Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
J Environ Sci Health B ; 56(5): 467-476, 2021.
Article in English | MEDLINE | ID: mdl-33852377

ABSTRACT

Different technologies to prepare long term pesticide forms include polymer coating, preparing composites and encapsulating pesticides in nanoparticles. A simple and low-cost method was proposed to obtain slow-release formulations by co-extrusion of a pesticide with a biodegradable polymer at a temperature above the melting points of both components. A herbicide metribuzin and low-melting polyester poly-ε-caprolactone were chosen for this work. Formulations containing 10%, 20%, and 40% herbicide were prepared. During 7 days of their exposition in water, it was released from 81% to 96% of initially loaded metribuzin; the highest release was detected for 40%-loaded forms. Biodegradation of the constructs and pesticide release were further studied in the model soil. Degradation rates of the specimens increased with an increase in pesticide content, from 9% to 20% over 14 weeks for the 10%/20%-loaded and the 40%-loaded specimens, respectively. The release of metribuzin reached, respectively, 37-38% and 55%. The herbicide content in soil was lower due to its partial degradation in soil; it reached 23-25% and 33%, respectively, from initially loaded into the polymer matrix. Release kinetics of metribuzin in water as in soil best fitted the First-order model. The used approach is promising for obtaining long-term release formulations for soil applications.


Subject(s)
Caproates/chemistry , Herbicides/chemistry , Lactones/chemistry , Polyesters/chemistry , Soil Pollutants/chemistry , Triazines/chemistry , Biodegradation, Environmental , Caproates/analysis , Caproates/pharmacokinetics , Delayed-Action Preparations , Drug Compounding , Herbicides/analysis , Herbicides/pharmacokinetics , Kinetics , Lactones/analysis , Lactones/pharmacokinetics , Polyesters/analysis , Polyesters/pharmacokinetics , Soil/chemistry , Soil Pollutants/analysis , Soil Pollutants/pharmacokinetics , Triazines/analysis , Triazines/pharmacokinetics
2.
Acta Biomater ; 106: 256-266, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32058082

ABSTRACT

Here, we describe innovative synthesis of well-defined biocompatible N-(2-hydroxypropyl) methacrylamide (HPMA)-based polymer carriers and their drug conjugates with pirarubicin intended for controlled drug delivery and pH-triggered drug activation in tumor tissue. Polymer carrier synthesis was optimized to obtain well-defined linear HPMA-based polymer precursor with dispersity close to 1 and molar mass close to renal threshold with minimal synthesis steps. The developed synthesis enables preparation of tailored polymer nanomedicines with highly enhanced biological behavior in vivo, especially the biodistribution, urine elimination, tumor accumulation and anticancer activity. STATEMENT OF SIGNIFICANCE: The manuscript reports on novel synthesis and detailed physicochemical characterization and in vivo evaluation of well-defined biocompatible hydrophilic copolymers based on N-(2-hydroxypropyl)methacrylamide (HPMA) and their drug conjugates with pirarubicin enabling controlled drug delivery and pH-triggered drug activation in tumor tissue. Polymer carrier synthesis was optimized to obtain well-defined linear HPMA-based polymer precursor with minimal synthesis steps using controlled polymerization. Compared to previously published HPMA-based polymer drug conjugates whose polymer carriers were prepared by classical route via free radical polymerization, the newly prepared polymer drug conjugates exhibited enhanced biological behavior in vivo, especially the prolonged blood circulation, urine elimination, tumor accumulation and excellent anticancer activity. We believe that the newly prepared well-defined polymer conjugates could significantly enhance tumor therapy in humans.


Subject(s)
Acrylamides/therapeutic use , Antineoplastic Agents/therapeutic use , Doxorubicin/analogs & derivatives , Sarcoma, Experimental/drug therapy , Acrylamides/chemical synthesis , Acrylamides/pharmacokinetics , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Caproates/chemical synthesis , Caproates/pharmacokinetics , Caproates/therapeutic use , Cell Line, Tumor , Doxorubicin/chemical synthesis , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Drug Delivery Systems , Mice , Nanomedicine/methods , Polymerization
3.
Ecotoxicol Environ Saf ; 185: 109666, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31542645

ABSTRACT

Perfluoroalkyl acids (PFAAs) are a type of persistent organic pollutants that are widely distributed in multiple environmental media and organisms and have a teratogenic effect on and toxicity to animals and humans. The residual levels of seventeen PFAAs in the tissues of two regular consumption fish species, Culter erythropterus and Aristichthys nobilis in Lake Chaohu were measured by a high-performance liquid chromatograph - mass spectrometer (HPLC-MS). The distributions of PFAAs and the effect of the lipid contents were analyzed, and the health risks of typical PFAAs were evaluated. The results showed that perfluorohexanoic acid (PFHxA) was the predominant contaminant (80.50 ±â€¯58.31 ng/g and 19.17 ±â€¯12.57 ng/g wet weight, ww), followed by perfluorooctanesulfonic acid (PFOS) (55.02 ±â€¯34.82 and 14.79 ±â€¯6.24 ng/g, ww) in both fish. The level of total PFAAs was the highest in the liver tissues of Culter erythropterus (359.87 ng/g, ww) and the lowest in the kidney tissues in A. nobilis (10.06 ng/g, ww). Due to the higher trophic level of C. erythropteru, the total PFAA concentrations were significantly higher in all tissues than those in A. nobilis. Liver muscle ratio of C. erythropteru was the highest, indicating the most accumulation in the liver. The concentrations of PFAAs in fish tissues were influenced by the lipid content, resulting in a difference between the lipid-normalized concentrations and the wet weight concentrations of the PFAAs. The non-carcinogenic risks of PFOS were higher than those of PFOA through the ingestion of C. erythropterus and A. nobilis. Both the carcinogenic and non-carcinogenic risks of C. erythropterus were greater than those of A. nobilis, and fish tissue intake could cause an increasing of risks up to 60%, indicating that long-term and large amount ingestion of carnivorous fish and related tissues with higher trophic level, such as C. erythropterus should be avoided.


Subject(s)
Alkanesulfonic Acids/toxicity , Caproates/toxicity , Cyprinidae/metabolism , Environmental Monitoring/methods , Fluorocarbons/toxicity , Lakes/chemistry , Water Pollutants, Chemical/toxicity , Alkanesulfonic Acids/pharmacokinetics , Animals , Caproates/pharmacokinetics , China , Fluorocarbons/pharmacokinetics , Food Chain , Humans , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Seafood/analysis , Species Specificity , Tissue Distribution , Water Pollutants, Chemical/pharmacokinetics
4.
J Neuroimmune Pharmacol ; 14(3): 391-400, 2019 09.
Article in English | MEDLINE | ID: mdl-31209775

ABSTRACT

HIV-associated neurocognitive disorders (HAND) have been linked to dysregulation of glutamate metabolism in the central nervous system (CNS) culminating in elevated extracellular glutamate and disrupted glutamatergic neurotransmission. Increased glutamate synthesis via upregulation of glutaminase (GLS) activity in brain immune cells has been identified as one potential source of excess glutamate in HAND. However, direct evidence for this hypothesis in an animal model is lacking, and the viability of GLS as a drug target has not been explored. In this brief report, we demonstrate that GLS inhibition with the glutamine analogue 6-diazo-5-oxo-L-norleucine (DON) can reverse cognitive impairment in the EcoHIV-infected mouse model of HAND. However, due to peripheral toxicity DON is not amenable to clinical use in a chronic disease such as HAND. We thus tested JHU083, a novel, brain penetrant DON prodrug predicted to exhibit improved tolerability. Systemic administration of JHU083 reversed cognitive impairment in EcoHIV-infected mice similarly to DON, and simultaneously normalized EcoHIV-induced increases in cerebrospinal fluid (CSF) glutamate and GLS activity in microglia-enriched brain CD11b + cells without observed toxicity. These studies support the mechanistic involvement of elevated microglial GLS activity in HAND pathogenesis, and identify JHU083 as a potential treatment option. Graphical Abstract Please provide Graphical Abstract caption.Glutamine Antagonist JHU083 Normalizes Aberrant Glutamate Production and Cognitive Deficits in the EcoHIV Murine Model of HIV-Associated Neurocognitive Disorders .


Subject(s)
AIDS Dementia Complex , Azo Compounds/therapeutic use , Caproates/therapeutic use , Cognition Disorders/drug therapy , Glutamates/biosynthesis , Glutamine/antagonists & inhibitors , Prodrugs/therapeutic use , Animals , Azo Compounds/pharmacokinetics , CD11b Antigen/analysis , Caproates/pharmacokinetics , Cognition Disorders/cerebrospinal fluid , Cognition Disorders/etiology , Cognition Disorders/virology , Conditioning, Classical/drug effects , Fear , Glutamates/cerebrospinal fluid , HIV-1/genetics , HIV-1/pathogenicity , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/pathogenicity , Male , Maze Learning/drug effects , Memory, Short-Term/drug effects , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Norleucine/analogs & derivatives , Norleucine/therapeutic use , Prodrugs/pharmacokinetics , Reassortant Viruses/genetics , Reassortant Viruses/pathogenicity , Spatial Learning/drug effects
5.
ACS Appl Mater Interfaces ; 10(48): 41012-41018, 2018 Dec 05.
Article in English | MEDLINE | ID: mdl-30403126

ABSTRACT

This study explored a new rosuvastatin calcium- and heparin-loaded poly(l-lactide- co-caprolactone) (PLCL) scaffold for covered stents for treating aneurysms. The mechanism of rosuvastatin-induced endothelialization via vascular endothelial growth factor (VEGF)-A elevation was further explored. Rosu50, Rosu75, Rosu100, and phosphate-buffered saline (PBS) nanofibrous scaffolds were fabricated by coaxial electrospinning and observed by electron microscopy. Anticoagulation and pro-endothelialization properties were tested. Sixteen rabbits were selected for an in vivo assay and underwent microsurgery to establish a carotid aneurysm model. The animals were treated with covered stents and followed for 4 months using digital subtraction angiography (DSA), electron microscopy, and histology. Rosuvastatin-treated human umbilical vein endothelial cell (HUVEC) viability, function, and VEGF-A modulation were further studied to elucidate the pro-endothelialization mechanism of rosuvastatin. Our study demonstrates that rosuvastatin and heparin can be incorporated into PLCL nanofibers via electrospinning. Rosu100 nanofiber scaffolds exhibited significant anticoagulation properties. The viability of HUVECs transferred to Rosu100 nanofiber scaffolds was increased significantly. In vivo, DSA revealed that the Rosu100 group had better outcomes than the PBS group. In addition, the Rosu100 stents induced more integrated endothelialization. Further study demonstrated that rosuvastatin promoted HUVEC viability and function in vitro. The effects of rosuvastatin may be attributed to an elevation in VEGF-A. We demonstrated that rosuvastatin- and heparin-loaded PLCL-covered stents show favorable anticoagulation and pro-endothelialization properties in vitro and in vivo in a rabbit aneurysm model. VEGF-A elevation played a crucial role in rosuvastatin-promoted endothelialization. This work provides an additional option for treating cerebral aneurysms with covered stents.


Subject(s)
Aneurysm , Carotid Arteries , Nanofibers/chemistry , Rosuvastatin Calcium , Stents , Vascular Endothelial Growth Factor A , Aneurysm/metabolism , Aneurysm/pathology , Aneurysm/surgery , Animals , Caproates/chemistry , Caproates/pharmacokinetics , Caproates/pharmacology , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Arteries/surgery , Heparin/chemistry , Heparin/pharmacokinetics , Heparin/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Lactones/chemistry , Lactones/pharmacokinetics , Lactones/pharmacology , Polyesters/chemistry , Polyesters/pharmacokinetics , Polyesters/pharmacology , Rabbits , Rosuvastatin Calcium/chemistry , Rosuvastatin Calcium/pharmacokinetics , Rosuvastatin Calcium/pharmacology , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/pharmacokinetics , Vascular Endothelial Growth Factor A/pharmacology
6.
J Toxicol Sci ; 42(3): 301-317, 2017.
Article in English | MEDLINE | ID: mdl-28496036

ABSTRACT

To examine the kinetics of low doses of perfluoro compounds (PFCs), we administered perfluorohexanoic acid (C6A), perfluorooctanoic acid (C8A), perfluorononanoic acid (C9A) and perfluorooctane sulfonate (C8S) with a single oral dose (50-100 µg/kg BW), and in drinking water at 1, 5, and 25 µg/L for one and three months to male rats; and examined the distribution in the brain, heart, liver, spleen, kidney, whole blood and serum. C6A was very rapidly absorbed, distributed and eliminated from the tissues with nearly the same tissue t1/2 of 2-3 hr. Considering serum Vd, and the tissue delivery, C6A was mainly in the serum with the lowest delivery to the brain; and no tissue accumulation was observed in the chronic studies as estimated from the single dose study. For the other PFCs, the body seemed to be an assortment of independent one-compartments with a longer elimination t1/2 for the liver than the serum. The concentration ratio of liver/serum increased gradually from C0 to a steady state. The high binding capacity of plasma protein may be the reason for the unusual kinetics, with only a very small fraction of free PFCs moving gradually to the liver. Although the tissue specific distribution was time dependent and different among the PFCs, the Vd and ke of each tissue were constant throughout the study. The possibility of extremely high C6A accumulation in the human brain and liver was suggested, by comparing the steady state tissue concentration of this study with the human data reported by Pérez et al. (2013).


Subject(s)
Alkanesulfonic Acids/administration & dosage , Alkanesulfonic Acids/toxicity , Caproates/pharmacokinetics , Caproates/toxicity , Caprylates/pharmacokinetics , Caprylates/toxicity , Fluorocarbons/administration & dosage , Fluorocarbons/pharmacokinetics , Fluorocarbons/toxicity , Animals , Brain/metabolism , Fatty Acids , Liver/metabolism , Male , Myocardium/metabolism , Organ Specificity , Rats, Wistar , Time Factors , Tissue Distribution
7.
Anticancer Agents Med Chem ; 17(8): 1144-1152, 2017.
Article in English | MEDLINE | ID: mdl-27924732

ABSTRACT

BACKGROUND: Lactam cyclized alpha-melanocyte stimulating hormone (α-MSH) analogues exhibit high stability and affinity for the MC1-R receptors over expressed in melanoma cells. Recently, we reported a novel 99mTc-HYNIC-cycMSH4-13 analogue with the HYNIC chelator directly attached to the lactam cyclized ring. OBJECTIVE: In this study we proposed the introduction of a 6-aminohexanoic acid (Ahx) linker between the HYNIC chelator and lactam cyclized peptide cycMSH4-13 to reduce steric hindrance and improve the melanoma targeting and imaging proprieties of the radiolabeled peptide. METHOD: HYNIC-Ahx-cycMSH4-13 peptide was synthesized on an automated peptide synthesizer and displayed an IC50 of 0.3 nM using B16/F1 cells. The 99mTc/tricine radiolabeled peptide was examined for radiochemical purity, stability and cell binding. In vivo, biodistribution and planar gamma imaging studies were performed in B16/F1 melanoma tumor bearing C57BK mice. RESULTS: 99mTc-HYNIC-Ahx-cycMSH4-13 was obtained with a radiochemical purity > 95%, was stable up to 24 h at room temperature and exhibited high binding and rapid internalization in B16/F1 cells. In vivo biodistribution studies showed a tumor uptake of 4.92 ± 0.92 % ID/g and 2.78 ± 1.48 % ID/g at 2 h and 4 h post injection, respectively. Whole-body clearance was rapid through urinary excretion. The melanoma tumors were clearly visualized by planar gamma imaging. CONCLUSION: 99mTc-HYNIC-Ahx-cycMSH4-13 was shown radiochemically stability and exhibited rapid and selective uptake in melanoma cells and tumors. Imaging studies yielded promising preclinical results, warranting further evaluation of 99mTc-HYNIC-cycMSH analogs as melanoma specific imaging agents.


Subject(s)
Caproates/pharmacokinetics , Neoplasms, Experimental/diagnosis , Organotechnetium Compounds/pharmacokinetics , Peptide Fragments/pharmacokinetics , alpha-MSH/pharmacokinetics , Animals , Caproates/chemistry , Mice , Molecular Structure , Organotechnetium Compounds/chemistry , Peptide Fragments/chemistry , Tissue Distribution , Tumor Cells, Cultured , alpha-MSH/chemistry
8.
AAPS PharmSciTech ; 18(6): 1917-1924, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27858252

ABSTRACT

We report the successful implementation of a novel melt co-extrusion process to fabricate ca. 1 µm diameter fibers of poly(caprolactone) (PCL) containing the antifungal compound clotrimazole in concentrations between 4 and 8 wt%. The process involves co-extrusion of a clotrimazole-loaded PCL along with poly(ethylene oxide) (PEO) as a co-feed, with subsequent removal of PEO to isolate PCL-clotrimazole fibers. In vitro tests of the clotrimazole-containing fibers against the fungus Aspergillus fumigatus, Candida albicans, and Trichophyton mentagrophytes strains demonstrated good antifungal activity which was maintained for more than 3 weeks. An in vivo study using a mouse model showed the lowest tissue fungal burden for PCL-clotrimazole when compared to a PCL-only patch and untreated controls. Comparative studies were conducted with clotrimazole-containing PCL fibers fabricated by electrospinning. Our data showed that the co-extruded, clotrimazole-containing fibers maintain activity for longer times vs. electrospun samples. This, coupled with the much higher throughput of the co-extrusion process vs. electrospinning, renders this new approach very attractive for the fabrication of drug-releasing polymer fibers.


Subject(s)
Antifungal Agents/chemistry , Chemistry, Pharmaceutical/methods , Nanofibers/chemistry , Polymers/chemistry , Animals , Antifungal Agents/pharmacokinetics , Candida albicans/drug effects , Candida albicans/metabolism , Caproates/chemistry , Caproates/pharmacokinetics , Clotrimazole/chemistry , Clotrimazole/pharmacokinetics , Drug Compounding , Drug Liberation/drug effects , Drug Liberation/physiology , Lactones/chemistry , Lactones/pharmacokinetics , Mice , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Polymers/pharmacokinetics , Trichophyton/drug effects , Trichophyton/metabolism
9.
Curr Environ Health Rep ; 2(1): 33-40, 2015 Mar.
Article in English | MEDLINE | ID: mdl-26231240

ABSTRACT

Due to their oleophobic and hydrophobic properties and stability, perfluorinated compounds (PFCs) are used in many applications, particularly as greaseproofing agents for food contact. However, PFCs 8-carbons in length or greater (C8-PFCs) have raised concerns regarding environmental biopersistence, bioaccumulation in humans, and potent toxicity that have resulted in their gradual phase-out for food contact use. Industry has replaced C8-PFCs with shorter-chained C6-based greaseproofing agents, which are intended to have the same favorable physicochemical properties without the problematic toxicological effects in humans and wildlife. Compared with the large body of data available for C8 compounds, however, the available database on toxicity and exposure to the C6 compounds is fairly limited. This article summarizes the information in this database, focusing on aspects of human exposure and potential health risks associated with two types of C6 PFCs found in food packaging: perfluorohexanoic acid (PFHxA) and 6-2 fluorotelomer alcohol (C6-FTOH).


Subject(s)
Caproates , Fluorocarbons , Food Packaging/standards , Surface-Active Agents , Animals , Caproates/chemistry , Caproates/pharmacokinetics , Caproates/toxicity , Environmental Pollutants/analysis , Fluorocarbons/chemistry , Fluorocarbons/pharmacokinetics , Fluorocarbons/toxicity , Food Packaging/methods , Humans , Mice , Models, Animal , Molecular Structure , Rats , Surface-Active Agents/chemistry , Surface-Active Agents/pharmacokinetics , Surface-Active Agents/toxicity
10.
J Pharm Biomed Anal ; 102: 417-24, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25459941

ABSTRACT

Alzheimer's disease (AD) is the most common cause of dementia. Since no causative treatment is available, new therapeutic options are utmost needed. Several pirinixic acid derivatives, including MH84 (2-((4,6-bis(4-(trifluoromethyl)phenethoxy)pyrimidin-2-yl)thio)hexanoic acid), have shown promising in vitro results as γ-secretase modulators as well as PPARγ activators as potential pharmacological compounds against AD. Using a newly developed and validated sensitive LC-MS (APCI-qTOF mass analyzer) method, the pharmacokinetic and long-term accumulating properties as well as the blood-brain-barrier permeability of MH84 were evaluated in a preclinical animal study. MH84 was administered to mice by oral gavage with a dose of 12 mg/kg. Nine time points from 0.5 to 48 h with 6 animals per point were investigated. Additionally 6 animals were fed daily, for 21 days with an identical dose to determine possible long-term accumulation in plasma and brain tissue. The sample preparation was performed by a liquid-liquid extraction on Extrelut(®) columns whereas the LC separation was operated on a MulthoHigh 100 RP 18-5 µ column (125 × 4 mm) using an isocratic mobile phase of formic acid (0.1% (v/v))-methanol mixture (11:89 (v/v)) at a flow rate of 1 ml/min. The validation confirmed the new LC-MS method to be precise, accurate and reliable. After oral application, Cmax and Tmax of unmetabolized MH84 was determined to be 10.90 µg/ml and 3h in plasma. In brain tissue a constant level of 300 to maximum 320.64 ng/g was found after 1.5-6h. Daily gavage for 21 days did not lead to a long-term drug accumulation in the brain. The efficacy of the obtained MH84 levels needs to be investigated in further preclinical pharmacodynamic animal studies.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Caproates/pharmacokinetics , PPAR gamma/agonists , PPAR gamma/metabolism , Pyrimidines/pharmacokinetics , Tandem Mass Spectrometry/methods , Animals , Caproates/chemistry , Chromatography, Liquid/methods , Mass Spectrometry/methods , Mice , Pyrimidines/chemistry , Swine
11.
J Med Chem ; 56(6): 2568-80, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23472952

ABSTRACT

Recent efforts to identify treatments for myocardial ischemia reperfusion injury have resulted in the discovery of a novel series of highly potent α,α-disubstituted amino acid-based arginase inhibitors. The lead candidate, (R)-2-amino-6-borono-2-(2-(piperidin-1-yl)ethyl)hexanoic acid, compound 9, inhibits human arginases I and II with IC50s of 223 and 509 nM, respectively, and is active in a recombinant cellular assay overexpressing human arginase I (CHO cells). It is 28% orally bioavailable and significantly reduces the infarct size in a rat model of myocardial ischemia/reperfusion injury. Herein, we report the design, synthesis, and structure-activity relationships (SAR) for this novel series of inhibitors along with pharmacokinetic and in vivo efficacy data for compound 9 and X-ray crystallography data for selected lead compounds cocrystallized with arginases I and II.


Subject(s)
Amino Acids/chemistry , Amino Acids/pharmacology , Arginase/antagonists & inhibitors , Boron Compounds/chemistry , Boron Compounds/pharmacology , Caproates/chemistry , Caproates/pharmacology , Drug Discovery , Myocardial Reperfusion Injury/drug therapy , Amino Acids/pharmacokinetics , Amino Acids/therapeutic use , Animals , Arginase/chemistry , Boron Compounds/pharmacokinetics , Boron Compounds/therapeutic use , CHO Cells , Caproates/pharmacokinetics , Caproates/therapeutic use , Cricetinae , Cricetulus , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Male , Models, Molecular , Protein Conformation , Rats , Structure-Activity Relationship
12.
Toxicol Sci ; 127(1): 216-24, 2012 May.
Article in English | MEDLINE | ID: mdl-22302310

ABSTRACT

Perfluoroalkyl acids (PFAAs), specifically perfluorinated sulfonates and carboxylates, are synthetic substances known for their chemical stability, resistance to degradation, and potential to biomagnify in food chains. The toxicological and biological effects of PFAAs in avian species are not well characterized, although there is some evidence to suggest that they can impact neurodevelopment and hatching success. Our laboratory recently reported significant effects of perfluorohexane sulfonate (PFHxS) and perfluorohexanoate (PFHxA) on messenger RNA (mRNA) levels of thyroid hormone (TH)-responsive genes in chicken embryonic neuronal cells. In this study, we determined in ovo effects of PFHxS and PFHxA exposure (maximum dose = 38,000 and 9700 ng/g egg, respectively) on embryonic death, developmental endpoints, tissue accumulation, mRNA expression in liver and cerebral cortex, and plasma TH levels. Pipping success was reduced to 63% at the highest dose of PFHxS; no effects were observed for PFHxA. PFHxS exposure (38,000 ng/g) decreased tarsus length and embryo mass. PFHxS and PFHxA accumulated in the three tissue compartments analyzed as follows: yolk sac > liver > cerebral cortex. Type II and type III 5'-deiodinases (D2 and D3) and cytochrome P450 3A37 mRNA levels were induced in liver tissue of chicken embryos exposed to PFHxS, whereas D2, neurogranin (RC3), and octamer motif binding factor 1 mRNA levels were upregulated in cerebral cortex. Plasma TH levels were reduced in a concentration-dependent manner following PFHxS exposure; PFHxA had no effect. This in ovo study successfully validated previous in vitro results concerning the modulation of TH-responsive genes and identified adverse effects associated with TH homeostasis in response to PFHxS treatment.


Subject(s)
Caproates/toxicity , Chick Embryo/drug effects , Embryonic Development/drug effects , Fluorocarbons/toxicity , Gene Expression Regulation, Developmental/drug effects , Liver/drug effects , Sulfonic Acids/toxicity , Thyroxine/blood , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Biomarkers/metabolism , Caproates/pharmacokinetics , Cerebral Cortex/drug effects , Cerebral Cortex/embryology , Cytochrome P450 Family 3 , Embryo Loss/chemically induced , Fluorocarbons/pharmacokinetics , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Liver/embryology , RNA, Messenger/metabolism , Sulfonic Acids/pharmacokinetics , Yolk Sac/drug effects , Yolk Sac/embryology
13.
Biochim Biophys Acta ; 1814(10): 1394-403, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21782982

ABSTRACT

The stromelysin-1 catalytic domain(83-247) (SCD) is stable for at least 16 h at pHs 6.0-8.4. At pHs 5.0 and 9.0 there is exponential irreversible denaturation with half lives of 38 and 68 min respectively. At pHs 4.5 and 10.0 irreversible denaturation is biphasic. At 25°C, C-terminal truncation of stromelysin-1 decreases the stability of the stromelysin-1 catalytic domain at pH values >8.4 and <6.0. We describe the conversion of the carboxylate group of (ßR)-ß-[[[(1S)-1-[[[(1S)-2-Methoxy-1-phenylethyl]amino]carbonyl]-2,2-dimethylpropyl]amino]carbonyl]-2-methyl-[1,1'-biphenyl]-4-hexanoic acid (UK-370106-COOH) a potent inhibitor of the metalloprotease stromelysin-1 to a glyoxal group (UK-370106-CO(13)CHO). At pH 5.5-6.5 the glyoxal inhibitor is a potent inhibitor of stromelysin-1 (K(i)=~1µM). The aldehyde carbon of the glyoxal inhibitor was enriched with carbon-13 and using carbon-13 NMR we show that the glyoxal aldehyde carbon is fully hydrated when it is in aqueous solutions (90.4ppm) and also when it is bound to SCD (~92.0ppm). We conclude that the hemiacetal hydroxyl groups of the glyoxal inhibitor are not ionised when the glyoxal inhibitor is bound to SCD. The free enzyme pK(a) values associated with inhibitor binding were 5.9 and 6.2. The formation and breakdown of the signal at ~92ppm due to the bound UK-370106-CO(13)CHO inhibitor depends on pK(a) values of 5.8 and 7.8 respectively. No strong hydrogen bonds are present in free SCD or in SCD-inhibitor complexes. We conclude that the inhibitor glyoxal group is not directly coordinated to the catalytic zinc atom of SCD.


Subject(s)
Catalytic Domain , Glyoxal/metabolism , Matrix Metalloproteinase 3/chemistry , Matrix Metalloproteinase 3/metabolism , Caproates/chemistry , Caproates/metabolism , Caproates/pharmacokinetics , Catalytic Domain/physiology , Enzyme Inhibitors/metabolism , Enzyme Stability , Glyoxal/chemistry , Glyoxal/pharmacokinetics , Humans , Hydrogen-Ion Concentration , Matrix Metalloproteinase Inhibitors , Models, Biological , Nuclear Magnetic Resonance, Biomolecular , Polycyclic Compounds , Protein Binding , Protons , Temperature , Valine/analogs & derivatives , Valine/chemistry , Valine/metabolism , Valine/pharmacokinetics
14.
Drug Chem Toxicol ; 34(4): 341-6, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21696261

ABSTRACT

Excretion patterns and rates of ammonium perfluorohexanoate (APFHx) after administration of a single and multiple (14 days) oral dose(s) at 50 mg/kg to male and female mice and rats were examined. The test substance was [(14)C]-labeled APFHx. After a single oral administration, total excretion was rapid, with mean recoveries of over 90% of the dose at 24 hours after administration, irrespective of gender or species. The major route of elimination was via the urine (means of percentage recovery between 73.0 and 90.2% of the dose), followed by the feces (means of percentage recovery between 7.0 and 15.5% of the dose). Elimination via expired air was negligible. For the multiple dose tests, multiple (13 daily doses) oral administration of APFHx was followed by a single oral administration of [(14)C]-APFHx. Excretion was rapid, with mean recoveries of over 90% of the administered dose (mean values >95% of the ultimately recovered material) at 24 hours after dosing, irrespective of gender or species. The major route of elimination was via the urine (means of percentage recovery between 77.8 and 83.4% of the dose), followed by the feces (means of percentage recovery between 9.6 and 12.9% of the dose).


Subject(s)
Caproates/pharmacokinetics , Fluorocarbons/pharmacokinetics , Administration, Oral , Animals , Caproates/urine , Dose-Response Relationship, Drug , Female , Fluorocarbons/urine , Male , Metabolic Clearance Rate , Mice , Mice, Inbred Strains , Rats , Rats, Sprague-Dawley , Sex Factors , Species Specificity , Time Factors , Tissue Distribution
15.
Toxicology ; 283(1): 55-62, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21349313

ABSTRACT

The absorption, tissue distribution, elimination, and metabolism of [1-¹4C]-PFHx in rats and mice dosed orally at 2 or 100 mg/kg was evaluated following a single dose or after 14 consecutive doses. Absorption was rapid in rats as evidenced by a short time to maximum concentration (C(max)) of 30 min in male rats and 15 min in female rats at both the 2 and 100mg/kg dose level. The plasma elimination half-life was somewhat longer in males (1.5-1.7 h) than in females (0.5-0.7 h). Absorption in the mouse was also rapid with the maximum plasma concentration occurring between 15 and 30 min after dosing. The maximum concentration was not appreciably different between male and female mice (8 µg equiv./g at 2 mg/kg; ~350 µg equiv./g at 100 mg/kg). The primary route of elimination was via the urine. PFHx was not metabolized in rat or mouse hepatocytes, nor were any metabolites observed after oral dosing in either rodent species. Essentially 100% of the dose was eliminated in urine within 24 h demonstrating that PFHx is readily absorbed and bioavailability approaches 100%, even at a dose as high as 100 mg/kg. The route and extent of elimination was unchanged after 14 days of daily dosing. Tissues were collected at three time points (rat: 0.5, 2, and 24 h; mice: 0.25, 1, and 24 h) after dosing to investigate the tissue clearance kinetics of PFHx following a single dose at 2 or 100 mg/kg. In all tissues except skin, PFHx was not quantifiable 24 h after dosing in both sexes of the two species.


Subject(s)
Caproates/pharmacokinetics , Fluorocarbons/pharmacokinetics , Animals , Area Under Curve , Caproates/blood , Caproates/urine , Carbon Radioisotopes , Female , Fluorocarbons/blood , Fluorocarbons/urine , Guinea Pigs , Half-Life , Hepatocytes/metabolism , Intestinal Absorption , Male , Mice , Rats , Rats, Sprague-Dawley , Tissue Distribution
16.
Lipids ; 45(11): 997-1009, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20811782

ABSTRACT

Fatty acid synthase (FAS) is responsible for the de novo synthesis of palmitate and stearate. This enzyme is activated by insulin and T(3), and inhibited by fatty acids. In this study, we show that insulin and T(3) have an inducing effect on FAS enzymatic activity, which is synergetic when both hormones are present. Octanoate and hexanoate specifically inhibit this hormonal effect. A similar inhibitory effect is observed at the level of protein expression. Transient transfections in HepG2 cells revealed that hexanoate inhibits, at least in part, FAS at a transcriptional level targeting the T(3) response element (TRE) on the FAS promoter. The effect of C6 on FAS expression cannot be attributed to a modification of insulin receptor activation or to a decrease in T(3) entry in the cells. Using bromo-hexanoate, we determined that hexanoate needs to undergo a transformation in order to have an effect. When incubating cells with triglyceride-hexanoate or carnitine-hexanoate, no effect on the enzymatic activity induced by insulin and T(3) is observed. A similar result was obtained when cells were incubated with betulinic acid, an inhibitor of the diacylglycerol acyltransferase. However, the incubation of cells with Triacsin C, a general inhibitor of acyl-CoA synthetases, completely reversed the inhibitory effect of hexanoate. Our results suggest that in hepatic cells, hexanoate needs to be activated into a CoA derivative in order to inhibit the insulin and T(3)-induced FAS expression. This effect is partially transcriptional, targeting the TRE on the FAS promoter.


Subject(s)
Caproates/pharmacology , Fatty Acid Synthases/biosynthesis , Insulin/pharmacology , Triiodothyronine/pharmacology , Animals , Caproates/pharmacokinetics , Cells, Cultured , Chick Embryo , Down-Regulation/drug effects , Down-Regulation/genetics , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Fatty Acids/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Hormone Antagonists/pharmacology , Humans , Insulin Antagonists/pharmacology , Promoter Regions, Genetic/drug effects , Triiodothyronine/antagonists & inhibitors
17.
Reprod Toxicol ; 27(3-4): 400-406, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19429410

ABSTRACT

The toxicokinetics of perfluorohexanoic acid (PFHxA) and nonafluoro-1-butanesulfonic acid (PFBS) were evaluated in Sprague-Dawley rats and cynomolgus monkeys. Systemic exposure to PFHxA was lower than for PFBS following single equivalent intravenous or oral (rat only) doses. Serum clearance was more rapid for PFHxA than for PFBS. In rats, exposure to PFHxA and PFBS was up to 8-fold (intravenous) and 4-fold (oral) higher for males than females and serum clearance of PFHxA and PFBS was more rapid in females than males; however, there was no appreciable difference in the extent or rate of urinary elimination between compounds or genders. There were no apparent differences between genders in the serum half-life for PFHxA following 26 days of repeated oral dosing in rats; exposure decreased upon repeated dosing.


Subject(s)
Caproates/pharmacokinetics , Caproates/toxicity , Fluorocarbons/pharmacokinetics , Fluorocarbons/toxicity , Sulfonic Acids/pharmacokinetics , Sulfonic Acids/toxicity , Administration, Oral , Animals , Area Under Curve , Blood Chemical Analysis , Caproates/administration & dosage , Dose-Response Relationship, Drug , Female , Fluorocarbons/administration & dosage , Half-Life , Injections, Intravenous , Macaca fascicularis , Male , Metabolic Clearance Rate , Rats , Rats, Sprague-Dawley , Species Specificity , Sulfonic Acids/administration & dosage , Urinalysis
18.
J Pharmacol Exp Ther ; 330(1): 191-7, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19349522

ABSTRACT

In humans and rats, a synergistic blood pressure reduction was observed when the fibrate gemcabene (CI-1027) was coadministered with the angiotensin-converting enzyme inhibitor quinapril. In a quinapril (3 mg/kg) pharmacokinetic rat study, there was a 40% decrease in urinary excretion and a 53% increase in plasma area under the curve from 0 to 24 h of the active metabolite quinaprilat when coadministered with gemcabene (30 mg/kg). This observation revealed a possible transporter-mediated drug-drug interaction (DDI) between gemcabene and quinapril. This led to a series of studies investigating the underlying clearance mechanisms associated with these compounds intended to elucidate renal transporter interactions between quinapril and gemcabene. In vitro transporter studies using human embryonic kidney 293 cells transfected with human or rat organic anion transporter 3 (hOAT3, rOat3) revealed that quinaprilat is a substrate in both species, with a K(m) value of 13.4 microM for hOAT3. Subsequent studies discovered that gemcabene inhibited quinaprilat uptake by hOAT3 and rOat3 at IC(50) values of 35 and 48 microM, respectively. Moreover, gemcabene acylglucuronide, the major metabolite of gemcabene glucuronidation, also inhibited hOAT3- and rOat3-mediated uptake of quinaprilat at IC(50) values of 197 and 133 microM, respectively. High plasma concentrations of gemcabene (>100 microM) achieved in humans and rats upon oral dosing corroborate with gemcabene inhibition of renal OAT3-mediated secretion of quinaprilat in vitro. This investigation established that a DDI between gemcabene and quinapril involving inhibition of renal transporters and subsequent elevation in plasma concentrations of quinaprilat is responsible for the apparent synergistic blood pressure reduction observed with these compounds.


Subject(s)
Caproates/metabolism , Kidney/metabolism , Organic Anion Transporters/physiology , Tetrahydroisoquinolines/metabolism , Animals , Caproates/blood , Caproates/pharmacokinetics , Cell Line , Drug Interactions/physiology , Drug Therapy, Combination , Humans , Male , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Independent/antagonists & inhibitors , Organic Anion Transporters, Sodium-Independent/metabolism , Organic Anion Transporters, Sodium-Independent/physiology , Quinapril , Rats , Rats, Inbred SHR , Tetrahydroisoquinolines/blood , Tetrahydroisoquinolines/pharmacokinetics
19.
Mol Nutr Food Res ; 51(3): 301-6, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17309117

ABSTRACT

Human metabolism of 2-ethylhexanoic acid (2-EHA), which is a known metabolite of important phthalates, was investigated using 2-EHA-contaminated food. The results of our studies reveal that the major catabolic pathway of 2-EHA in human is beta-oxidation. The dominant final urinary metabolite was identified and quantified as 3-oxo-2-ethylhexanoic acid (3-oxo-2-EHA), but only after immediate methylation of the extract from urine and prior to GC-MS analysis. Former studies without the precaution of immediate methylation had found 4-heptanone as the major metabolite, which is obviously an artifact arising from the decarboxylation of 3-oxo-2-EHA.


Subject(s)
Caproates/administration & dosage , Caproates/pharmacokinetics , Caproates/urine , Adult , Female , Food Contamination , Gas Chromatography-Mass Spectrometry , Humans , Kinetics , Magnetic Resonance Imaging , Male , Oxidation-Reduction
20.
Eur J Clin Nutr ; 61(5): 686-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17180156

ABSTRACT

OBJECTIVE: To determine whether exercise augments the improvements in fractional synthetic rate (FSR) of albumin observed with nutrition alone. DESIGN: Randomized crossover study. Each patient randomly participated in two protein metabolism kinetic studies using primed-constant infusion of (13C) leucine 2 h before, during and 2 h after hemodialysis. Plasma enrichments of (13C) leucine and (13C) ketoisocaproate were examined to determine the FSR of albumin. SETTING: General Clinical Research Center at Vanderbilt University Medical Center. SUBJECTS: Five chronic hemodialysis (CHD) patients. INTERVENTIONS: Intra-dialytic parenteral nutrition (IDPN) with or without exercise. RESULTS: Exercise performance during hemodialysis significantly improves the FSR of albumin beyond what is observed with IDPN alone (26.2+/-3.1% per day versus 17.7+/-1.9% per day, P<0.05). CONCLUSION: Exercise improves albumin fractional synthetic rate beyond what is observed with IDPN alone in the acute setting in CHD patients.


Subject(s)
Caproates/pharmacokinetics , Exercise/physiology , Kidney Failure, Chronic/therapy , Leucine/pharmacokinetics , Renal Dialysis , Serum Albumin/analysis , Albumins/metabolism , Carbon Isotopes , Cross-Over Studies , Female , Humans , Kidney Failure, Chronic/blood , Male , Middle Aged , Parenteral Nutrition
SELECTION OF CITATIONS
SEARCH DETAIL
...