines as well as primary neurons and astrocytes using pharmacological agents as well as dominant negative dynamin and Rab5 to perturb uptake and intracellular trafficking. We find that uptake of ANG is largely clathrin/dynamin independent and microtubule depolymerisation has a marginal effect. Perturbation of membrane ruffling and macropinocytosis significantly inhibited ANG uptake suggesting an uptake mechanism similar to RNase A. Our findings shed light on why mutations which do not overtly affect RNase activity but cause impaired localization are associated with neurodegenerative disease.


Subject(s)
Astrocytes/metabolism , Dynamins/metabolism , Microglia/metabolism , Neurons/metabolism , Ribonuclease, Pancreatic/pharmacokinetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Astrocytes/drug effects , Cell Line , Clathrin/metabolism , Humans , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice , Microglia/drug effects , Neurons/drug effects
4.
Biomacromolecules ; 18(8): 2509-2520, 2017 Aug 14.
Article in English | MEDLINE | ID: mdl-28649842

ABSTRACT

Here, we report novel lipo-oligoaminoamide nanoformulations for targeted intracellular protein delivery. Formulations are generated by first bioreversibly conjugating a sequence-defined amphiphilic lipo-oligomer 728 to the cargo protein via disulfide bonds, followed by formulation of the formed 728-SS-protein conjugate with different helper lipids in various compositions. The triblock oligoaminoamide 728 contains cysteines for reversible covalent protein conjugation and cross-link-stabilization of formed nanoparticles, polyethylene glycol (PEG) for shielding, and providing a hydrophilic domain, eight cationizable succinoyl tetraethylene pentamine (Stp) repeats for endosomal buffering and escape into the cytosol, and a tetra-oleic acid block for hydrophobic stabilization. The added helper lipids are supposed to enhance serum stability of the nanoparticles and provide targeting by lipid-anchored folic acid (FA)-PEG. The optimized protein nanoparticles, including 728, DOPS, cholesterol, DMPE-PEG2000, and the FA-PEG conjugated lipid 1042, presented a high colloidal stability without significant size increase in 72 h. Using cytotoxic ribonuclease A (RNase A) as cargo protein, FA-728-DOPS-DMPE-RNase A nanoformulation could be identified with highest potency of targeted RNase A-mediated folate-receptor-positive KB carcinoma cell killing among all tested formulations, resulting in 85% KB cell killing at a low concentration of 2 µM. These approximately 50 nm sized nanoparticles induced superior 70% KB cell killing even in the presence of 20% serum. Efficient targeted cytosolic delivery by coformulation with helper lipids was also demonstrated by FA-728-DOPS-DMPE-nlsEGFP nanoformulation using enhanced green fluorescent protein (EGFP) as cargo. Furthermore, partial nlsEGFP was imported into the nuclei of KB cells, validating effective endosomal escape, and following nuclear transport mediated by nuclear localization signal on nlsEGFP. As demonstrated, the screening and optimization of nanoformulations with helper lipids and coformulation agents is considered to be an important and rational next step in the development of intracellular biopharmaceuticals, following initial protein conjugate synthesis.


Subject(s)
Drug Delivery Systems/methods , Nanoparticles/chemistry , Phosphatidylethanolamines , Polyethylene Glycols , Ribonuclease, Pancreatic , Animals , Cell Line, Tumor , Humans , Mice , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/pharmacokinetics , Phosphatidylethanolamines/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/pharmacology , Ribonuclease, Pancreatic/chemistry , Ribonuclease, Pancreatic/pharmacokinetics , Ribonuclease, Pancreatic/pharmacology
5.
Biosci Biotechnol Biochem ; 81(3): 604-607, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27756186

ABSTRACT

Bovine angiogenin is a major component of the bone resorption inhibitory activity of milk basic protein (MBP). The intestinal absorption of bovine angiogenin was investigated in a rat model, where it was detected in an intact form in the peripheral blood after the oral administration of MBP. This finding demonstrates that orally administered bovine angiogenin is absorbed without being degraded.


Subject(s)
Milk Proteins/administration & dosage , Ribonuclease, Pancreatic/blood , Ribonuclease, Pancreatic/pharmacokinetics , Administration, Oral , Animals , Cattle , Intestinal Absorption , Male , Milk Proteins/pharmacokinetics , Rats, Sprague-Dawley
6.
J Control Release ; 183: 35-42, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24657950

ABSTRACT

The original aim of the study was to investigate the transdermal iontophoretic delivery of lysozyme and to gain further insight into the factors controlling protein electrotransport. Initial experiments were done using porcine skin. Lysozyme transport was quantified by using an activity assay based on the lysis of Micrococcus lysodeikticus and was corrected for the release of endogenous enzyme from the skin during current application. Cumulative iontophoretic permeation of lysozyme during 8h at 0.5mA/cm(2) (0.7mM; pH6) was surprisingly low (5.37±3.46µg/cm(2) in 8h) as compared to electrotransport of cytochrome c (Cyt c) and ribonuclease A (RNase A) under similar conditions (923.0±496.1 and 170.71±92.13µg/cm(2), respectively) - despite its having a higher electrophoretic mobility. The focus of the study then became to understand and explain the causes of its poor iontophoretic transport. Lowering formulation pH to 5 increased histidine protonation in the protein and decreased the ionisation of fixed negative charges in the skin (pI ~4.5) and resulted in a small but statistically significant increase in permeation. Co-iontophoresis of acetaminophen revealed a significant inhibition of electroosmosis; inhibition factors of 12-16 were indicative of strong lysozyme binding to skin. Intriguingly, lidocaine electrotransport, which is due almost exclusively to electromigration, was also decreased (approximately 2.7-fold) following skin pre-treatment by lysozyme iontophoresis (cf. iontophoresis of buffer solution) - suggesting that lysozyme was also able to influence subsequent cation electromigration. In order to elucidate the site of skin binding, different porcine skin models were tested (dermatomed skin with thicknesses of 250 and 750µm, tape-stripped skin and heat-separated dermis). Although no difference was seen between permeation across 250 and 750µm dermatomed skin (13.57±12.20 and 5.37±3.46µg/cm(2), respectively), there was a statistically significant increase across tape-stripped skin and heat-separated dermis (36.86±7.48 and 43.42±13.11µg/cm(2), respectively) - although transport was still much less than that seen across intact skin for Cyt c or RNase A. Furthermore, electroosmotic inhibition factors fell to 2.2 and 1.0 for tape-stripped skin and heat-separated dermis - indicating that lysozyme affected convective solvent flow through interactions with the epidermis and predominantly the stratum corneum. Finally, cation exchange and hydrophobic interaction chromatography confirmed that although lysozyme had greater positive charge than Cyt c or RNase A under the conditions used for iontophoresis, it also possessed the highest surface hydrophobicity, which may have facilitated the interactions with the transport pathways and encouraged aggregation in the skin microenvironment. Thus, high charge and electrophoretic mobility seem to be inadequate descriptors to predict the transdermal iontophoretic transport of proteins whose complex three dimensional structures can facilitate interactions with cutaneous transport pathways.


Subject(s)
Iontophoresis , Muramidase/administration & dosage , Muramidase/pharmacokinetics , Skin Absorption/physiology , Skin/drug effects , Acetaminophen/administration & dosage , Acetaminophen/pharmacokinetics , Animals , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Cytochromes c/administration & dosage , Cytochromes c/pharmacokinetics , Electroosmosis , Feasibility Studies , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , In Vitro Techniques , Iontophoresis/methods , Lidocaine/administration & dosage , Lidocaine/pharmacokinetics , Ribonuclease, Pancreatic/administration & dosage , Ribonuclease, Pancreatic/pharmacokinetics , Skin/enzymology , Skin/metabolism , Species Specificity , Static Electricity , Sus scrofa
7.
Nanoscale ; 5(3): 1009-17, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23249999

ABSTRACT

Ultra-small gold nanoclusters (AuNCs) have unique size-dependent optical, electrical and chemical properties. They have emerged as a new nanomaterial with broad applications in optoelectronics, catalysis, biosensing, and bioimaging. Several strategies have been exploited to prepare AuNCs of different "magic number" sizes, using different templates e.g. dendrimers, polyethyleneimines, peptides, and more recently, proteins. Notwithstanding, almost all bio-template-protected AuNCs reported so far exhibit fairly low fluorescence quantum yields (QYs), typically <5%, which is especially true for AuNCs prepared using the protein templates. In this paper, we report a facile, one-pot aqueous synthesis of highly fluorescent AuNCs using bovine pancreatic ribonuclease A (RNase-A) as the bio-template. The as-prepared AuNCs not only fluoresce strongly at the near-infrared (NIR) region (λ(em) = 682 nm), but also exhibit an elevated QY of ∼12%. Additionally, the RNase-A-encapsulated AuNC (RNase-A-AuNC) displays an exceptionally large Stokes shift of ∼210 nm as well as a single dominant fluorescence lifetime of ∼1.5 µs, about three orders of magnitude longer than biological autofluorescence. Furthermore, by coupling vitamin B(12) (VB(12)) to the RNase-A-AuNC, we develop a multifunctional nanoplatform that is suitable for simultaneous targeting and imaging of cancer at the cellular level using Caco-2 cell lines as an in vitro model. Since VB(12) has effective uptake pathways in the digestive system, this nanoplatform may have potential for targeted oral drug delivery in vivo.


Subject(s)
Gold/therapeutic use , Metal Nanoparticles/therapeutic use , Microscopy, Fluorescence/methods , Neoplasms, Experimental/drug therapy , Ribonuclease, Pancreatic/pharmacokinetics , Caco-2 Cells , Coated Materials, Biocompatible/chemical synthesis , Coated Materials, Biocompatible/pharmacokinetics , Gold/chemistry , Humans , Materials Testing , Neoplasms, Experimental/pathology
8.
Biochemistry ; 49(49): 10535-42, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21062079

ABSTRACT

In the redox-coupled oxidative folding of a protein having several SS bonds, two folding phases are usually observed, corresponding to SS formation (oxidation) with generation of weakly stabilized heterogeneous structures (a chain-entropy losing phase) and the subsequent intramolecular SS rearrangement to search for the native SS linkages (a conformational folding phase). By taking advantage of DHS(ox) as a highly strong and selective oxidant, the former SS formation phase was investigated in detail in the oxidative folding of RNase A. The folding intermediates obtained at 25 °C and pH 4.0 within 1 min (1S°-4S°) showed different profiles in the HPLC chromatograms from those of the intermediates obtained at pH 7.0 and 10.0 (1S-4S). However, upon prolonged incubation at pH 4.0 the profiles of 1S°-3S° transformed slowly to those similar to 1S-3S intermediate ensembles via intramolecular SS reshuffling, accompanying significant changes in the UV and fluorescence spectra but not in the CD spectrum. Similar conversion of the intermediates was observed by pH jump from 4.0 to 8.0, while the opposite conversion from 1S-4S was observed by addition of guanidine hydrochloride to the folding solution at pH 8.0. The results demonstrated that the preconformational folding phase coupled with SS formation can be divided into two distinct subphases, a kinetic (or stochastic) SS formation phase and a thermodynamic SS reshuffling phase. The transition from kinetically formed to thermodynamically stabilized SS intermediates would be induced by hydrophobic nucleation as well as generation of the native interactions.


Subject(s)
Disulfides/chemistry , Protein Folding , Ribonuclease, Pancreatic/chemistry , Ribonuclease, Pancreatic/pharmacokinetics , Thermodynamics , Animals , Cattle , Disulfides/metabolism , Disulfides/pharmacokinetics , Entropy , Kinetics , Oxidation-Reduction , Ribonuclease, Pancreatic/metabolism
9.
J Control Release ; 145(3): 203-9, 2010 Aug 03.
Article in English | MEDLINE | ID: mdl-20423719

ABSTRACT

The purpose of the study was to demonstrate the feasibility of using transdermal iontophoresis to deliver a functional protein, ribonuclease A (RNAse; 13.6 kDa), non-invasively across the skin. Iontophoretic transport experiments were conducted using porcine skin in vitro and established the effect of current density and protein concentration on delivery kinetics. A methylene blue-based assay was used to quantify RNAse transport and to simultaneously demonstrate that protein functionality was retained post-iontophoresis. The results confirmed that intact functional RNAse was indeed delivered across the skin; cumulative permeation and steady state flux after 8h iontophoresis at 0.3 mA/cm(2) were 224.37+/-72.34 microg/cm(2) and 68.28+/-23.87 microg/cm(2)h, respectively. Significant amounts of protein were also deposited within the membrane (e.g., 1425.13+/-312.09 microg/cm(2) at 0.3 mA/cm(2)). In addition to the evidence provided by the enzymatic assay with regards to RNAse integrity and functionality, SDS-PAGE gels and MALDI-TOF spectra were also used to characterize RNAse present in the receiver phase (MALDI-TOF spectra: RNAse control, 13.690 kDa cf. RNAse from permeation samples, 13.692 kDa). Co-iontophoresis of acetaminophen showed that, despite its molecular weight, electromigration was the predominant electrotransport mechanism, accounting for >80% of RNAse total flux. Increasing RNAse concentration from 0.35 to 0.7 mM in the formulation did not result in a statistically significant increase in delivery. Iontophoretic transport of RNAse across human skin was statistically equivalent to that seen with porcine skin under the same conditions; cumulative permeation across human and porcine skin was 241.48+/-60.01 and 170.71+/-92.13 microg/cm(2), respectively. Laser scanning confocal microscopy was used to visualize the distribution of rhodamine B-labelled RNAse in the epidermis and dermis as a function of depth following 8h iontophoresis (results were compared to control experiments involving passive administration of the same formulation for 8h). Although fluorescence was localized at the skin surface following passive administration, it was visible throughout the membrane after current application. In conclusion, the results demonstrate that non-invasive transdermal iontophoresis can be used to deliver significant amounts of a structurally intact, functional protein across skin.


Subject(s)
Iontophoresis/methods , Ribonuclease, Pancreatic/administration & dosage , Ribonuclease, Pancreatic/pharmacokinetics , Skin/metabolism , Administration, Cutaneous , Animals , Humans , Skin Absorption , Swine
10.
Biotechnol Bioeng ; 101(4): 837-42, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18623227

ABSTRACT

Using a quartz crystal microbalance with dissipative monitoring (QCM-D) we have determined the adsorption reversibility and viscoelastic properties of ribonuclease A adsorbed to hydrophobic self-assembled monolayers. Consistent with previous work with proteins unfolding on hydrophobic surfaces, high protein solution concentrations, reduced adsorption times, and low ammonium sulfate concentrations lead to increased adsorption reversibility. Measured rigidity of the protein layers normalized for adsorbed protein amounts, a quantity we term specific dissipation, correlated with adsorption reversibility of ribonuclease A. These results suggest that specific dissipation may be correlated with changes in structure of adsorbed proteins.


Subject(s)
Hydrophobic and Hydrophilic Interactions , Ribonuclease, Pancreatic/chemistry , Ribonuclease, Pancreatic/pharmacokinetics , Adsorption
11.
Nano Lett ; 7(7): 1991-5, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17559285

ABSTRACT

This paper reports on the unfolding behavior of ribonuclease A (RNase A) on silica nanoparticle surfaces and quantitively demonstrates that nanoscale size and surface curvature play key roles in influencing the stability of adsorbed proteins. Urea denaturation analyses showed that the thermodynamic stability of RNase A decreased upon adsorption onto the nanoparticles, with greater decrease on larger nanoparticles. The stability changes of RNase A correlate well with the changes in the protein-nanoparticle interactions, which increase as the surface contact area and surface charge interaction increases. This study, therefore, provides fundamental information on the effect of nanoscale surfaces on protein structure and function.


Subject(s)
Nanoparticles , Protein Folding , Ribonuclease, Pancreatic/chemistry , Silicon Dioxide , Adsorption , Animals , Cattle , Enzyme Stability , Ribonuclease, Pancreatic/pharmacokinetics
12.
Anticancer Res ; 26(2A): 857-63, 2006.
Article in English | MEDLINE | ID: mdl-16619480

ABSTRACT

BACKGROUND: We previously showed the usefulness of a fused protein of human pancreatic ribonuclease1 (hRNase1) with human epidermal growth factor (hEGF) for molecular targeting of EGF receptor (EGFR)-overexpressing cells. In this study, the mechanisms underlying the inhibition of cell growth by RNase-EGF fused proteins was confirmed. MATERIALS AND METHODS: Des.1-7 hRNase1 was genetically fused to hEGF. The fused proteins were expressed and isolated from Escherichia coli. The internalization of hRNase1-hEGF was confirmed by confocal fluorescence microscopy. The growth-inhibitory effect of the fused proteins was evaluated by MTT assay. RESULTS: FITC-labelled hRNase1-hEGF was internalized into EGFR-overexpressing A431 cells. The internalization was not observed in A431 cells pre-treated with hEGF and EGFR-deficient H69 cells. The growth-inhibitory effect of des.1-7 hRNase1-hEGF against A431 cells was statistically significantly more pronounced than that of hRNase1-hEGF. CONCLUSION: RNase-EGF fused proteins are internalized through EGFR and inhibit cell growth by exerting their ribonucleolytic activity in the cytosol.


Subject(s)
Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Recombinant Fusion Proteins/pharmacology , Ribonuclease, Pancreatic/pharmacology , Amino Acid Sequence , Carcinoma, Small Cell/drug therapy , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacokinetics , ErbB Receptors/biosynthesis , ErbB Receptors/deficiency , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Fluorescein-5-isothiocyanate/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Microscopy, Fluorescence , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/metabolism , Ribonuclease, Pancreatic/pharmacokinetics , Vulvar Neoplasms/drug therapy , Vulvar Neoplasms/metabolism , Vulvar Neoplasms/pathology
13.
Bull Exp Biol Med ; 142(4): 464-6, 2006 Oct.
Article in English, Russian | MEDLINE | ID: mdl-17415438

ABSTRACT

Dynamics of angiogenin content in the serum, thymus, bone marrow, and brain of mice was studied after intravenous injection. The maximum angiogenin retention was detected in the thymus, while high rate of its elimination in the brain.


Subject(s)
Brain/metabolism , Immunocompetence/physiology , Milk/physiology , Ribonuclease, Pancreatic/pharmacokinetics , Animals , Bone Marrow/metabolism , Male , Mice , Mice, Inbred BALB C , Thymus Gland/metabolism , Tissue Distribution
14.
J Nucl Med ; 45(8): 1373-80, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15299064

ABSTRACT

UNLABELLED: Direct radiolabeling of proteins can result in the loss of targeting activity, requires highly customized procedures, and yields heterogeneous products. Here we describe a novel imaging complex comprised of a standardized (99m)Tc-radiolabeled adapter protein noncovalently bound to a "Docking tag" fused to a "Targeting protein". The assembly of this complex is based on interactions between human 109-amino acid (HuS) and 15-amino acid (Hu-tag) fragments of ribonuclease I, which serve as an "Adapter protein" and a Docking tag, respectively. METHODS: HuS modified with hydrazinonicotinamide (HYNIC) was radiolabeled using (99m)Tc-tricine to a specific activity of 3.4-7.4 MBq/microg. Protein complexes were then formed by mixing (99m)Tc-HuS with equimolar amounts of either Hu-tagged VEGF(121) (Hu-VEGF [vascular endothelial growth factor]) or Hu-tagged anti-VEGFR-2 single-chain antibody (Hu-P4G7) and incubating on ice for 15 min. 4T1 luc/gfp luciferase-expressing murine mammary adenocarcinoma cells (1 x 10(4)) were implanted subcutaneously or injected intravenously into BALB/c mice. Bioluminescent imaging (BLI) was performed 10 d later. Immediately after BLI visualization of tumor, 18.5-37 MBq of tracer (5-10 microg of protein) were injected via tail vein. One hour later planar or SPECT images were obtained, followed by killing the mice. RESULTS: There was significantly (P = 0.0128) increased uptake of (99m)Tc-HuS/Hu-VEGF (n = 10) within subcutaneous tumor as compared with (99m)Tc-HuS/Hu-P4G7 (n = 5) at biodistribution assay (2.68 +/- 0.75 vs. 1.8 +/- 0.21; tumor-to-subcutaneous tissue [ratio of specific activities], respectively), despite similar molecular weights. The focal (99m)Tc-HuS/Hu-VEGF uptake seen on planar images (3.44 +/- 1.16 [tumor to soft-tissue background]) corresponded directly to the locations of tumor observed by BLI. Region of interest analyses of SPECT images revealed a significant increase of (99m)Tc-HuS/Hu-VEGF (n = 5) within the lungs with BLI-detectable pulmonary tumor nodules as compared with controls (n = 4) (right: 4.47 +/- 2.07 vs. 1.79 +/- 0.56; left: 3.66 +/- 1.65 vs. 1.62 +/- 0.45, tumor lung [counts/pixel]/normal lung [counts/pixel], respectively). CONCLUSION: (99m)Tc-HuS/Hu-VEGF complex is stable for at least 1 h in vivo and can be effectively used to image mouse tumor neovasculature in lesions as small as several millimeters in soft tissue. We expect that a similar approach can be adapted for in vivo delivery of other targeting proteins of interest without affecting their bioactivity.


Subject(s)
Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/metabolism , Ribonuclease, Pancreatic/pharmacokinetics , Vascular Endothelial Growth Factor A/pharmacokinetics , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/metabolism , Animals , Humans , Isotope Labeling/methods , Male , Metabolic Clearance Rate , Mice , Mice, Inbred BALB C , Organ Specificity , Radionuclide Imaging , Radiopharmaceuticals/blood , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/pharmacokinetics , Reproducibility of Results , Ribonuclease, Pancreatic/blood , Ribonuclease, Pancreatic/genetics , Sensitivity and Specificity , Technetium/blood , Technetium/pharmacokinetics , Tissue Distribution , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/genetics
15.
Biomed Khim ; 49(6): 588-96, 2003.
Article in Russian | MEDLINE | ID: mdl-16119087

ABSTRACT

Conjugates of pancreatic RNase and ligand-free human serum albumin (LFHSA) have been obtained. The number of hydrophobic binding sites both for initial HSA and LFHSA has been determined by the polarised luminescence method. Interaction between RNase and HSA involves additional electrovalent linkage. Unlike initial enzyme, conjugates exhibit activity toward double-strand RNA. After intravenous injection, transferase activity of unmodified enzyme remains in the blood during 20 min., whereas 30-40% of this activity is detected at the fourth day after administration of RNase conjugates. A single dose administration of LFHSA-RNAse conjugates exhibited high antiviral activity in mice, infected with influenza A and influenza B viruses.


Subject(s)
Ribonuclease, Pancreatic/chemistry , Serum Albumin/chemistry , Animals , Binding Sites , Humans , Ligands , Metabolic Clearance Rate , Rabbits , Rats , Rats, Wistar , Ribonuclease, Pancreatic/metabolism , Ribonuclease, Pancreatic/pharmacokinetics , Serum Albumin/metabolism , Serum Albumin/pharmacokinetics , Tissue Distribution
16.
Biomol Eng ; 19(2-6): 273-80, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12202195

ABSTRACT

Surface modification by deposition of ordered protein systems constitutes one of the major objectives of bio-related chemistry and biotechnology. In this respect a concept has recently been reported aimed at fabricating multilayers by the consecutive adsorption of positively and negatively charged polyelectrolytes. We investigate the adsorption processes between polyelectrolyte multilayers and a series of positively and negatively charged proteins. The film buildup and adsorption experiments were followed by Scanning Angle Reflectometry (SAR). We find that proteins strongly interact with the polyelectrolyte film whatever the sign of the charge of both the multilayer and the protein. When charges of the multilayer and the protein are similar, one usually observes the formation of protein monolayers, which can become dense. We also show that when the protein and the multilayer become oppositely charged, the adsorbed amounts are usually larger and the formation of thick protein layers extending up to several times the largest dimension of the protein can be observed. Our results confirm that electrostatic interactions dominate protein/polyelectrolyte multilayer interactions.


Subject(s)
Coated Materials, Biocompatible/chemistry , Materials Testing/methods , Membranes, Artificial , Polyamines/chemistry , Polyethyleneimine/chemistry , Polystyrenes/chemistry , Proteins/pharmacokinetics , Adsorption , Electrochemistry/methods , Humans , Ion Exchange Resins/chemistry , Lactalbumin/pharmacokinetics , Muramidase/pharmacokinetics , Myoglobin/pharmacokinetics , Ribonuclease, Pancreatic/pharmacokinetics , Serum Albumin/pharmacokinetics
17.
Izv Akad Nauk Ser Biol ; (2): 205-8, 2002.
Article in Russian | MEDLINE | ID: mdl-11963547

ABSTRACT

The cow milk angiogenin consumed by mice perorally penetrated from the gastrointestinal tract in the blood flow. In the experimental animals, the blood level of exogenous angiogenin first increased to reach a maximum, and then gradually decreased to zero. The dynamics of this process depends on the age of animals. The data obtained suggest that the cycle of perorally introduced cow milk angiogenin in blood is realized in the infantile-juvenile mice at a higher rate than in the adult-presenile mice.


Subject(s)
Ribonuclease, Pancreatic/blood , Ribonuclease, Pancreatic/pharmacokinetics , Administration, Oral , Age Factors , Animals , Cattle , Female , Intestinal Absorption , Male , Mice , Mice, Inbred BALB C , Ribonuclease, Pancreatic/administration & dosage
18.
Cytokine ; 12(6): 786-90, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10843765

ABSTRACT

A hybrid human protein was produced in E. coli by fusing the genes encoding human pancreatic RNase1 (hpRNase1) and human IL-2 (hIL-2). The recombinant hpRNase1-hIL-2 inhibited protein synthesis in HTLV-1-infected, malignant T cells, which hyperproduce high affinity IL-2 receptors, with an IC(50)of 2x10(-8) M, whereas no inhibition was detectable in control cells with lower affinity receptors. HpRNase1 alone had an IC(50)of almost 10(-3) M. A molar excess of hIL-2 blocked the protein synthesis inhibition dose-dependently. In a human mixed lymphocyte culture, hpRNase1-hIL-2 inhibited the proliferation of responder cells with potency comparable to that of cyclosporine, while non-effective doses of FK506 importantly improved its potency. Despite its short half-life in animals, hpRNase1-hIL-2 rapidly enters cells in a few minutes and arrests the protein translation in less than 10 h. Thus, hpRNase1-hIL-2 may be useful to selectively eliminate activated lymphocytes hyperproducing high affinity IL-2 receptors, as in allograft rejection, graft-versus-host disease, autoimmune disorders, adult T cell leukaemia and other lymphoproliferative or retroviral malignancies including HIV infection, without inducing general immunosuppression. As an entirely human "immunotoxin analogue" it may alleviate the dose limiting toxicity and immunogenicity of conventional immunotoxins.


Subject(s)
Human T-lymphotropic virus 1/physiology , Interleukin-2/toxicity , Lymphocyte Activation/physiology , Ribonuclease, Pancreatic/toxicity , T-Lymphocytes/drug effects , Animals , Cell Division/drug effects , Cloning, Molecular , Escherichia coli , Humans , Interleukin-2/pharmacokinetics , Interleukin-2/pharmacology , Leukemia, T-Cell , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed , Lymphoma, T-Cell , Metabolic Clearance Rate , Rats , Rats, Inbred Lew , Receptors, Interleukin-2/physiology , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/toxicity , Ribonuclease, Pancreatic/pharmacokinetics , Ribonuclease, Pancreatic/pharmacology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Tumor Cells, Cultured
19.
J Control Release ; 56(1-3): 231-7, 1998 Dec 04.
Article in English | MEDLINE | ID: mdl-9801446

ABSTRACT

A major challenge in correcting disorders affecting the central nervous system is to induce blood-brain barrier (BBB) crossing of exogenous biological compounds such as proteins or specific nucleic acid sequences. Fatty acids, due to their high membrane affinity and low toxicity, are good potential candidates to promote this barrier crossing when covalently bound to proteins. In this paper, we report that regiospecific monoacylation of ribonuclease A (RNase A) enables its transport across an in vitro model of the BBB. Myristoylated, palmitoylated and stearoylated RNases A were prepared using reversed micelles as microreactors. All the purified acylated RNases A kept their original enzymatic activity. A single fatty acid moiety was linked to RNase A through the alpha-amino group of its N-terminal lysine as shown by powerful analytical techniques. The ability of monoacylated RNases A to cross an in vitro model of the BBB is strictly dependent on the acyl chain length, which must be at least 16 carbon atoms long.


Subject(s)
Blood-Brain Barrier/physiology , Ribonuclease, Pancreatic/pharmacokinetics , Acetylation , Animals , Biological Transport , Cattle , Chromatography, High Pressure Liquid/methods , Kinetics , Mass Spectrometry/methods , Micelles , Peptide Mapping , Ribonuclease, Pancreatic/metabolism
20.
Cancer Res ; 56(18): 4180-6, 1996 Sep 15.
Article in English | MEDLINE | ID: mdl-8797589

ABSTRACT

The similarities and differences among members of the RNase A superfamily provide an ideal opportunity to examine the molecular basis for differences in their pharmacokinetics and biodistribution. Plasma clearances in BALB/c mice are similar among the five RNases studied: human pancreatic RNase, angiogenin, eosinophil-derived neurotoxin, onconase, and bovine seminal RNase. The average clearance is 0.13 ml/min or 60% of the glomerular filtration rate (measured by [14C]inulin clearance during continuous infusion from an i.p. implanted osmotic pump). Angiogenin has a higher volume of distribution and plasma-to-muscle transport rate than the other RNases, suggestive of binding to endothelial cells. Organ distribution differs dramatically among these RNases. The RNase most toxic to tumor cells, onconase, exhibits the longest retention in the kidneys: at 180 min, 50% of the injected dose is found in the kidneys, whereas only 1% or less of the other RNases is retained in the kidneys. Slower elimination of onconase from the kidneys may be due to a higher degree of binding in the kidney or a resistance to proteolytic degradation. To elucidate the molecular determinants involved in tissue uptake, we examined the biodistribution of recombinant onconase and two onconasepancreatic RNase chimeric proteins. The tissue retention property of onconase appears to be located in at least two regions, one of which is in the NH2-terminal 9-amino acid alpha-helix. The NH2-terminal pyroglutamate of onconase, a residue essential for ribonucleolytic activity and cytotoxicity, does not play a role in kidney retention.


Subject(s)
Ribonuclease, Pancreatic/pharmacokinetics , Ribonucleases/pharmacokinetics , Animals , Cattle , Female , Glomerular Filtration Rate , Humans , Infusions, Parenteral , Inulin/pharmacokinetics , Male , Metabolic Clearance Rate , Mice , Mice, Inbred BALB C , Ovum/enzymology , Proteins/pharmacokinetics , Rana pipiens , Ribonuclease, Pancreatic/blood , Ribonuclease, Pancreatic/isolation & purification , Ribonucleases/blood , Ribonucleases/isolation & purification , Semen/enzymology , Seminal Vesicles/enzymology , Time Factors , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...