Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 219
Filter
1.
Biomed Pharmacother ; 174: 116569, 2024 May.
Article in English | MEDLINE | ID: mdl-38603886

ABSTRACT

Alpha-alpha diaspirin-crosslinked human hemoglobin (DCLHb or ααHb) was a promising early generation red blood cell (RBC) substitute. The DCLHb was developed through a collaborative effort between the United States Army and Baxter Healthcare. The core design feature underlying its development was chemical stabilization of the tetrameric structure of hemoglobin (Hb) to prevent Hb intravascular dimerization and extravasation. DCLHb was developed to resuscitate warfighters on the battlefield, who suffered from life-threatening blood loss. However, extensive research revealed toxic side effects associated with the use of DCLHb that contributed to high mortality rates in clinical trials. This study explores whether scavenging Hb and heme via the apohemoglobin-haptoglobin (apoHb-Hp) complex can reduce DCLHb associated toxicity. Awake Golden Syrian hamsters were equipped with a window chamber model to characterize the microcirculation. Each group was first infused with either Lactated Ringer's or apoHb-Hp followed by a hypovolemic infusion of 10% of the animal's blood volume of DCLHb. Our results indicated that animals pretreated with apoHb-Hb exhibited improved microhemodynamics vs the group pretreated with Lactated Ringer's. While systemic acute inflammation was observed regardless of the treatment group, apoHb-Hp pretreatment lessened those effects with a marked reduction in IL-6 levels in the heart and kidneys compared to the control group. Taken together, this study demonstrated that utilizing a Hb and heme scavenger protein complex significantly reduces the microvasculature effects of ααHb, paving the way for improved HBOC formulations. Future apoHb-Hp dose optimization studies may identify a dose that can completely neutralize DCLHb toxicity.


Subject(s)
Haptoglobins , Hemoglobins , Animals , Hemoglobins/pharmacology , Hemoglobins/metabolism , Humans , Haptoglobins/metabolism , Male , Mesocricetus , Apoproteins/chemistry , Apoproteins/pharmacology , Blood Substitutes/pharmacology , Blood Substitutes/chemistry , Cross-Linking Reagents/chemistry , Cricetinae
2.
J Antimicrob Chemother ; 74(9): 2631-2639, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31170282

ABSTRACT

BACKGROUND: New strategies are needed to slow the emergence of antibiotic resistance among bacterial pathogens. In particular, society is experiencing a crisis of antibiotic-resistant infections caused by Gram-negative bacterial pathogens and novel therapeutics are desperately needed to combat such diseases. Acquisition of iron from the host is a nearly universal requirement for microbial pathogens-including Gram-negative bacteria-to cause infection. We have previously reported that apo-transferrin (lacking iron) can inhibit the growth of Staphylococcus aureus in culture and diminish emergence of resistance to rifampicin. OBJECTIVES: To define the potential of apo-transferrin to inhibit in vitro growth of Klebsiella pneumoniae and Acinetobacter baumannii, key Gram-negative pathogens, and to reduce emergence of resistance to antibiotics. METHODS: The efficacy of apo-transferrin alone or in combination with meropenem or ciprofloxacin against K. pneumoniae and A. baumannii clinical isolates was tested by MIC assay, time-kill assay and assays for the selection of resistant mutants. RESULTS: We confirmed that apo-transferrin had detectable MICs for all strains tested of both pathogens. Apo-transferrin mediated an additive antimicrobial effect for both antibiotics against multiple strains in time-kill assays. Finally, adding apo-transferrin to ciprofloxacin or meropenem reduced the emergence of resistant mutants during 20 day serial passaging of both species. CONCLUSIONS: These results suggest that apo-transferrin may have promise to suppress the emergence of antibiotic-resistant mutants when treating infections caused by Gram-negative bacteria.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Apoproteins/pharmacology , Drug Resistance, Bacterial/drug effects , Gram-Negative Bacteria/drug effects , Gram-Negative Bacterial Infections/drug therapy , Transferrin/pharmacology , Acinetobacter baumannii/drug effects , Ciprofloxacin/therapeutic use , Gram-Negative Bacterial Infections/microbiology , Humans , Klebsiella pneumoniae/drug effects , Meropenem/therapeutic use , Microbial Sensitivity Tests , Mutation
3.
Sci Rep ; 9(1): 1780, 2019 02 11.
Article in English | MEDLINE | ID: mdl-30741951

ABSTRACT

Cancer remains one of the biggest threats to human society. There are massive demands for compounds to selectively kill cancerous cells. Earlier studies have shown that bovine α -lactalbumin made lethal to tumor cells (BAMLET) becomes cytotoxic against cancer cells in complex with oleic acid {Hoque, M. et. al., PLoSOne 8, e68390 (2013)}. In our study, we obtained bovine α-lactalbumin complexed with lanthanum ion (La3+-B-α-LA) and determined its high resolution crystal structure. The natural calcium binding site of bovine α-lactalbumin is replaced by lanthanum. The La3+ complex formation by B-α-apo-LA was also supported by various biophysical methods. Interestingly, our complex, La3+-B-α-LA exhibits much greater anticancer activity against breast cancer cells as compared to the reported BAMLET-oleic acid complex. This study shows that La3+-B-α-LA complex is preferentially more toxic to MCF-7 cells as compared to KB (oral cancer) and HeLa (cervical) cells, while almost non-toxic to the healthy cells that we studied. Our data indicates that the cytotoxicity of La3+-B-α-LA against cancer cells is through apoptotic path way. The higher anticancer activity of La3+-B-α-LA is attributable to the requisite structural changes induced in the protein by La3+ binding as supported by the crystal structure of the complex.


Subject(s)
Apoproteins/pharmacology , Lactalbumin/pharmacology , Lanthanum/metabolism , Animals , Apoproteins/chemistry , Apoproteins/metabolism , Calcium/metabolism , Cattle , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Humans , Lactalbumin/chemistry , Lactalbumin/metabolism , Lanthanum/chemistry , Molecular Structure , Protein Binding
4.
Article in English | MEDLINE | ID: mdl-30782989

ABSTRACT

There has been renewed interest in combining traditional small-molecule antimicrobial agents with nontraditional therapies to potentiate antimicrobial effects. Apotransferrin, which decreases iron availability to microbes, is one such approach. We conducted a 48-h one-compartment in vitro infection model to explore the impact of apotransferrin on the bactericidal activity of ciprofloxacin. The challenge panel included four Klebsiella pneumoniae isolates with ciprofloxacin MIC values ranging from 0.08 to 32 mg/liter. Each challenge isolate was subjected to an ineffective ciprofloxacin monotherapy exposure (free-drug area under the concentration-time curve over 24 h divided by the MIC [AUC/MIC ratio] ranging from 0.19 to 96.6) with and without apotransferrin. As expected, the no-treatment and apotransferrin control arms showed unaltered prototypical logarithmic bacterial growth. We identified relationships between exposure and change in bacterial density for ciprofloxacin alone (R2 = 0.64) and ciprofloxacin in combination with apotransferrin (R2 = 0.84). Addition of apotransferrin to ciprofloxacin enabled a remarkable reduction in bacterial density across a wide range of ciprofloxacin exposures. For instance, at a ciprofloxacin AUC/MIC ratio of 20, ciprofloxacin monotherapy resulted in nearly 2 log10 CFU increase in bacterial density, while the combination of apotransferrin and ciprofloxacin resulted in 2 log10 CFU reduction in bacterial density. Furthermore, addition of apotransferrin significantly reduced the emergence of ciprofloxacin-resistant subpopulations compared to monotherapy. These data demonstrate that decreasing the rate of bacterial replication with apotransferrin in combination with antimicrobial therapy represents an opportunity to increase the magnitude of the bactericidal effect and to suppress the growth rate of drug-resistant subpopulations.


Subject(s)
Anti-Bacterial Agents/pharmacology , Apoproteins/pharmacology , Ciprofloxacin/pharmacology , Transferrin/pharmacology , Fluoroquinolones/pharmacology , Klebsiella/drug effects , Microbial Sensitivity Tests
5.
Int J Pharm ; 514(2): 399-406, 2016 Dec 05.
Article in English | MEDLINE | ID: mdl-27628783

ABSTRACT

The airways of most people with cystic fibrosis are colonized with biofilms of the Gram-negative, opportunistic pathogen Pseudomonas aeruginosa. Delivery of antibiotics directly to the lung in the form of dry powder aerosols offers the potential to achieve high local concentrations directly to the biofilms. Unfortunately, current aerosolised antibiotic regimes are unable to efficiently eradicate these biofilms from the airways. We investigated the ability of the innate antimicrobial, lactoferrin, to enhance the activity of two aminoglycoside antibiotics (tobramycin and gentamicin) against biofilms of P. aeruginosa strain PAO1. Biofilms were prepared in 96 well polystyrene plates. Combinations of the antibiotics and various lactoferrin preparations were spray dried. The bacterial cell viability of the various spray dried combinations was determined. Iron-free lactoferrin (apo lactoferrin) induced a 3-log reduction in the killing of planktonic cell by the aminoglycoside antibiotics (p<0.01) and also reduced both the formation and persistence of P. aeruginosa biofilms (p<0.01). Combinations of lactoferrin and an aminoglycoside displays potential as an effective new therapeutic strategy in the treatment of P. aeruginosa biofilms infections such as those typical of the CF lungs.


Subject(s)
Anti-Infective Agents/administration & dosage , Anti-Infective Agents/pharmacology , Gentamicins/pharmacology , Lactoferrin/administration & dosage , Lactoferrin/pharmacology , Tobramycin/pharmacology , Anti-Infective Agents/chemistry , Apoproteins/administration & dosage , Apoproteins/chemistry , Apoproteins/pharmacology , Biofilms/drug effects , Cell Survival/drug effects , Drug Compounding , Drug Synergism , Lactoferrin/chemistry , Powders , Pseudomonas aeruginosa/drug effects
6.
Adv Mind Body Med ; 30(1): 4-11, 2016.
Article in English | MEDLINE | ID: mdl-26878676

ABSTRACT

CONTEXT: The changes in verbal learning and working memory that often occur with aging may result in reduced social and intellectual interactions. These changes significantly affect an individual's quality of life. As humans age, the body's ability to regulate and maintain calcium levels is diminished. Pharmacological manipulation of the entry of free calcium (Ca2+) has been shown to be effective in increasing some aspects of cognitive function in the aged brain. Apoaequorin has been shown in laboratory studies to regulate levels of intracellular calcium in neuronal cells and to provide protection against ischemic cell death. OBJECTIVE: The study was designed to assess the effects of a supplement of apoaequorin on verbal learning and working memory. DESIGN: The current study, the Madison Memory Study, was a randomized, double-blind, placebo-controlled trial. SETTING: The study occurred in Madison, WI, USA. PARTICIPANTS: Participants were 218 community-dwelling adults, aged 40-91 y, with self-reported memory concerns. INTERVENTION: Participants were randomly assigned to receive either apoaequorin (apoaequorin group) or a matched placebo (control group) for 90 d. OUTCOME MEASURES: The study used quantitative, computerized tools for cognitive assessment the CogState International Shopping List (ISL) and the CogState ISL-Delayed Recall (ISL-DR). Scores from computerized cognitive tasks were measured at baseline and at several points during the 90-d study. RESULTS: No significant differences existed between the intervention and control groups in any parameter at baseline. The intervention group (apoaequorin group) showed a statistically significant improvement in verbal learning and recall on the ISL and the ISL-DR, respectively, during the 90-d study. Apoaequorin was tolerated very well in the study. CONCLUSIONS: The results indicated a strong relationship between apoaequorin and improvements on a quantitative measure of cognitive function, specifically verbal learning. The study found that apoaequorin is a well-tolerated supplement that improved cognitive function in aging adults. The results suggest potential utility for apoaequorin in addressing the declines in cognitive function associated with aging.


Subject(s)
Aequorin/administration & dosage , Aequorin/pharmacology , Apoproteins/administration & dosage , Apoproteins/pharmacology , Dietary Supplements , Verbal Learning/drug effects , Adult , Aequorin/adverse effects , Aged , Aged, 80 and over , Aging , Apoproteins/adverse effects , Cognition/drug effects , Female , Humans , Male , Middle Aged , Neuropsychological Tests , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacology
7.
Int J Nanomedicine ; 11: 25-44, 2016.
Article in English | MEDLINE | ID: mdl-26730188

ABSTRACT

This study used the Eri silk nanoparticles (NPs) for delivering apo-bovine lactoferrin (Apo-bLf) (~2% iron saturated) and Fe-bLf (100% iron saturated) in MDA-MB-231 and MCF-7 breast cancer cell lines. Apo-bLf and Fe-bLf-loaded Eri silk NPs with sizes between 200 and 300 nm (±10 nm) showed a significant internalization within 4 hours in MDA-MB-231 cells when compared to MCF-7 cells. The ex vivo loop assay with chitosan-coated Fe-bLf-loaded silk NPs was able to substantiate its future use in oral administration and showed the maximum absorption within 24 hours by ileum. Both Apo-bLf and Fe-bLf induced increase in expression of low-density lipoprotein receptor-related protein 1 and lactoferrin receptor in epidermal growth factor (EGFR)-positive MDA-MB-231 cells, while transferrin receptor (TfR) and TfR2 in MCF-7 cells facilitated the receptor-mediated endocytosis of NPs. Controlled and sustained release of both bLf from silk NPs was shown to induce more cancer-specific cytotoxicity in MDA-MB-231 and MCF-7 cells compared to normal MCF-10A cells. Due to higher degree of internalization, the extent of cytotoxicity and apoptosis was significantly higher in MDA-MB-231 (EGFR+) cells when compared to MCF-7 (EGFR-) cells. The expression of a prominent anticancer target, survivin, was found to be downregulated at both gene and protein levels. Taken together, all the observations suggest the potential use of Eri silk NPs as a delivery vehicle for an anti-cancer milk protein, and indicate bLf for the treatment of breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Drug Delivery Systems/methods , Lactoferrin/pharmacology , Nanoparticles/administration & dosage , Silk/chemistry , Animals , Apoproteins/administration & dosage , Apoproteins/pharmacology , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cattle , Chitosan/chemistry , Chitosan/metabolism , Drug Delivery Systems/instrumentation , Female , Humans , Iron/chemistry , Lactoferrin/administration & dosage , MCF-7 Cells/drug effects , Mice , Moths/chemistry , Nanoparticles/chemistry , Nanoparticles/metabolism , Receptors, Cell Surface/metabolism
8.
Anticancer Drugs ; 27(1): 24-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26457548

ABSTRACT

Neocarzinostatin (NCS) consists of an enediyne chromophore and an apoprotein (NCP). Lidamycin (LDM) is composed of another active enediyne chromophore (AE) and an acidic protein (LDP). Although the structures of NCP and LDP are very similar, LDM has been shown to have an increased tumor-suppressive activity than that of NCS. The aim of this study was to construct a chimeric protein (CMP) that consists of both the terminus residue of NCP and an LDP pocket-forming residue that can bind AE. This CMP will have a structure similar to NCS and an antitumor activity similar to LDM. The assembling efficiency of LDP, CMP, and NCP was 73.9, 1.5, and 1.1%, respectively. The cytotoxicity was consistent with their assembling efficiency of AE in proteins. When CMP-AE and NCP-AE were administered at equivalent AE doses of LDM, the inhibition rate of CMP-AE was the same as LDM and significantly higher than that of NCP-AE. Our study implied that the binding activity between LDP and AE was very specific. The terminus residue of LDP could affect the specifically binding activity. The pocket-forming residue could confer a protective function to the chromophore. Further investigation of its bioactivity might serve as a new drug design strategy and drug-delivery carrier in targeted cancer therapy.


Subject(s)
Aminoglycosides/chemistry , Antineoplastic Agents/chemistry , Apoproteins/chemistry , Enediynes/chemistry , Recombinant Fusion Proteins/chemistry , Zinostatin/chemistry , Aminoglycosides/genetics , Animals , Antineoplastic Agents/pharmacology , Apoproteins/genetics , Apoproteins/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Mice , Neoplasm Transplantation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology
9.
Haematologica ; 101(3): 297-308, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26635037

ABSTRACT

Iron overload results in significant morbidity and mortality in ß-thalassemic patients. Insufficient hepcidin is implicated in parenchymal iron overload in ß-thalassemia and approaches to increase hepcidin have therapeutic potential. We have previously shown that exogenous apo-transferrin markedly ameliorates ineffective erythropoiesis and increases hepcidin expression in Hbb(th1/th1) (thalassemic) mice. We utilize in vivo and in vitro systems to investigate effects of exogenous apo-transferrin on Smad and ERK1/2 signaling, pathways that participate in hepcidin regulation. Our results demonstrate that apo-transferrin increases hepcidin expression in vivo despite decreased circulating and parenchymal iron concentrations and unchanged liver Bmp6 mRNA expression in thalassemic mice. Hepatocytes from apo-transferrin-treated mice demonstrate decreased ERK1/2 pathway and increased serum BMP2 concentration and hepatocyte BMP2 expression. Furthermore, hepatocyte ERK1/2 phosphorylation is enhanced by neutralizing anti-BMP2/4 antibodies and suppressed in vitro in a dose-dependent manner by BMP2, resulting in converse effects on hepcidin expression, and hepatocytes treated with MEK/ERK1/2 inhibitor U0126 in combination with BMP2 exhibit an additive increase in hepcidin expression. Lastly, bone marrow erythroferrone expression is normalized in apo-transferrin treated thalassemic mice but increased in apo-transferrin injected wild-type mice. These findings suggest that increased hepcidin expression after exogenous apo-transferrin is in part independent of erythroferrone and support a model in which apo-transferrin treatment in thalassemic mice increases BMP2 expression in the liver and other organs, decreases hepatocellular ERK1/2 activation, and increases nuclear Smad to increase hepcidin expression in hepatocytes.


Subject(s)
Apoproteins/pharmacology , Bone Morphogenetic Protein 2/genetics , Hepcidins/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Transferrin/pharmacology , beta-Thalassemia/genetics , Animals , Antibodies, Neutralizing/pharmacology , Bone Morphogenetic Protein 2/agonists , Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , Butadienes/pharmacology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepcidins/agonists , Hepcidins/antagonists & inhibitors , Hepcidins/metabolism , Humans , Liver/drug effects , Liver/metabolism , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Nitriles/pharmacology , Phosphorylation/drug effects , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Smad Proteins/genetics , Smad Proteins/metabolism , beta-Thalassemia/metabolism , beta-Thalassemia/pathology
10.
Basic Clin Pharmacol Toxicol ; 117(2): 105-16, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25615234

ABSTRACT

Gelatinases play important roles in tumour invasion and metastasis and are thus considered promising targets for cancer therapy. In this study, a new single-chain variable fragment (scFv)-based fusion protein Fv-LDP, composed of the anti-gelatinases scFv and lidamycin apoprotein (LDP), was prepared, and its combination with angiogenesis inhibitor Endostar was then investigated. The fusion protein Fv-LDP specifically bound to various tumour cells, and its binding capability to human pulmonary giant cell carcinoma (PG) cells was higher than that of LDP. Fv-LDP inhibited the expression and secretion of gelatinases and could be internalized into tumour cells via endocytosis. Fv-LDP also suppressed the growth of human hepatoma cells and murine hepatoma 22 transplanted in Kunming mice in various degrees. In addition, Endostar could enhance the synergistic or additive inhibition of Fv-LDP on the growth, migration or invasion of human hepatoma cells shown by a colony formation assay and a transwell-based migration or invasion assay, respectively. In vivo, Fv-LDP/Endostar combination showed a significantly synergistic effect on the growth of a human hepatoma xenograft, with an inhibition rate of 80.8% compared with the Fv-LDP (44.1%) or Endostar (8.9%)-treated group. The above-mentioned results indicate that the fusion protein Fv-LDP is effective against transplantable hepatoma in mice and human hepatoma xenografts in athymic mice. Moreover, Endostar can potentiate the inhibition effect of Fv-LDP on the growth of human hepatoma cells and xenografts. These data will provide a new combined strategy for improving the therapeutic efficacy of treatments for hepatoma or other gelatinase-overexpressing tumours.


Subject(s)
Aminoglycosides/pharmacology , Carcinoma, Hepatocellular/drug therapy , Endostatins/pharmacology , Enediynes/pharmacology , Liver Neoplasms/drug therapy , Single-Chain Antibodies/pharmacology , Aminoglycosides/administration & dosage , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Animals , Apoproteins/administration & dosage , Apoproteins/pharmacology , Carcinoma, Giant Cell/drug therapy , Carcinoma, Giant Cell/enzymology , Carcinoma, Giant Cell/pathology , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Drug Synergism , Endostatins/administration & dosage , Enediynes/administration & dosage , Female , Gelatinases/metabolism , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Recombinant Proteins , Single-Chain Antibodies/administration & dosage , Xenograft Model Antitumor Assays
11.
Chembiochem ; 16(1): 149-55, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25476866

ABSTRACT

Fibrils formed by human serum transferrin [(1-3 µM) apo-Tf, partially iron-saturated (Fe0.6 -Tf) and holo-Tf (Fe2 -Tf) forms], from dilute bicarbonate solutions, were deposited on formvar surfaces and studied by electron microscopy. We observed that possible bacterial contamination appears to give rise to long, pea-pod-like (PPL) structures for Fe2 -Tf, attributable to the formation of polyhydroxybutyrate (PHB) storage granules, under the nutrient-limiting conditions used. These PPL structures contained periodic nanomineralisation sites susceptible to uranyl stain. Extended incubation of transferrin solutions (about four days) gave rise to extensive transferrin fibril structures. Optical microscopy and AFM studies showed that red blood cells (RBCs) readily adhere to these fibrils. Moreover, the fibrils appear to penetrate RBC membranes and to induce rapid cell destruction (within about 5 h). It is speculated that in situations in vivo where transferrin fibrils can form, such interactions might have adverse physiological consequences, and further studies could aid the understanding of related pathological events.


Subject(s)
Apoproteins/chemistry , Bacteria/metabolism , Erythrocytes/drug effects , Transferrin/chemistry , Apoproteins/pharmacology , Apoproteins/ultrastructure , Bacteria/growth & development , Binding Sites , Cell Adhesion , Erythrocytes/cytology , Hemolysis/drug effects , Humans , Microscopy, Electron, Transmission , Polymerization , Prohibitins , Protein Binding , Protein Conformation , Sodium Bicarbonate , Solutions , Transferrin/pharmacology , Transferrin/ultrastructure
12.
Arch Pharm Res ; 37(2): 270-5, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24155020

ABSTRACT

It has been reported that transferrin has antibacterial and antifungal activities via iron chelation in the environment surrounding the microbes. In the present study, we investigated whether the binding of transferrin to Candida albicans mediates growth inhibition. By using cultures that contained iron-free (apo)transferrin glycoprotein either in contact with candidal cells or separated from candidal cells by a dialysis membrane, we distinguished the growth inhibition by transferrin-cell interaction from that of simple iron chelation. Maximal growth inhibition always occurred when the apotransferrin interacted directly with the cells. Additionally, there was partial inhibition even when candidal cells were in contact with iron-saturated transferrin. Binding studies with (59)Fe(3+) radiolabeled-transferrin indicated that the apo-protein can bind to the candidal cell surface. The binding sites were saturable and it was dose dependent. Chemicals (hydrogen peroxide, dithiothreitol, sodium dodecyl sulfate) blocked transferrin binding to C. albicans, and among the three, hydrogen peroxide (HP) was the most effective for the blocking. When HP-treated yeast cells were added to the culture that was pretreated with apotransferrin, candidal cell growth increased by 5-fold as compared to the growth of HP-untreated candidal cells under apotransferrin-regulation (P < 0.05). Combined all data together, it was concluded that transferrin has a second mechanism of anticandidal activity that is mediated by binding to the surface of C. albicans yeast cells.


Subject(s)
Antifungal Agents/pharmacology , Apoproteins/pharmacology , Candida albicans/drug effects , Iron/metabolism , Transferrin/pharmacology , Binding Sites , Candida albicans/growth & development , Candida albicans/metabolism , Dose-Response Relationship, Drug , Hydrogen Peroxide/pharmacology , Radioligand Assay , Time Factors
13.
PLoS One ; 8(12): e80768, 2013.
Article in English | MEDLINE | ID: mdl-24324625

ABSTRACT

Neurodegenerative diseases are associated with misfolding and deposition of specific proteins, either intra or extracellularly in the nervous system. Advanced glycation end products (AGEs) originate from different molecular species that become glycated after exposure to sugars. Several proteins implicated in neurodegenerative diseases have been found to be glycated in vivo and the extent of glycation is related to the pathologies of the patients. Although it is now accepted that there is a direct correlation between AGEs formation and the development of neurodegenerative diseases, several questions still remain unanswered: whether glycation is the triggering event or just an additional factor acting on the aggregation pathway. To this concern, in the present study we have investigated the effect of glycation on the aggregation pathway of the amyloidogenic W7FW14F apomyoglobin. Although this protein has not been related to any amyloid disease, it represents a good model to resemble proteins that intrinsically evolve toward the formation of amyloid aggregates in physiological conditions. We show that D-ribose, but not D-glucose, rapidly induces the W7FW14F apomyoglobin to generate AGEs in a time-dependent manner and protein ribosylation is likely to involve lysine residues on the polypeptide chain. Ribosylation of the W7FW14F apomyoglobin strongly affects its aggregation kinetics producing amyloid fibrils within few days. Cytotoxicity of the glycated aggregates has also been tested using a cell viability assay. We propose that ribosylation in the W7FW14F apomyoglobin induces the formation of a cross-link that strongly reduces the flexibility of the H helix and/or induce a conformational change that favor fibril formation. These results open new perspectives for AGEs biological role as they can be considered not only a triggering factor in amyloidosis but also a player in later stages of the aggregation process.


Subject(s)
Amyloidogenic Proteins/chemistry , Apoproteins/chemistry , Glycation End Products, Advanced/chemistry , Myoglobin/chemistry , Ribose/chemistry , Amyloidogenic Proteins/genetics , Amyloidogenic Proteins/pharmacology , Animals , Apoproteins/genetics , Apoproteins/pharmacology , Cell Survival/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Flocculation , Gene Expression , Glucose/chemistry , Glycosylation , Humans , Mice , Models, Molecular , Myoglobin/genetics , Myoglobin/pharmacology , NIH 3T3 Cells , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
14.
PLoS One ; 8(11): e79002, 2013.
Article in English | MEDLINE | ID: mdl-24244400

ABSTRACT

Ischemic stroke affects ∼795,000 people each year in the U.S., which results in an estimated annual cost of $73.7 billion. Calcium is pivotal in a variety of neuronal signaling cascades, however, during ischemia, excess calcium influx can trigger excitotoxic cell death. Calcium binding proteins help neurons regulate/buffer intracellular calcium levels during ischemia. Aequorin is a calcium binding protein isolated from the jellyfish Aequorea victoria, and has been used for years as a calcium indicator, but little is known about its neuroprotective properties. The present study used an in vitro rat brain slice preparation to test the hypothesis that an intra-hippocampal infusion of apoaequorin (the calcium binding component of aequorin) protects neurons from ischemic cell death. Bilaterally cannulated rats received an apoaequorin infusion in one hemisphere and vehicle control in the other. Hippocampal slices were then prepared and subjected to 5 minutes of oxygen-glucose deprivation (OGD), and cell death was assayed by trypan blue exclusion. Apoaequorin dose-dependently protected neurons from OGD--doses of 1% and 4% (but not 0.4%) significantly decreased the number of trypan blue-labeled neurons. This effect was also time dependent, lasting up to 48 hours. This time dependent effect was paralleled by changes in cytokine and chemokine expression, indicating that apoaequorin may protect neurons via a neuroimmunomodulatory mechanism. These data support the hypothesis that pretreatment with apoaequorin protects neurons against ischemic cell death, and may be an effective neurotherapeutic.


Subject(s)
Aequorin/pharmacology , Apoproteins/pharmacology , Brain Ischemia/metabolism , CA1 Region, Hippocampal/metabolism , Neurons/metabolism , Animals , Brain Ischemia/drug therapy , Brain Ischemia/pathology , CA1 Region, Hippocampal/pathology , Cell Death/drug effects , Glucose/metabolism , Humans , Neurons/pathology , Oxygen/metabolism , Oxygen Consumption/drug effects , Rats , Recombinant Proteins/pharmacology
15.
J Neurochem ; 127(6): 819-36, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24032544

ABSTRACT

Oligodendroglial damage and demyelination are common pathological features characterizing white matter and neurodegenerative disorders. Identifying the signaling pathways involved in myelin repair through oligodendroglial progenitor maturation is essential for the development of new therapies. This article investigated the role of the Notch signaling pathway in CNS demyelination and apotransferrin-induced remyelination in a focal lysolecithin-induced demyelination model in rats. Notch was found activated in Nestin-expressing neural progenitor cells and in NG2-expressing oligodendroglial precursor cells in the subventricular zone and corpus callosum of lysolecithin-demyelinated rats. Notch activation seemed to be driven by Jagged1, which led to a high expression of downstream gene Hes5 in the subventricular zone of demyelinated rats. Apotransferrin injection induced remyelination, while the injection of the γ-secretase inhibitor reversed this effect. In addition, 24 h after apotransferrin injection, evidence showed Notch activation concomitantly with an increase in F3/contactin levels and the up-regulation of the myelin-associated glycoprotein gene in the subventricular zone and corpus callosum of demyelinated rats. Collected evidence supports the participation of both canonical and non-canonical Notch signaling pathways in demyelination/remyelination. Notch activation was found to trigger Hes5 expression as a consequence of focal demyelination, which might promote oligodendroglial precursor cell proliferation. During apotransferrin-induced remyelination, Notch activation seemed to be mediated by the expression of F3/contactin, which might induce apotransferrin-mediated oligodendroglial maturation. Evidence of the participation of Notch signaling in the demyelination/remyelination process will help further understand demyelinating disorders such as Multiple Sclerosis and the use of aTf should be taken into consideration as a possible therapeutic intervention.


Subject(s)
Apoproteins/pharmacology , Brain/metabolism , Demyelinating Diseases/metabolism , Myelin Sheath/metabolism , Receptors, Notch/metabolism , Transferrin/pharmacology , Animals , Brain/drug effects , Brain/pathology , Corpus Callosum/drug effects , Corpus Callosum/metabolism , Corpus Callosum/pathology , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Female , Lysophosphatidylcholines , Male , Neural Stem Cells/pathology , Oligodendroglia/pathology , Rats , Rats, Wistar , Signal Transduction
16.
Biomed Pharmacother ; 67(2): 164-71, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23206754

ABSTRACT

Targeting and inhibiting angiogenesis is a promising strategy for treatment of cancer. NGR peptide motif is a tumor-homing peptide, which could bind with CD13 expressed on tumor blood vessels. Lidamycin is a highly potent antitumor antibiotic, which is composed of an apoprotein (LDP) and an active enediyne chromophore (AE). Here, an NGR-integrated and enediyne-energized apoprotein composed of cyclic NGR peptide and lidamycin was developed by a two-step procedure. Firstly, we prepared the fusion protein composed of NGR peptide and LDP by recombinant DNA technology. Then, AE was reloaded to the fusion protein to get NGR-LDP-AE. Our experiments showed that NGR-LDP could bind to CD13-expressing HT-1080 cells, whereas the recombinant LDP (rLDP) showed weak binding. NGR-LDP-AE exerted highly potent cytotoxicity to cultured tumor cells in vitro. In vivo antitumor activity was evaluated in murine hepatoma 22 (H22) model and human fibrosarcoma HT-1080 model. At the tolerable dose, NGR-LDP-AE and lidamycin inhibited H22 tumor growth by 94.8 and 66.9%, and the median survival time of the mice was 62 and 37 days, respectively. In the HT-1080 model, NGR-LDP-AE inhibited tumor growth by 88.6%, which was statistically different from that of lidamycin (74.5%). Immunohistochemical study showed that NGR-LDP could bind to tumor blood vessels. Conclusively, these results demonstrate that fusion of LDP with CNGRC peptide delivers AE to tumor blood vessels and improves its antitumor activity.


Subject(s)
Apoproteins/pharmacology , CD13 Antigens/metabolism , Enediynes/pharmacology , Oligopeptides/pharmacology , Aminoglycosides/pharmacology , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Apoproteins/genetics , Apoproteins/metabolism , Cell Line, Tumor , Female , Fibrosarcoma/drug therapy , Hep G2 Cells , Humans , Liver Neoplasms, Experimental/drug therapy , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Oligopeptides/genetics , Oligopeptides/metabolism , Protein Binding/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology
17.
Biometals ; 25(6): 1247-59, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23001680

ABSTRACT

Apo-form of human lactoferrin (LF) is a potent iron chelator, this feature being similar to the iron-binding properties of a synthetic chelator desferoxamine (DFO). The latter stabilizes the principal adaptive transcriptional hypoxia-inducible factor-1 alpha (HIF-1α). Since DFO is known as a pharmacological mimetic of hypoxia it was decided to test whether apo-LF is able to perform as such. Mice either injected intraperitoneally or given per os apo-LF displayed HIF-1α in liver, lungs, heart, brain, spleen and kidneys, as judged by results of Western blotting. Similar administration of iron-saturated LF (75 mg/kg) did not bring forth such effect. Synthesis of erythropoietin and ceruloplasmin became increased in the first case, which is explained by the respective genes being targets for HIF-1α. Apo-LF, but not Fe-LF, injected intraperitoneally to hypoxia low-resistant mice 24 h before animals were subjected to normobaric hypoxia with hypercapnia caused a significant increase of life-time by 40 %. The results obtained show that, like DFO, apo-LF performs as a normoxic mimetic of hypoxia, capable of stabilizing HIF-1α. Protective features of LF and DFO and their pharmacological properties involving HIF-1α are discussed.


Subject(s)
Apoproteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Lactoferrin/metabolism , Apoproteins/pharmacology , Humans , Lactoferrin/pharmacology
18.
Eur J Pharm Sci ; 47(4): 781-9, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-22982402

ABSTRACT

Arginine-rich peptides belong to a subclass of cell penetrating peptides that are taken up by living cells and can be detected freely diffusing inside the cytoplasm and nucleoplasm. This phenomenon has been attributed to either an endocytotic mode of uptake and a subsequent release from vesicles or a direct membrane penetration. Lidamycin is an antitumor antibiotic, which consists of an active enediyne chromophore (AE) and a noncovalently bound apoprotein (LDP). In the present study, a fusion protein (Arg)(9)-LDP composed of cell penetrating peptide (Arg)(9) and LDP was prepared by DNA recombination, and the enediyne-energized fusion protein (Arg)(9)-LDP-AE was prepared by molecular reconstitution. The data in fixed cells demonstrated that (Arg)(9)-LDP could rapidly enter cells, and the results based on fluorescence activated cell sorting indicated that the major route for (Arg)(9)-mediated cellular uptake of protein molecules was endocytosis. (Arg)(9)-LDP-AE demonstrated more potent cytotoxicity against different carcinoma cell lines than lidamycin in vitro. In the mouse hepatoma 22 model, (Arg)(9)-LDP-AE (0.3mg/kg) suppressed the tumor growth by 89.2%, whereas lidamycin (0.05 mg/kg) by 74.6%. Furthermore, in the glioma U87 xenograft model in nude mice, (Arg)(9)-LDP-AE at 0.2mg/kg suppressed tumor growth by 88.8%, compared with that of lidamycin by 62.9% at 0.05 mg/kg. No obvious toxic effects were observed in all groups during treatments. The results showed that energized fusion protein (Arg)(9)-LDP-AE was more effective than lidamycin and would be a promising candidate for glioma therapy. In addition, this approach to manufacturing fusion proteins might serve as a technology platform for the development of new cell penetrating peptides-based drugs.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoproteins/pharmacology , Cell-Penetrating Peptides/pharmacology , Enediynes/pharmacology , Recombinant Fusion Proteins/pharmacology , Aminoglycosides/pharmacology , Animals , Antibiotics, Antineoplastic/metabolism , Apoproteins/genetics , Apoproteins/metabolism , Arginine/genetics , Arginine/metabolism , Base Sequence , Cell Line, Tumor , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/metabolism , Endocytosis/drug effects , Endocytosis/physiology , Enediynes/metabolism , Female , Hep G2 Cells , Humans , MCF-7 Cells , Mice , Mice, Nude , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
19.
PLoS One ; 7(3): e33937, 2012.
Article in English | MEDLINE | ID: mdl-22479482

ABSTRACT

Neural stem and progenitor cells (NSC/NPCs) are multipotent self-renewing cells that are able to generate neurons, astrocytes and oligodendrocytes (OLs) within the adult central nervous system. We cultured NSC/NPCs from the rat subventricular zone as neurospheres (NS) and studied apoTransferrin (aTf) effects on oligodendroglial specification and maturation. Our findings suggest that aTf acts at different stages during progression from NSC to mature oligodendrocytes. On the one hand, an early event associated with the activation of NSC/NPCs proliferation and commitment toward the oligodendroglial fate, as indicated by increased BrdU incorporation, larger neurospheres production, and higher ability to generate OL precursors (OPCs) from undifferentiated cultures. On the other hand, aTf exposure during differentiating conditions favours OL maturation from OPCs by promoting OL morphological development. This evidence supports a key role of Tf on the generation of OL from NSC/NPCs and highlights its potential in demyelinating disorder treatment.


Subject(s)
Apoproteins/metabolism , Choroid Plexus/metabolism , Neural Stem Cells/metabolism , Transferrin/metabolism , Animals , Antigens/metabolism , Apoproteins/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Choroid Plexus/cytology , Female , Male , Models, Biological , Neural Stem Cells/cytology , Oligodendroglia/cytology , Oligodendroglia/metabolism , Primary Cell Culture , Proteoglycans/metabolism , Rats , Rats, Wistar , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptors, Transferrin/immunology , Receptors, Transferrin/metabolism , Transferrin/pharmacology
20.
Biochem Cell Biol ; 90(3): 442-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22332831

ABSTRACT

Influenza is a highly contagious, acute respiratory illness, which represents one of the main plagues worldwide. Even though some antiviral drugs are available, the alarming increase of virus strains resistant to them highlights the need to find new antiviral compounds. As we have recently demonstrated that bovine lactoferrin (bLf) prevents influenza virus-induced apoptosis, in the present wor,k we have attempted to investigate in depth the mechanism of the anti-influenza virus effect of this protein. To this aim, experiments have been carried out whereby different forms of bLf were added to the cells during different phases of viral infection. Results obtained showed that bLf was able to prevent influenza virus cytopathic effects when incubated with the cells after the adsorption step, independently from ion saturation or carbohydrate content. Moreover, the influence of iron saturations or sialic acid/carbohydrates removal on bLf activity on the early phases of infection has been observed. Our results provide further insights on the antiviral activity of bLf and suggest novel strategies for treatment of influenza virus infection.


Subject(s)
Antiviral Agents/pharmacology , Apoproteins/pharmacology , Influenza A Virus, H3N2 Subtype/drug effects , Lactoferrin/pharmacology , Orthomyxoviridae Infections/virology , Animals , Antiviral Agents/chemistry , Apoproteins/chemistry , Apoptosis , Cattle , Cell Line , Cell Shape , Dogs , Dose-Response Relationship, Drug , Glycosylation , Host-Pathogen Interactions , Influenza A Virus, H3N2 Subtype/physiology , Inhibitory Concentration 50 , Iron/chemistry , Lactoferrin/chemistry , Manganese/chemistry , Protein Binding , Virus Attachment/drug effects , Zinc/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL