Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Mutagenesis ; 36(6): 445-455, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34612487

ABSTRACT

Advanced glycation end products (AGEs) are formed via non-enzymatic reactions between amino groups of proteins and the carbonyl groups of reducing sugars. Previous studies have shown that highly glycated albumin prepared using a glucose-bovine serum albumin (Glu-BSA) model system incubated at 60°C for 6 weeks induces genotoxicity in WIL2-NS cells at 9 days of exposure measured by the cytokinesis-block micronucleus cytome (CBMNcyt) assay. However, this AGE model system is not physiologically relevant as normal body temperature is 37°C and the degree of glycation may exceed the extent of albumin modification in vivo. We hypothesised that the incubation temperature and purification method used in these studies may cause changes to the chemical profile of the glycated albumin and may influence the extent of genotoxicity observed at 3, 6 and 9 days of exposure. We prepared AGEs generated using Glu-BSA model systems incubated at 60°C or 37°C purified using trichloroacetic acid (TCA) precipitation or ultrafiltration (UF) and compared their chemical profile (glycation, oxidation, and aggregation) and genotoxicity in WIL2-NS cells using the CBMNcyt assay after 3, 6 and 9 days of exposure. The number of micronuclei (MNi) was significantly higher for cells treated with Glu-BSA incubated at 60°C and purified via TCA (12 ± 1 MNi/1000 binucleated cells) compared to Glu-BSA incubated at 37°C and purified using UF (6 ± 1 MNi/1000 binucleated cells) after 9 days (P < 0.0001). The increase in genotoxicity observed could be explained by a higher level of protein glycation, oxidation, and aggregation of the Glu-BSA model system incubated at 60°C relative to 37°C. This study highlighted that the incubation temperature, purification method and cell exposure time are important variables to consider when generating AGEs in vitro and will enable future studies to better reflect in vivo situations of albumin glycation.


Subject(s)
Cytokinesis/drug effects , Glycation End Products, Advanced/toxicity , Serum Albumin/toxicity , Toxicity Tests/methods , Cell Line , Glucose/metabolism , Glycation End Products, Advanced/metabolism , Glycosylation , Humans , Micronucleus Tests/methods , Serum Albumin/metabolism , Temperature , Glycated Serum Albumin
2.
Mol Cell Endocrinol ; 479: 87-92, 2019 01 05.
Article in English | MEDLINE | ID: mdl-30253183

ABSTRACT

Vitamin D3 (1,25-dihydroxyvitamin D3, VD3) in vitro attenuates the effect of the pro-inflammatory advanced glycation end products (AGEs) on steroidogenesis in human granulosa cells (GCs) by downregulating the receptor for AGEs (RAGE). It has been shown that VD3 alone downregulates anti-Mullerian hormone (AMH) type 2 receptor (AMHR-2) gene expression and suppresses AMH-induced SMAD 1/5/8 phosphorylation in granulosa cells. However, the effect of AGEs, in the absence or presence of VD3, on AMH action in GCs has not been studied. Using human GCs, this study showed that human glycated albumin (HGA), an in vitro representative for AGEs, upregulated AMHR-2 mRNA but did not alter AMH mRNA expression levels. VD3 inhibited the HGA-induced increase in AMHR-2 mRNA expression levels. In KGN granulosa cell line, recombinant AMH induced SMAD 1/5/8 phosphorylation. HGA augmented the recombinant AMH-induced SMAD 1/5/8 phosphorylation while the addition of VD3 to HGA attenuated the recombinant AMH-induced SMAD 1/5/8 phosphorylation. Thus, AGEs could potentially affect folliculogenesis as reflected by changes in AMH signaling. These findings have significant implications for women with polycystic ovary syndrome who have significantly elevated serum and ovarian AGEs.


Subject(s)
Anti-Mullerian Hormone/metabolism , Glycation End Products, Advanced/toxicity , Signal Transduction , Vitamin D/pharmacology , Adult , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Female , Gene Expression Regulation/drug effects , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Humans , Luteinization/drug effects , Phosphorylation/drug effects , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Recombinant Proteins/pharmacology , Serum Albumin/toxicity , Smad Proteins/metabolism , Glycated Serum Albumin
3.
Int J Pharm ; 522(1-2): 198-209, 2017 Apr 30.
Article in English | MEDLINE | ID: mdl-28279738

ABSTRACT

Gene therapy bears great potential for the cure of a multitude of human diseases. Research efforts focussed on the use of viral delivery vectors in the past decades, neglecting non-viral gene therapies of physical or chemical origin due to low transfection efficiency. However, side effects such as activation of oncogenes and inflammatory reactions upon immune cell activation are major obstacles impeding the clinical applicability of viral gene therapy vectors. The aim of this study was the development of a non-viral gene delivery system based on plasmid-loaded human serum albumin nanoparticles, which are biocompatible, biodegradable, and non-toxic in relevant concentrations. The surface of said nanoparticles was modified with different cell penetrating peptides, namely Tat, nona-arginine R9, and the penetratin analogue EB1. We hypothesise that the surface modified nanoparticles can effectively enter HEK 293T cells based on the cell penetrating properties of the different peptides attached. A variety of inhibitors were used targeting distinct uptake pathways in an effort to understand the mechanisms utilized by the various cell penetrating peptides on the surface of the nanoparticles. A significant increase in transfection efficiency compared to free DNA or polyplexes was seen for these novel delivery vectors.


Subject(s)
Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/chemistry , Gene Transfer Techniques , Genetic Therapy/methods , Nanoparticles/chemistry , Plasmids/chemistry , Serum Albumin/chemistry , Cell Survival/drug effects , DNA/administration & dosage , DNA/chemistry , Excipients , Gene Products, tat/chemistry , HEK293 Cells , Humans , Serum Albumin/antagonists & inhibitors , Serum Albumin/toxicity , Transfection
4.
AAPS PharmSciTech ; 18(6): 1987-1997, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27933587

ABSTRACT

Gambogic acid (GA) has been proven to be a potent chemotherapeutic agent for the treatment of lung cancer in clinical trials. However, GA is limited in its therapeutic value by properties such as poor water solubility and low chemical stability. In clinical trials, cationic arginine (Arg) was added to solubilize GA, and this may also cause other side effects. Here, we have designed and developed a more efficient human serum albumin (HSA)-based delivery system for GA with low toxicity which helps improve its solubility, chemical stability and increases its antitumor efficacy. The GA-HSA nanoparticles (NPs) were prepared by albumin-bound (nabTM) technology, with a particle size of 135.2 ± 35.03 nm, a zeta potential of -21.81 ± 1.24 mV, and a high entrapment efficiency. Compared with GA-Arg solution, the physical and chemical stability of the NPs were improved when stored at pH 7.4 in PBS or freeze-dried. The in vitro drug release showed that GA-HSA NPs had a more sustained release than GA-Arg solution. Furthermore, HSA NPs improved the therapeutic efficacy of GA and were less toxic compared with GA-Arg solution in A549-bearing mice. Therefore, this delivery system is a promising polymeric carrier for GA when used for tumor therapy.


Subject(s)
Drug Carriers/toxicity , Lung Neoplasms/drug therapy , Serum Albumin/toxicity , Tumor Burden/drug effects , Xanthones/toxicity , A549 Cells , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Drug Carriers/chemistry , Drug Liberation , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, SCID , Nanoparticles/chemistry , Particle Size , Serum Albumin/chemistry , Treatment Outcome , Tumor Burden/physiology , Xanthones/chemistry
5.
Bioconjug Chem ; 28(2): 449-460, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27992181

ABSTRACT

In recent years, we and others have become interested in evaluating the use of surface-enhanced Raman scattering (SERS) tags for early cancer detection and in designing new approaches to demonstrate the applicability of this spectroscopic technique in the clinic. SERS-based imaging in particular offers ultra sensitivity up to the single molecule, multiplexing capability, and increased photostability and has been shown to outperform fluorescence. However, to employ SERS tags for early cancer detection, it is important to understand their interaction with cells and determine their cytotoxicity. We have been particularly interested for quite some time in determining if and how gold nanostars, which have been demonstrated as outstanding SERS enhancing substrates, can be safely employed in living systems and translated to the clinic. In this study, we carried out a multiparametric in vitro study to look at the cytotoxicity and cellular uptake of gold nanoparticles on human glioblastoma and human dermal fibroblast cell lines. Cytotoxicity was evaluated by incubating cells with three different morphologies of AuNPs, namely nanospheres, nanorods, and nanostars, each having three different surface chemistries (cetyltrimethylammonium bromide (CTAB), poly(ethylene glycol) (PEG), and human serum albumin (HSA)). Our results showed that the surface chemistry of the nanoparticles had predominant effects on cytotoxicity, and the morphology and size of the nanoparticles only slightly affected cell viability. CTAB-coated particles were found to be the most toxic to cells, and PEGylated nanostars were determined to be the least toxic. Caspase-3 assay and LDH assay revealed that cell death occurs via apoptosis for cancerous cells and via necrosis for healthy ones. Cellular uptake studies carried out via TEM showed that the particles retain their shape even at long incubation times, which may be beneficial for in vivo SERS-based disease detection. Overall, this study provides valuable information on gold-nanoparticle-induced cytotoxicity that can be leveraged for the development of safe and effective nanoparticle-based therapeutic and diagnostic systems.


Subject(s)
Cell Survival/drug effects , Gold/toxicity , Metal Nanoparticles/toxicity , Caspase 3/metabolism , Cell Line, Tumor , Cells, Cultured , Cetrimonium , Cetrimonium Compounds/chemistry , Cetrimonium Compounds/toxicity , Gold/chemistry , Humans , Metal Nanoparticles/chemistry , Metal Nanoparticles/ultrastructure , Polyethylene Glycols/chemistry , Polyethylene Glycols/toxicity , Serum Albumin/chemistry , Serum Albumin/toxicity , Spectrum Analysis, Raman/methods , Surface Properties
6.
Int J Pharm ; 511(2): 969-82, 2016 Sep 25.
Article in English | MEDLINE | ID: mdl-27498282

ABSTRACT

The majority of active agents do not readily permeate into brain due to the presence of the blood-brain barrier and blood-cerebrospinal fluid barrier. Currently, the most innovative and promising non-invasive strategy in brain delivery is the design and preparation of nanocarriers, which can move through the brain endothelium. Niosomes can perform brain delivery, in fact polysorbates, can act as an anchor for apolipoprotein E from blood plasma. The particles mimic LDL and interact with the LDL receptor leading to the endothelial cells uptake. The efficacy of niosomes for anticancer therapeutic applications was correlated to their physicochemical and drug delivery properties. Dimensions and ζ-potential were characterized using dynamic light scattering and asymmetric flow-field fractionation system. Lipid bilayer was characterized measuring the fluidity, polarity and microviscosity by fluorescent probe spectra evaluation. Morphology and homogeneity were characterized using atomic force microscopy. Physicochemical stability and serum stability (45% v/v fetal bovine and human serum) were evaluated as a function of time using dynamic light scattering. U87-MG human glioblastoma cells were used to evaluate vesicle cytotoxicity and internalisation efficiency. From the obtained data, the systems appear useful to perform a prolonged (modified) release of biological active substances to the central nervous system.


Subject(s)
Blood-Brain Barrier/drug effects , Brain/drug effects , Drug Delivery Systems/methods , Liposomes/administration & dosage , Liposomes/toxicity , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Cattle , Cell Line, Tumor , Humans , Liposomes/chemistry , Serum Albumin/administration & dosage , Serum Albumin/chemistry , Serum Albumin/toxicity
7.
Am J Physiol Renal Physiol ; 310(5): F409-15, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26739891

ABSTRACT

Our previous studies have suggested a critical role of reticulon (RTN)1A in mediating endoplasmic reticulum (ER) stress in kidney cells of animal models and humans with kidney diseases. A large body of evidence suggests that proteinuria itself can cause tubular cell injury leading to the progression of kidney disease. In the present study, we determined whether RTN1A mediates proteinuria-induced tubular cell injury through increased ER stress. We found that incubation of HK2 cells with human serum albumin induced the expression of RTN1A and ER stress markers, whereas knockdown of RTN1A expression attenuated human serum albumin-induced ER stress and tubular cell apoptosis in vitro. In vivo, we found that tubular cell-specific RTN1 knockdown resulted in a significant attenuation of tubular cell ER stress, apoptosis, and renal fibrosis in a model of albumin overload nephropathy. Based on these findings, we conclude that RTN1A is a key mediator for proteinuria-induced tubular cell toxicity and renal fibrosis.


Subject(s)
Albuminuria/metabolism , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Kidney Tubules/drug effects , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/metabolism , Renal Insufficiency, Chronic/prevention & control , Serum Albumin, Bovine , Serum Albumin/toxicity , Albuminuria/chemically induced , Albuminuria/genetics , Albuminuria/pathology , Animals , Disease Models, Animal , Disease Progression , Fibrosis , Genotype , HEK293 Cells , Humans , Kidney Tubules/metabolism , Kidney Tubules/pathology , Mice, Knockout , Nerve Tissue Proteins/genetics , Phenotype , RNA Interference , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Serum Albumin, Human , Signal Transduction/drug effects , Time Factors , Transfection
8.
J Fluoresc ; 25(6): 1721-6, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26410776

ABSTRACT

The non-enzymatic reaction between reducing sugars and proteins has received increased attention in nutritional and medical research recently. In the current manuscript, effect of glycation in structural changes of human serum albumin (HSA) by the metabolites of glucose such as glyoxal, methylglyoxal and glyceraldehyde was studied using different spectroscopy techniques. Glycation of HSA was monitored by following advanced glycation end-products (AGEs) fluorescence changes, HSA intrinsic fluorescence measurement, extrinsic fluorescence using 8-analino 1-nephthlene sulfonic acid (ANS) dye, and circular dichroism (CD) studies. AGEs were formed within 7 days of incubation with glyoxal, methylglyoxal and glyceraldehyde. However, methylglyoxal induced significant structural changes in HSA compared with glyoxal and glyceraldehydes. Moreover, ANS binding to native and glycated-HSA showed difference in binding pattern of these metabolites to HSA. The CD spectrum revealed changes in the secondary structure of HSA upon glycation when compared to native HSA. Furthermore, the MTT (3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide) assay established the cytotoxicity of the glycated- HSA towards human liver carcinoma (HepG2) cell lines via the production of reactive oxygen species.


Subject(s)
Cytotoxins/metabolism , Cytotoxins/toxicity , Glucose/metabolism , Oxidative Stress/drug effects , Serum Albumin/metabolism , Serum Albumin/toxicity , Glycation End Products, Advanced , Glycosylation , Hep G2 Cells , Humans , Reactive Oxygen Species/metabolism , Glycated Serum Albumin
9.
Exp Eye Res ; 137: 50-6, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26070987

ABSTRACT

Diabetic macular edema (DME) is a major cause of visual impairment. Although DME is generally believed to be a microvascular disease, dysfunction of the retinal pigment epithelium (RPE) can also contribute to its development. Advanced glycation end-products (AGE) are thought to be one of the key factors involved in the pathogenesis of diabetes in the eye, and we have previously demonstrated a rapid breakdown of RPE function following glycated-albumin (Glyc-alb, a common AGE mimetic) administration in monolayer cultures of fetal human RPE cells. Here we present new evidence that this response is attributed to apically oriented AGE receptors (RAGE). Moreover, time-lapse optical coherence tomography in Dutch-belted rabbits 48 h post intravitreal Glyc-alb injections demonstrated a significant decrease in RPE-mediated fluid resorption in vivo. In both the animal and tissue culture models, the response to Glyc-alb was blocked by the relatively selective RAGE antagonist, FPS-ZM1 and was also inhibited by ZM323881, a relatively selective vascular endothelial growth factor receptor 2 (VEGF-R2) antagonist. Our data establish that the Glyc-alb-induced breakdown of RPE function is mediated via specific RAGE and VEGF-R2 signaling both in vitro and in vivo. These results are consistent with the notion that the RPE is a key player in the pathogenesis of DME.


Subject(s)
Macular Edema/physiopathology , Retinal Pigment Epithelium/physiopathology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Blood-Retinal Barrier , Cadaver , Cells, Cultured , Disease Models, Animal , Glycation End Products, Advanced , Humans , Immunoblotting , Immunohistochemistry , Macular Edema/chemically induced , Macular Edema/pathology , Rabbits , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Serum Albumin/toxicity , Tomography, Optical Coherence , Glycated Serum Albumin
10.
Br J Pharmacol ; 172(5): 1237-49, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25363737

ABSTRACT

BACKGROUND AND PURPOSE: Nuclear factor erythroid 2-related factor 2 (Nrf2) is a redox-sensitive transcription factor that binds to antioxidant response elements located in the promoter region of genes encoding many antioxidant enzymes and phase II detoxifying enzymes. Activation of the Nrf2 pathway seems protective for many organs, and although a well-known Nrf2 activator, bardoxolone methyl, was evaluated clinically for treating chronic kidney disease, it was found to induce adverse events. Many bardoxolone methyl derivatives, mostly derived by chemical modifications, have already been studied. However, we adopted a biotransformation technique to obtain a novel Nrf2 activator. EXPERIMENTAL APPROACH: The potent novel Nrf2 activator, RS9, was obtained from microbial transformation products. Its Nrf2 activity was evaluated by determining NADPH:quinone oxidoreductase-1 induction activity in Hepa1c1c7 cells. We also investigated the effects of RS9 on oxygen-induced retinopathy in rats and glycated albumin-induced blood-retinal barrier permeability in rabbits because many ocular diseases are associated with oxidative stress and inflammation. KEY RESULTS: Bardoxolone methyl doubled the specific activity of Nrf2 in Hepa1c1c7 cells at a much higher concentration than RS9. Moreover, the induction of Nrf2-targeted genes was observed at a one-tenth lower concentration of RS9. Interestingly, the cytotoxicity of RS9 was substantially reduced compared with bardoxolone methyl. Oral and intravitreal administration of RS9 ameliorated the pathological scores and leakage in the models of retinopathy in rats and ocular inflammation in rabbits respectively. CONCLUSION AND IMPLICATIONS: Nrf2 activators are applicable for treating ocular diseases and novel Nrf2 activators have potential as a unique method for prevention and treatment of retinovascular disease.


Subject(s)
Blood-Retinal Barrier/drug effects , NF-E2-Related Factor 2/metabolism , Triterpenes/pharmacology , Animals , Cell Line , Female , Glycation End Products, Advanced , Humans , Male , Mice , Oxygen/toxicity , Permeability/drug effects , Rabbits , Rats , Rats, Sprague-Dawley , Retinopathy of Prematurity/chemically induced , Retinopathy of Prematurity/pathology , Serum Albumin/antagonists & inhibitors , Serum Albumin/toxicity , Triterpenes/chemistry , Glycated Serum Albumin
11.
J Control Release ; 194: 130-7, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25173842

ABSTRACT

Iron oxide-containing magnetic nanoparticles (MNPs) have certain advantages over currently used contrast agents for tumor imaging by magnetic resonance imaging (MRI) as they offer the possibility of functionalization with ligands and tracers. Functionalized MNPs also may be used for targeted tumor therapy. In the current study nanoparticles (NPs) consisting of recombinant human serum albumin (rHSA) with incorporated hydrophilic (NH4)2Ce(IV)(NO3)6-γ-Fe2O3 particles (CAN maghemite particles) for medical imaging were produced and characterized. For this purpose CAN maghemite particles were incorporated into an rHSA matrix to yield rHSA-NPs. The resulting NPs were analyzed by transmission electron microscopy, photon correlation spectroscopy, and atomic absorption. The sizes of the manufactured NP were 170 ± 10 nm, and the zeta-potential was -50 ± 3 mV. The NPs remained stable when stored after lyophilization with sucrose 3% [w/v] as a cryoprotector. They showed pro-inflammatory properties without cell and animal toxicity in vivo and were highly contrasting in MRI. In conclusion, this report introduces novel rHSA NP with favorable properties containing iron oxide for detection by MRI.


Subject(s)
Contrast Media , Diagnostic Imaging/methods , Ferric Compounds , Magnetite Nanoparticles , Serum Albumin , Animals , Cell Line, Tumor , Cell Survival/drug effects , Contrast Media/administration & dosage , Contrast Media/toxicity , Drug Stability , Electrochemistry , Ferric Compounds/administration & dosage , Ferric Compounds/chemistry , Ferric Compounds/toxicity , Humans , Magnetic Resonance Imaging , Magnetics , Magnetite Nanoparticles/administration & dosage , Magnetite Nanoparticles/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neoplasms/diagnosis , Particle Size , Recombinant Proteins/administration & dosage , Recombinant Proteins/toxicity , Serum Albumin/administration & dosage , Serum Albumin/toxicity
12.
Cardiovasc Diabetol ; 13: 8, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24397367

ABSTRACT

BACKGROUND: High blood and tissue concentrations of glucose and advanced glycation end-products are believed to play an important role in the development of vascular complications in patients with diabetes mellitus (DM) and chronic kidney disease. MicroRNAs (miRNA) are non-coding RNAs that regulate gene expression in a sequence specific manner. MiRNA are involved in various biological processes and become novel biomarkers, modulators and therapeutic targets for diseases such as cancer, atherosclerosis, and DM. Calcitriol (the active form of vitamin D) may inhibit endothelial proliferation, blunt angiogenesis, and be a cardioprotective agent. Calcitriol deficiency is a risk factor for DM and hypertension. The aim of this project was to study the miRNA microarray expression changes in human umbilical vein endothelial cells (HUVEC) treated in a diabetic-like environment with the addition of calcitriol. METHODS: HUVEC were treated for 24 h with 200 µg/ml human serum albumin (HSA) and 100 mg/dl glucose (control group) or 200 µg/ml AGE-HSA, and 250 mg/dl glucose (diabetic-like environment), and physiological concentrations (10-10 mol/l) of calcitriol. miRNA microarray analysis and real time PCR to validate the miRNA expression profile and mRNA target gene expression were carried out. RESULTS: Compared to control, 31 mature human miRNA were differentially expressed in the presence of a diabetic-like environment. Addition of physiological concentrations of calcitriol revealed 39 differentially expressed mature human miRNA. MiR-181c, miR-15a, miR-20b, miR-411, miR-659, miR-126 and miR-510 were selected for further analysis because they are known to be modified in DM and in other biological disorders. The predicted targets of these miRNA (such as KLF6, KLF9, KLF10, TXNIP and IL8) correspond to molecular and biological processes such as immune and defense responses, signal transduction and regulation of RNA. CONCLUSION: This study identified novel miRNA in the field of diabetic vasculopathy and might provide new information about the effect of vitamin D on gene regulation induced by a diabetic-like environment. New gene targets that are part of the molecular mechanism and the therapeutic treatment in diabetic vasculopathy are highlighted.


Subject(s)
Diabetes Mellitus/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , MicroRNAs/biosynthesis , Vitamin D/pharmacology , Cells, Cultured , Diabetes Mellitus/chemically induced , Diabetes Mellitus/drug therapy , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/physiology , Glucose/toxicity , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Serum Albumin/toxicity , Vitamin D/therapeutic use
13.
Cell Stress Chaperones ; 19(1): 105-14, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23719694

ABSTRACT

In diabetic kidney disease (DKD), epithelial-to-mesenchymal transition (EMT) is a classic pathological process in tubular damage. Oxidative stress is considered to play an important role in DKD. Astragaloside IV (A-IV), one of the main active ingredients of Astragalus membranaceus, exhibits a wide range of biological activities. However, the effect of A-IV on regulating EMT in tubular cells is unclear. This study aims to determine whether A-IV could attenuate glycated albumin (GA)-induced EMT in the NRK-52E cell line by inhibiting oxidative stress. GA and A-IV-induced cytotoxicity were assayed by CCK-8. The intercellular reactive oxygen species (ROS) level was detected by H2DCFDA. The activity of NADPH oxidase was assayed by adding exogenous NADPH oxidase, and the superoxide dismutase (SOD) units were observed by NBT. We used a microscope to examine the morphology of the NRK-52E cell line. We conducted a wound healing assay to measure cell mobility. To determine mRNA and protein expressions of α-SMA and E-cadherin, we used real-time polymerase chain reaction (real-time PCR), immunofluorescence, and western blot analysis. A-IV significantly attenuated GA-induced amplification of ROS, lowered the increased level of NADPH oxidase activity, and elevated the decreased level of SOD units. The GA-induced NRK-52E cell line showed increased expression of α-SMA and decreased expression of E-cadherin in mRNA and protein levels, whereas A-IV alleviated the expression of α-SMA and increased the expression of E-cadherin. Our data demonstrate that GA could induce NRK-52E cell line EMT through oxidative stress. This effect could be attenuated by A-IV via regulation of the impaired redox balance.


Subject(s)
Oxidative Stress/drug effects , Saponins/pharmacology , Serum Albumin/toxicity , Triterpenes/pharmacology , Actins/genetics , Actins/metabolism , Animals , Antioxidants/pharmacology , Astragalus Plant/chemistry , Astragalus Plant/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line , Cell Movement/drug effects , Cell Survival/drug effects , Epithelial-Mesenchymal Transition/drug effects , Glycation End Products, Advanced , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , RNA, Messenger/metabolism , Rats , Reactive Oxygen Species/metabolism , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/metabolism , Glycated Serum Albumin
14.
J Appl Toxicol ; 34(7): 795-804, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24193864

ABSTRACT

Aflatoxins (AFs) and fumonisins (FBs) can co-contaminate foodstuffs and have been associated with hepatocellular and esophageal carcinomas in humans at high risk for exposure. One strategy to reduce exposure (and toxicity) from contaminated foodstuffs is the dietary inclusion of a montmorillonite clay (UPSN) that binds AFs and FBs in the gastrointestinal tract. In this study, the binding capacity of UPSN was evaluated for AFB1, FB1 and a combination thereof in Fischer 344 rats. Rats were pre-treated with different dietary levels of UPSN (0.25% or 2%) for 1 week. Rats were gavaged with a single dose of either 0.125 mg AFB1 or 25 mg FB1 per kg body weight and a combination thereof in the presence and absence of an aqueous solution of UPSN. The kinetics of mycotoxin excretion were monitored by analyzing serum AFB1 -albumin, urinary AF (AFM1) and FB1 biomarkers over a period of 72 h. UPSN decreased AFM1 excretion by 88-97%, indicating highly effective binding. FB1 excretion was reduced, to a lesser extent, ranging from 45% to 85%. When in combination, both AFB1 and FB1 binding occurred, but capacity was decreased by almost half. In the absence of UPSN, the combined AFB1 and FB1 treatment decreased the urinary biomarkers by 67% and 45% respectively, but increased levels of AFB1 -albumin, presumably by modulating its cytochrome metabolism. UPSN significantly reduced bioavailability of both AFB1 and FB1 when in combination; suggesting that it can be utilized to reduce levels below their respective thresholds for affecting adverse biological effects.


Subject(s)
Aflatoxin B1/toxicity , Aluminum Silicates/pharmacology , Bentonite/pharmacology , Calcium/pharmacology , Fumonisins/toxicity , Serum Albumin/toxicity , Aflatoxin B1/blood , Aflatoxin B1/urine , Aluminum Silicates/chemistry , Animals , Bentonite/chemistry , Biomarkers/blood , Biomarkers/urine , Calcium/chemistry , Clay , Fumonisins/blood , Fumonisins/urine , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Male , Rats , Rats, Inbred F344
15.
Fertil Steril ; 100(2): 544-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23602317

ABSTRACT

OBJECTIVE: To screen human serum albumin (HSA) preparations for toxicity and investigate causes of variation. DESIGN: Experimental laboratory study. SETTING: University-based laboratory. ANIMAL(S): FVB and CF1 mice crossed to create embryos used in experiments. INTERVENTION(S): Mouse embryo assay performed with 5% or 15% HSA (100 mg/mL albumin) from three samples from three separate manufacturers (A, B, C). MAIN OUTCOME MEASURE(S): Blastocyst rates calculated at 96 hours of culture (experiments repeated in triplicate). RESULT(S): The HSA preparations were desalted to remove stabilizers added during HSA processing, then mass spectrometry was used to determine the relative variation in stabilizer concentrations; the effect of the stabilizer octanoic acid on embryo development was tested. At 5% HSA, all samples had blastocyst rates ≥ 70%; at 15% HSA, the blastocyst rates for samples B and C were <50%. Desalting did not affect sample B but did improve the blastocyst rates of sample C. Mass spectrometry revealed high levels of octanoic acid in sample C compared with sample A. The addition of octanoic acid to sample A produced toxicity similar to sample C. CONCLUSION(S): The stabilizer octanoic acid varies by lot and inhibits embryo development. Because octanoic acid is known to cause disruptions in mitochondrial bioenergetics, reduce intracellular pH, and induce oxidative damage in peripheral tissues, its use in embryo culture should be monitored and limited.


Subject(s)
Caprylates/toxicity , Culture Media/standards , Embryo Culture Techniques/standards , Embryo, Mammalian/drug effects , Excipients/toxicity , Animals , Culture Media/chemistry , Culture Media/toxicity , Dose-Response Relationship, Drug , Drug Stability , Embryo Culture Techniques/methods , Embryo Culture Techniques/statistics & numerical data , Embryo, Mammalian/cytology , Female , Humans , Mice , Quality Control , Salts/chemistry , Serum Albumin/chemistry , Serum Albumin/pharmacology , Serum Albumin/toxicity
16.
Inflamm Bowel Dis ; 19(4): 683-90, 2013.
Article in English | MEDLINE | ID: mdl-23429466

ABSTRACT

OBJECTIVE: Treatment of Crohn's disease (CD) with anti-tumor necrosis factor α (TNFα) decreases intestinal inflammation, but the effect on fibrosis remains unclear. We hypothesized that treatment with rat-specific anti-TNFα will decrease the development of intestinal fibrosis in a rat model of CD. We further hypothesized that magnetization transfer magnetic resonance imaging (MT-MRI) will be sensitive in detecting these differences in collagen content. METHODS: Rats were injected in the distal ileum and cecum with peptidoglycan-polysaccharide (PG-PS) or human serum albumin (control) at laparotomy and then received intraperitoneal injections of rat-specific anti-TNFα or vehicle daily for 21 days after laparotomy. Rats underwent MT-MRI abdominal imaging on day 19 or 20. MT ratio was calculated in the cecal wall. Cecal tissue histologic inflammation was scored. Cecal tissue procollagen, cytokine, and growth factor messenger RNAs were measured by quantitative real-time PCR. RESULTS: PG-PS-injected rats treated with anti-TNFα had less histologic inflammation, and cecal tissue expressed lower levels of proinflammatory cytokine messenger RNAs than vehicle-treated PG-PS-injected rats (IL-1ß: 5.59 ± 1.53 versus 10.41 ± 1.78, P = 0.02; IL-6: 23.23 ± 9.33 versus 45.89 ± 11.79, P = 0.07). PG-PS-injected rats treated with anti-TNFα developed less intestinal fibrosis than vehicle-treated PG-PS-injected rats by tissue procollagen I (2.87 ± 0.66 versus 9.28 ± 1.11; P = 0.00002), procollagen III (2.25 ± 0.35 versus 7.28 ± 0.76; P = 0.0000009), and MT-MRI (MT ratio: 17.79 ± 1.61 versus 27.95 ± 1.75; P = 0.0001). Insulin-like growth factor I (2.52 ± 0.44 versus 5.14 ± 0.60; P = 0.0007) and transforming growth factor ß1 (2.34 ± 0.29 versus 3.45 ± 0.29; P = 0.006) were also decreased in anti-TNFα-treated PG-PS-injected rats. CONCLUSIONS: Anti-TNFα prevents the development of bowel wall inflammation and fibrosis in the PG-PS rat model of CD. MT-MRI measurably demonstrates this decrease in intestinal fibrosis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Crohn Disease/prevention & control , Fibrosis/prevention & control , Intestinal Diseases/prevention & control , Magnetic Resonance Imaging , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Colitis/chemically induced , Colitis/pathology , Colitis/prevention & control , Crohn Disease/chemically induced , Crohn Disease/pathology , Cytokines/genetics , Disease Models, Animal , Female , Fibrosis/chemically induced , Fibrosis/pathology , Humans , Inflammation/chemically induced , Inflammation/pathology , Inflammation/prevention & control , Insulin-Like Growth Factor I/genetics , Intestinal Diseases/chemically induced , Intestinal Diseases/pathology , Magnetics , Peptidoglycan/toxicity , Procollagen/genetics , Rats , Rats, Inbred Lew , Real-Time Polymerase Chain Reaction , Serum Albumin/toxicity
17.
Diab Vasc Dis Res ; 10(1): 57-64, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22610271

ABSTRACT

Mainstream and sidestream tobacco smoke extracts have been shown to increase platelet activation directly. Furthermore, advanced glycation end products, which are present in the diabetic vasculature, have also been shown to enhance platelet activity. However, the combined effects of these two risk factors on platelet functions remain unclear. Platelets were exposed to tobacco extracts concurrently with advanced glycation end products. Timed samples were removed to assess the extent of platelet activity. The presence of smoke extracts enhanced platelet activity as compared to control conditions, this was especially prevalent for sidestream extracts. With the addition of irreversibly glycated albumin, there was an additive effect, further enhancing platelet responses. This was at least partially regulated by α-granule release and CD41 expression. The combination of cardiovascular risk factors can significantly enhance platelet activation and aggregation, and therefore it is possible to accelerate cardiovascular diseases through the interactions of multiple cardiovascular risk factors.


Subject(s)
Blood Platelets/drug effects , Nicotiana/toxicity , Nicotine/toxicity , Serum Albumin/toxicity , Smoke/adverse effects , Tobacco Smoke Pollution/adverse effects , Diabetes Mellitus/metabolism , Diabetes Mellitus/physiopathology , Glycation End Products, Advanced/pharmacology , Humans , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Platelet Glycoprotein GPIIb-IIIa Complex/drug effects , Smoking/metabolism , Smoking/physiopathology , Glycated Serum Albumin
18.
Biochem Biophys Res Commun ; 430(3): 1157-63, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23246834

ABSTRACT

The presence of glycated albumin (GA) is associated with increased diabetic complications. This study investigated the effect of angiotensin-(1-7) on the expression of GA-induced endothelial interleukin-6 (IL-6) in human aortic endothelial cells (HAECs). We also evaluated whether miR-146a is involved in the post-transcriptional regulation of angiotensin-(1-7). HAECs were stimulated with GA with or without angiotensin-(1-7) pretreatment. Inflammatory cytokine screening approach identified that angiotensin-(1-7) (10(-7) M) potently inhibited GA (200 µg/mL)-stimulated endothelial IL-6 expression in conditioned medium. ELISA confirmed this finding. Real-time PCR showed that angiotensin-(1-7) decreased GA-induced intracellular IL-6 mRNA expression and western blotting showed that angiotensin-(1-7) decreased GA-induced intracellular IL-6 protein expression. Bioinformatics' miR target analysis identified homology between miR-146a and the 3'-UTR of the human IL-6 mRNA, suggesting a potential regulation of IL-6 by miR-146a. Treatment with GA decreased endothelial miR-146a expression to 37.2% of the albumin control, while angiotensin-(1-7) increased endothelial miR-146a expression to 1.9-times that of the medium control. Pretreatment with angiotensin-(1-7) inhibited the GA-mediated downregulation of miR-146a to 78.9% of the albumin control levels. Furthermore, the inhibitory effect of angiotensin-(1-7) on IL-6 expression was abolished in GA-treated, miR-146a inhibitor-transfected HAECs. In conclusion, these results suggest that angiotensin-(1-7) exerted an endothelial protective effect through IL-6 downregulation, and miR-146a modulation is involved in this protective effect.


Subject(s)
Angiotensin I/pharmacology , Cytoprotection , Endothelium, Vascular/drug effects , Interleukin-6/biosynthesis , MicroRNAs/metabolism , Peptide Fragments/pharmacology , Serum Albumin/metabolism , Cells, Cultured , Culture Media, Conditioned/pharmacology , Down-Regulation , Endothelium, Vascular/metabolism , Glycation End Products, Advanced , Humans , Interleukin-6/antagonists & inhibitors , Serum Albumin/toxicity , Glycated Serum Albumin
19.
J Gastroenterol Hepatol ; 28(2): 369-76, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23173780

ABSTRACT

BACKGROUND AND AIMS: Advanced glycation end products (AGEs) are nonenzymatic modifications of proteins by reducing sugars. These compounds accumulate in a number of chronic disease states, contributing to tissue injury via several mechanisms, including activation of the receptor for advanced glycation end products (RAGE). We aimed to investigate whether AGEs can exacerbate chronic liver injury and contribute to hepatic fibrosis. METHODS: We initially studied the effects of chronic hepatic exposure to high levels of AGEs given intraperitoneally as AGE-rat serum albumin. In a separate experiment, we examined the impact of high AGE exposure in rats following bile duct ligation (BDL). RESULTS: In normal rats, chronic AGE-rat serum albumin administration induced significant increases in α-smooth muscle actin gene and protein expression but did not induce fibrosis or biochemical evidence of liver injury. However, in BDL animals, AGE-bovine serum albumin administration significantly increased hepatic fibrosis as evidenced by increased collagen content and α-smooth muscle actin expression, compared with BDL alone. Furthermore, AGEs increased hepatic oxidative stress and receptor for advanced glycation end products gene expression. CONCLUSIONS: These findings suggest that AGEs may contribute to the pathogenesis of chronic liver injury and fibrosis.


Subject(s)
Glycation End Products, Advanced/toxicity , Liver Cirrhosis, Experimental/chemically induced , Liver/drug effects , Serum Albumin/toxicity , Actins/genetics , Actins/metabolism , Animals , Biomarkers/metabolism , Collagen/metabolism , Common Bile Duct/surgery , Glycation End Products, Advanced/administration & dosage , Injections, Intraperitoneal , Ligation , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Receptor for Advanced Glycation End Products , Receptors, Immunologic/drug effects , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Serum Albumin/administration & dosage
20.
Nanotechnology ; 24(3): 035103, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23262833

ABSTRACT

Polymeric and biodegradable nanoparticles are frequently used in drug delivery systems. In this study silk fibroin-albumin blended nanoparticles were prepared using the desolvation method without any surfactant. These nanoparticles are easily internalized by the cells, reside within perinuclear spaces and act as carriers for delivery of the model drug methotrexate. Methotrexate loaded nanoparticles have better encapsulation efficiency, drug loading ability and less toxicity. The in vitro release behavior of methotrexate from the nanoparticles suggests that about 85% of the drug gets released after 12 days. The encapsulation and loading of a drug would depend on factors such as size, charge and hydrophobicity, which affect drug release. MTT assay and conjugation of particles with FITC demonstrate that the silk fibroin-albumin nanoparticles do not affect the viability and biocompatibility of cells. This blended nanoparticle, therefore, could be a promising nanocarrier for the delivery of drugs and other bioactive molecules.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Drug Carriers/chemistry , Fibroins/chemistry , Methotrexate/administration & dosage , Nanoparticles/chemistry , Serum Albumin/chemistry , Animals , Antimetabolites, Antineoplastic/pharmacology , Bombyx/chemistry , Breast Neoplasms/drug therapy , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Female , Fibroins/toxicity , Hemolysis/drug effects , Humans , Methotrexate/pharmacology , Nanoparticles/toxicity , Nanoparticles/ultrastructure , Particle Size , Serum Albumin/toxicity , Spectroscopy, Fourier Transform Infrared
SELECTION OF CITATIONS
SEARCH DETAIL
...