Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 387(2): 214-225, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37643795

ABSTRACT

Hereditary angioedema (HAE) is a rare autosomal dominant disorder caused by a deficiency in functional C1 esterase inhibitor, a serpin family protein that blocks the activity of plasma kallikrein. Insufficient inhibition of plasma kallikrein results in the overproduction of bradykinin, a vasoactive inflammatory mediator that produces both pain and unpredictable swelling during HAE attacks, with potentially life-threatening consequences. We describe the generation of STAR-0215, a humanized IgG1 antibody with a long circulating half-life (t1/2) that potently inhibits plasma kallikrein activity, with a >1000-fold lower affinity for prekallikrein and no measurable inhibitory activity against other serine proteases. The high specificity and inhibitory effect of STAR-0215 is demonstrated through a unique allosteric mechanism involving N-terminal catalytic domain binding, destabilization of the activation domain, and reversion of the active site to the inactive zymogen state. The YTE (M252Y/S254T/T256E) modified fragment crystallizable (Fc) domain of STAR-0215 enhances pH-dependent neonatal Fc receptor binding, resulting in a prolonged t1/2 in vivo (∼34 days in cynomolgus monkeys) compared with antibodies without this modification. A single subcutaneous dose of STAR-0215 (≥100 mg) was predicted to be active in patients for 3 months or longer, based on simulations using a minimal physiologically based pharmacokinetic model. These data indicate that STAR-0215, a highly potent and specific antibody against plasma kallikrein with extended t1/2, is a potential agent for long-term preventative HAE therapy administered every 3 months or less frequently. SIGNIFICANCE STATEMENT: STAR-0215 is a YTE-modified immunoglobulin G1 monoclonal antibody with a novel binding mechanism that specifically and potently inhibits the enzymatic activity of plasma kallikrein and prevents the generation of bradykinin. It has been designed to be a long-lasting prophylactic treatment to prevent attacks of HAE and to decrease the burden of disease and the burden of treatment for people with HAE.

2.
J Pediatr ; 206: 56-65.e8, 2019 03.
Article in English | MEDLINE | ID: mdl-30471715

ABSTRACT

OBJECTIVE: To investigate recombinant human insulin-like growth factor 1 complexed with its binding protein (rhIGF-1/rhIGFBP-3) for the prevention of retinopathy of prematurity (ROP) and other complications of prematurity among extremely preterm infants. STUDY DESIGN: This phase 2 trial was conducted from September 2014 to March 2016. Infants born at a gestational age of 230/7 weeks to 276/7 weeks were randomly allocated to rhIGF-1/rhIGFBP-3 (250 µg/kg/ 24 hours, continuous intravenous infusion from <24 hours of birth to postmenstrual age 296/7 weeks) or standard neonatal care, with follow-up to a postmenstrual age of 404/7 weeks. Target exposure was ≥70% IGF-1 measurements within 28-109 µg/L and ≥70% intended therapy duration. The primary endpoint was maximum severity of ROP. Secondary endpoints included time to discharge from neonatal care, bronchopulmonary dysplasia, intraventricular hemorrhage, and growth measures. RESULTS: Overall, 61 infants were allocated to rhIGF-1/rhIGFBP-3, 60 to standard care (full analysis set); 24 of 61 treated infants achieved target exposure (evaluable set). rhIGF-1/rhIGFBP-3 did not decrease ROP severity or ROP occurrence. There was, however, a 53% decrease in severe bronchopulmonary dysplasia in the full analysis set (21.3% treated vs 44.9% standard care), and an 89% decrease in the evaluable set (4.8% vs 44.9%; P = .04 and P = .02, respectively) for severity distribution between groups. There was also a nonsignificant trend toward decrease in grades 3-4 intraventricular hemorrhage in the full analysis set (13.1% vs 23.3%) and in the evaluable set (8.3% vs 23.3%). Fatal serious adverse events were reported in 19.7% of treated infants (12/61) and 11.7% of control infants (7/60). No effect was observed on time to discharge from neonatal care/growth measures. CONCLUSIONS: rhIGF-1/rhIGFBP-3 did not affect development of ROP, but decreased the occurrence of severe bronchopulmonary dysplasia, with a nonsignificant decrease in grades 3-4 intraventricular hemorrhage. TRIAL REGISTRATION: ClinicalTrials.gov: NCT01096784.


Subject(s)
Cerebral Hemorrhage/prevention & control , Insulin-Like Growth Factor I/therapeutic use , Recombinant Proteins/therapeutic use , Retinopathy of Prematurity/prevention & control , Bronchopulmonary Dysplasia/prevention & control , Cerebral Hemorrhage/therapy , Female , Gestational Age , Humans , Infant, Extremely Premature , Infant, Newborn , Infant, Premature , Infusions, Intravenous , Insulin-Like Growth Factor Binding Protein 3/therapeutic use , Male , Retinopathy of Prematurity/mortality , Retinopathy of Prematurity/therapy , Severity of Illness Index , Treatment Outcome
3.
J Ocul Pharmacol Ther ; 34(1-2): 224-232, 2018.
Article in English | MEDLINE | ID: mdl-29267138

ABSTRACT

PURPOSE: Lifitegrast is approved in the United States for the treatment of dry eye disease (DED). We assessed lifitegrast's ocular distribution/pharmacokinetic profile in rabbits, and 14C-lifitegrast mass balance/excretion in dogs. METHODS: Female pigmented rabbits received a single topical ocular dose of lifitegrast (Formulation No. 1, n = 25; No. 2, n = 25) per eye twice daily (target, 1.75 mg/eye/dose). Blood/ocular tissues were collected on day 5. Beagle dogs received single intravenous (n = 10; target, 3 mg, 262 µCi/animal) and ocular (n = 8, target, 3 mg, 30 µCi/eye) doses of 14C-lifitegrast (∼8 weeks between doses). Blood, excreta, and cage rinse/wipes were collected. Concentrations were measured by mass spectrometry/liquid scintillation counting. Pharmacokinetic analyses (noncompartmental) included maximum concentration (Cmax), time to Cmax (tmax), and area under the concentration-time curve from 0 to 8 h (AUC0-8). RESULTS: In rabbits, lifitegrast Cmax and AUC0-8 were similar between formulations. Cmax was highest in ocular anterior segment tissues: 5,190-14,200 ng/g [conjunctiva (palpebral/bulbar), cornea, anterior sclera]. Posterior segment tissues had lower concentrations (0-826 ng/g). AUC0-8 followed a similar trend. Plasma concentrations were low (Cmax <18 ng/mL). Tissue/plasma tmax was ∼0.25-1 h. In dogs, after intravenous/ocular doses, 14C-lifitegrast was eliminated primarily through feces. Excreted radioactivity was mainly unchanged lifitegrast. CONCLUSIONS: High exposure of lifitegrast in rabbit ocular anterior segment tissues and low exposure in posterior segment tissues/plasma suggests that lifitegrast reaches target tissues for DED treatment, with low potential for off-target systemic/ocular effects. Excretion of unchanged 14C-lifitegrast suggests minimal drug metabolism in vivo. This is consistent with lifitegrast clinical trial efficacy/safety data.


Subject(s)
Cornea/drug effects , Dry Eye Syndromes/drug therapy , Ophthalmic Solutions/pharmacokinetics , Phenylalanine/analogs & derivatives , Sulfones/pharmacokinetics , Administration, Topical , Animals , Cornea/pathology , Dogs , Dry Eye Syndromes/pathology , Female , Ophthalmic Solutions/administration & dosage , Phenylalanine/administration & dosage , Phenylalanine/pharmacokinetics , Rabbits , Sulfones/administration & dosage
4.
Int J Mol Sci ; 18(12)2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29194406

ABSTRACT

Mucopolysaccharidosis III type A (MPS IIIA; Sanfilippo syndrome), a genetic lysosomal disorder causing a deficiency of heparan N-sulfatase (HNS), leads to progressive cognitive decline from an early age. An effective enzyme replacement therapy (ERT) for MPS IIIA requires central nervous system (CNS) biodistribution. Recombinant human heparan N-sulfatase (rhHNS), an investigatory ERT for MPS IIIA, has been formulated for intrathecal (IT) administration since intravenous (IV) administration cannot cross the blood brain barrier (BBB) in sufficient amounts to have a therapeutic effect. In this study, systemic and CNS distribution of rhHNS in cynomolgus monkeys following IV and IT administration was evaluated by quantitation of rhHNS in serum, cerebral spinal fluid (CSF) and various tissues, and positron emission tomography (PET) imaging of live animals. Following IV administration, rhHNS levels were low to non-detectable in the CSF, and systemic clearance was rapid (≤2 h). With IT administration, rhHNS was observable in CNS tissues in ≤1 h, with varying Tmax (1-24 h). Appreciable systemic distribution was observed up to 7 days. This provides evidence that in this animal model, intrathecal administration of rhHNS delivers the replacement enzyme to therapeutically relevant tissues for the treatment of Sanfilippo Syndrome type A. Penetration into grey matter and cortex was 3-4 times greater than concentrations in white matter and deeper parenchymal regions, suggesting some limitations of this ERT strategy.


Subject(s)
Central Nervous System/chemistry , Sulfatases/administration & dosage , Sulfatases/pharmacokinetics , Administration, Intravenous , Animals , Central Nervous System/diagnostic imaging , Disease Models, Animal , Humans , Injections, Spinal , Macaca fascicularis , Male , Mucopolysaccharidosis III/drug therapy , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Tissue Distribution
5.
Pediatr Res ; 81(3): 504-510, 2017 03.
Article in English | MEDLINE | ID: mdl-27870826

ABSTRACT

BACKGROUND: rhIGF-1/rhIGFBP-3 is being investigated for prevention of retinopathy of prematurity in extremely preterm infants. METHODS: A population pharmacokinetic model was developed using data from phase I/II (Sections A-C) trials of rhIGF-1/rhIGFBP-3 and additional studies in preterm infants to predict optimal dosing to establish/maintain serum IGF-1 within physiological intrauterine levels. In Section D of the phase II study, infants (gestational age (GA) (wk+d) 23+0 to 27+6) were randomized to rhIGF-1/rhIGFBP-3, administered at the model-predicted dose of 250 µg/kg/d continuous i.v. infusion up to postmenstrual age (PMA) 29 wk+6 d or standard of care. An interim pharmacokinetic analysis was performed for the first 10 treated infants to verify dosing. RESULTS: Serum IGF-1 data were reviewed for 10 treated/9 control infants. Duration of therapy in treated infants ranged 1-34.5 d. At baseline (before infusion and <24 h from birth), mean (SD) IGF-1 was 19.2 (8.0) µg/l (treated) and 15.4 (4.7) µg/l (controls). Mean (SD) IGF-1 increased to 45.9 (19.6) µg/l at 12 h in treated infants, and remained within target levels for all subsequent timepoints. For treated infants, 88.8% of the IGF-1 measurements were within target levels (controls, 11.1%). CONCLUSION: Through the reported work, we determined appropriate rhIGF-1/rhIGFBP-3 dosing to achieve physiological intrauterine serum IGF-1 levels in extremely preterm infants.


Subject(s)
Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor I/therapeutic use , Recombinant Proteins/blood , Recombinant Proteins/therapeutic use , Blood Glucose , Computer Simulation , Female , Gestational Age , Humans , Infant, Extremely Premature , Infant, Newborn , Infant, Premature , Male , Reference Values , Time Factors
6.
PLoS One ; 11(10): e0164765, 2016.
Article in English | MEDLINE | ID: mdl-27764180

ABSTRACT

Enzyme replacement therapy with intravenous idursulfase (recombinant iduronate-2-sulfatase) is approved for the treatment of Hunter syndrome. Intravenous administration does not, however, treat the neurological manifestations, due to its low central nervous system bioavailability. Using intrathecal-lumbar administration, iduronate-2-sulfatase is delivered directly to the central nervous system. This study investigates the central nervous system biodistribution of intrathecal-lumbar administered iduronate-2-sulfatase in cynomolgus monkeys. Twelve monkeys were administered iduronate-2-sulfatase in one 30 mg intrathecal-lumbar injection. Brain, spinal cord, liver, and kidneys were collected for iduronate-2-sulfatase concentration (measured by an enzyme linked immunosorbent assay) and enzyme activity measurement (via a method utilizing 4-methylumbelliferyl-α-iduronate-2-sulfate) at 1, 2, 5, 12, 24, and 48 hours following administration. The tissue enzyme linked immunosorbent assay confirmed iduronate-2-sulfatase uptake to the brain, spinal cord, kidneys, and liver in a time-dependent manner. In spinal cord and brain, iduronate-2-sulfatase appeared as early as 1 hour following administration, and peak concentrations were observed at ~2 and ~5 hours. Iduronate-2-sulfatase appeared in liver and kidneys 1 hour post intrathecal-lumbar dose with peak concentrations between 5 and 24 hours. Liver iduronate-2-sulfatase concentration was approximately 10-fold higher than kidney. The iduronate-2-sulfatase localization and enzyme activity in the central nervous system, following intrathecal administration, demonstrates that intrathecal-lumbar treatment with iduronate-2-sulfatase may be considered for further investigation as a treatment for Hunter syndrome patients with neurocognitive impairment.


Subject(s)
Enzyme Replacement Therapy , Iduronate Sulfatase/administration & dosage , Mucopolysaccharidosis II/drug therapy , Animals , Brain/enzymology , Drug Evaluation, Preclinical , Female , Humans , Iduronate Sulfatase/pharmacokinetics , Injections, Spinal , Kidney/enzymology , Liver/enzymology , Macaca fascicularis , Male , Spinal Cord/enzymology , Time Factors , Tissue Distribution
7.
Mol Genet Metab ; 118(3): 198-205, 2016 07.
Article in English | MEDLINE | ID: mdl-27211612

ABSTRACT

OBJECTIVE: This was an open-label, phase 1/2 dose-escalation, safety trial of intrathecal recombinant human heparan-N-sulfatase (rhHNS) administered via intrathecal drug delivery device (IDDD) for treating mucopolysaccharidosis IIIA (NCT01155778). STUDY DESIGN: Twelve patients received 10, 45, or 90mg of rhHNS via IDDD once monthly for a total of 6 doses. Primary endpoints included adverse events (AEs) and anti-rhHNS antibodies. Secondary endpoints included standardized neurocognitive assessments, cortical gray matter volume, and pharmacokinetic/pharmacodynamic analyses. RESULTS: All patients experienced treatment-emergent AEs; most of mild-to-moderate severity. Seven patients reported a total of 10 serious AEs (SAEs), all but one due to hospitalization to revise a nonfunctioning IDDD. No SAEs were considered related to rhHNS. Anti-rhHNS antibodies were detected in the serum of 6 patients and in the cerebrospinal fluid (CSF) of 2 of these. CSF heparan sulfate levels were elevated at baseline and there were sustained declines in all tested patients following the first rhHNS dose. No impact of anti-rhHNS antibodies on any pharmacodynamic or safety parameters was evident. 4 of 12 patients showed a decline in developmental quotient, 6 were stable, and 2 patients had only a single data point. No dose group showed a clearly different response pattern. CONCLUSIONS: rhHNS administration via IDDD appeared generally safe and well tolerated. Treatment resulted in consistent declines in CSF heparan sulfate, suggesting in vivo activity in the relevant anatomical compartment. Results of this small study should be interpreted with caution. Future studies are required to assess the potential clinical benefits of rhHNS and to test improved IDDD models.


Subject(s)
Heparitin Sulfate/cerebrospinal fluid , Mucopolysaccharidosis III/drug therapy , Sulfatases/administration & dosage , Adolescent , Antibodies/blood , Antibodies/cerebrospinal fluid , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Humans , Injections, Spinal/instrumentation , Male , Mucopolysaccharidosis III/cerebrospinal fluid , Sulfatases/adverse effects , Sulfatases/immunology , Treatment Outcome , Young Adult
8.
PLoS One ; 10(4): e0122453, 2015.
Article in English | MEDLINE | ID: mdl-25836678

ABSTRACT

Intravenous enzyme replacement therapy with iduronate-2-sulfatase is an approved treatment for Hunter syndrome, however, conventional intravenous delivery cannot treat the neurologic manifestations of the disease due to its limited central nervous system penetration. Intrathecal administration of iduronate-2-sulfatase for delivery to the central nervous system is currently under investigation. The objective of this study was to evaluate the pharmacokinetics of idursulfase in the central nervous system of cynomolgus monkeys (Macaca fasicularis) after intravenous and intrathecal administration. Twenty-seven monkeys, treatment-naïve to enzyme replacement therapy, were placed into 4 groups according to body weight: Group 1 was administered 0.5 mg/kg idursulfase intravenously, Groups 2-4 were administered an intrathecal formulation (1-, 10-, and 30-mg doses). Blood samples and cerebrospinal fluid (sampled at the cisterna magna or lumbar level) were collected at the same time points for 72 hours post dosing. Following intravenous administration, a high maximum serum concentration and rapid distribution of iduronate-2-sulfatase out of the central compartment were observed (elimination half-life: 4.3 hours). Iduronate-2-sulfatase exposure in the cerebrospinal fluid was limited, suggesting intravenous administration provided minimal penetration of the blood-brain barrier. Following intrathecal administration, a high maximum observed concentration was immediately noted and elimination half-life ranged between 7.8-10 hours and 5.9-6.7 hours (cisterna magna and lumbar sampling, respectively). Cerebrospinal fluid pharmacokinetic profiles at different doses of iduronate-2-sulfatase were similar and the dose/exposure relationship was proportional. After intrathecal administration, movement of iduronate-2-sulfatase from cerebrospinal fluid to serum was observed (systemic bioavailability was 40-83%). The clear penetration of iduronate-2-sulfatase into the cerebrospinal fluid and the dose response suggest that intrathecal delivery of iduronate-2-sulfatase may be suitable for treating the central nervous system manifestations associated with Hunter syndrome.


Subject(s)
Iduronate Sulfatase/administration & dosage , Iduronate Sulfatase/pharmacokinetics , Administration, Intravenous , Animals , Biological Availability , Enzyme Replacement Therapy , Female , Humans , Iduronate Sulfatase/cerebrospinal fluid , Injections, Spinal , Macaca fascicularis , Male , Mucopolysaccharidosis II/drug therapy , Mucopolysaccharidosis II/enzymology , Recombinant Proteins/administration & dosage , Recombinant Proteins/cerebrospinal fluid , Recombinant Proteins/pharmacokinetics
9.
Toxicol Lett ; 210(3): 353-9, 2012 May 05.
Article in English | MEDLINE | ID: mdl-22366341

ABSTRACT

Styrene is one of the most important industrial intermediates consumed in the world and is mainly used as a monomer for reinforced plastics and rubber. Styrene has been found to be hepatotoxic and pneumotoxic in humans and experimental animals. The toxicity of styrene is suggested to be metabolism-dependent. Styrene-7,8-oxide has been considered as the major metabolite responsible for styrene-induced cytotoxicity. The objective of the study was to investigate the correlation between cytotoxicity of styrene and chemical and biochemical properties of the vinyl group of styrene by development of structure activity relationships (SAR). 4-Fluorostyrene, 4-chlorostyrene and 4-bromostyrene were selected for the SAR study. Cytotoxicity of styrene and the halogenated styrene derivatives with an order of 4-bromostyrene>4-chlorostyrene>4-fluorostyrene≈styrene was observed in CYP2E1 transgenic cells. Similar orders in the efficiency of the metabolism of styrene and the halogenated styrene analogues to their oxides and in the electrophilicity of the corresponding oxides were observed. Additionally, the order of the potency of cellular glutathione depletion and the degree of protein adduction induced by styrene and the halogenated styrenes were consistent with that of their cytotoxicities. The wild-type cells were less susceptible to the toxicity of the corresponding model compounds than CYP2E1 cells. The present study provided insight into the roles of the biochemical and chemical properties of styrene in its cytotoxicity.


Subject(s)
Cytochrome P-450 CYP2E1/physiology , Hydrocarbons, Halogenated/toxicity , Styrenes/toxicity , Cell Line , Cells, Cultured , Cysteamine/metabolism , Glutathione/metabolism , Humans , Protein Binding , Structure-Activity Relationship
10.
Arterioscler Thromb Vasc Biol ; 32(4): 902-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22282355

ABSTRACT

OBJECTIVE: We investigated the stability, pharmacokinetic, and pharmacodynamic profile of the 2(nd) generation anti-von Willeband factor aptamer ARC15105. METHODS AND RESULTS: Platelet plug formation was measured by collagen/adenosine diphosphate-induced closure time with the platelet function analyzer-100 and platelet aggregation by multiple electrode aggregometry. Platelet adhesion was measured on denuded porcine aortas and in a flow chamber. Aptamer stability was assessed by incubation in nuclease rich human, monkey, and rat serum for up to 72 hours. Pharmacokinetic and pharmacodynamic profiles were tested in cynomolgus monkeys after IV and SC administration. The median IC(100) and IC(50) to prolong collagen/adenosine diphosphate-induced closure timewere 27 nmol/L and 12 nmol/L, respectively. ARC15105 (1.3 µmol/L) completely inhibited ristocetin-induced platelet aggregation in whole blood (P<0.001), but also diminished collagen, ADP, arachidonic acid, and thrombin receptor activating peptide-induced platelet aggregation to some extent (P<0.05). ARC15105 (40 nmol/L) decreased platelet adhesion by >90% on denuded porcine aortas (P<0.001), which was comparable to the degree of inhibition obtained with abciximab. ARC15105 (100 nmol/L) also inhibited platelet adhesion to collagen under arterial shear in a flow chamber by >90% (P<0.001). The IV and SC terminal half-lives in cynomolgus monkeys were 67 and 65 hours, respectively, and the SC bioavailability was ≈98%. Allometric scaling estimates the human T(1/2) would be ≈217 hours. Pharmacodynamic analysis confirms that ARC15105 inhibits von Willeband factor activity >90% in blood samples taken 300 hours after a 20 mg/kg IV or SC dose in monkeys. CONCLUSIONS: The potency, pharmacokinetic profile, and SC bioavailability of ARC15105 support its clinical investigation for chronic inhibition of von Willeband factor -mediated platelet activation.


Subject(s)
Aptamers, Nucleotide/therapeutic use , Aptamers, Peptide/pharmacology , Blood Platelets/drug effects , Myocardial Infarction/blood , Platelet Activation/drug effects , Platelet Adhesiveness/drug effects , Platelet Aggregation Inhibitors/pharmacology , von Willebrand Factor/antagonists & inhibitors , Aged , Animals , Aptamers, Nucleotide/administration & dosage , Aptamers, Nucleotide/pharmacokinetics , Aptamers, Peptide/administration & dosage , Aptamers, Peptide/pharmacokinetics , Austria , Biological Availability , Blood Platelets/metabolism , Boston , Case-Control Studies , Collagen/metabolism , Cross-Sectional Studies , Dose-Response Relationship, Drug , Drug Stability , Female , Half-Life , Humans , Injections, Intravenous , Injections, Subcutaneous , Macaca fascicularis , Male , Middle Aged , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Function Tests , Protein Binding , Quebec , Rats , Swine , von Willebrand Factor/metabolism
11.
Chem Biol Interact ; 186(3): 323-30, 2010 Aug 05.
Article in English | MEDLINE | ID: mdl-20470765

ABSTRACT

Styrene is one of the most important industrial intermediates consumed in the world. Human exposure to styrene occurs mainly in the reinforced plastics industry, particularly in developing countries. Styrene has been found to be hepatotoxic and pneumotoxic in humans and animals. The biochemical mechanisms of styrene-induced toxicities remain unknown. Albumin and hemoglobin adduction derived from styrene oxide, a major reactive metabolite of styrene, has been reported in blood samples obtained from styrene-exposed workers. The objectives of the current study focused on cellular protein covalent binding of styrene metabolite and its correlation with cytotoxicity induced by styrene. We found that radioactivity was bound to cellular proteins obtained from mouse airway trees after incubation with (14)C-styrene. Microsomal incubation studies showed that the observed protein covalent binding required the metabolic activation of styrene. The observed radioactivity binding in protein samples obtained from the cultured airways and microsomal incubations was significantly suppressed by co-incubation with disulfiram, a CYP2E1 inhibitor, although disulfiram apparently did not show a protective effect against the cytotoxicity of styrene. A 2-fold increase in radioactivity bound to cellular proteins was detected in cells stably transfected with CYP2E1 compared to the wild-type cells after (14)C-styrene exposure. With the polyclonal antibody developed in our lab, we detected cellular protein adduction derived from styrene oxide at cysteinyl residues in cells treated with styrene. Competitive immunoblot studies confirmed the modification of cysteine residues by styrene oxide. Cell culture studies showed that the styrene-induced protein modification and cell death increased with the increasing concentration of styrene exposure. In conclusion, we detected cellular protein covalent modification by styrene oxide in microsomal incubations, cultured cells, and mouse airways after exposure to styrene and found a good correlation between styrene-induced cytotoxicity and styrene oxide-derived cellular protein adduction.


Subject(s)
Cytotoxins/adverse effects , Cytotoxins/metabolism , Proteins/metabolism , Styrene/adverse effects , Styrene/metabolism , Animals , Cell Line , Cytochrome P-450 CYP2E1/metabolism , Epoxy Compounds/adverse effects , Epoxy Compounds/metabolism , Humans , Male , Mice , Protein Binding
12.
Toxicology ; 226(2-3): 99-106, 2006 Sep 21.
Article in English | MEDLINE | ID: mdl-16872732

ABSTRACT

Styrene has been found to be toxic to the respiratory system, and the toxicity of styrene is metabolism-dependent. CYP2E1 is suggested to be one of the cytochrome P450 enzymes responsible for the bioactivation of styrene. Our work focused on the roles of CYP2E1 and epoxide, a metabolite of styrene epoxidation, in the cytotoxicity of styrene. Styrene was found to be more toxic to h2E1 cells than to the wild type, while there was no difference found when styrene oxide was administered. Both soluble and microsomal epoxide hydrolase inhibitors dramatically enhanced styrene toxicity. Glutathione and glutathione ethyl ester showed protection against styrene cytotoxicity. Cytotoxicity of a selection of styrene analogues, such as ethylbenzene, vinylcyclohexane, and ethylcyclohexane, was assessed to determine if unsaturation is required for styrene toxicity. Ethylbenzene and vinylcyclohexane were found to be as toxic as styrene to h2E1 cells, whereas little toxicity of ethylcyclohexane to h2E1 cells was observed. This indicates the importance of vinyl group of styrene in its cytotoxicity, but saturation of the vinyl group does not necessarily eliminate styrene toxicity. An N-acetylcysteine conjugate derived from styrene oxide was identified by LC/MS/MS in the sample obtained from the incubation of h2E1 cell lysate with styrene in the presence of N-acetylcysteine. Formation of the N-acetylcysteine conjugate was found to be NADPH-dependent. These studies provided strong evidence in support of toxic role of styrene epoxide metabolite in styrene toxicity.


Subject(s)
Cytochrome P-450 CYP2E1/genetics , Styrene/metabolism , Styrene/toxicity , Acetylcysteine/chemical synthesis , Animals , Animals, Genetically Modified , Biotransformation , Cell Survival/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/antagonists & inhibitors , Epoxy Compounds/metabolism , Epoxy Compounds/toxicity , Glutathione/metabolism , Humans , Styrenes/chemical synthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...