Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
J Chromatogr A ; 1621: 461050, 2020 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-32265050

RESUMO

Consumption of ciguatoxin-contaminated seafood can lead to ciguatera poisoning (CP). The diagnosis of CP in humans is based on the clinical symptoms after eating the fish from tropical or subtropical areas because no confirmatory clinical tests are available. One of the challenges for ciguatoxin analysis is their extremely low but toxicologically relevant concentration in biological samples. We previously reported a method using acetonitrile to precipitate proteins and extract the ciguatoxins simultaneously in whole blood samples from animals for toxin quantification by N2A cell-based assay. However, a test method for unambiguous confirmation of exposure of marine animals or humans to ciguatoxins is still needed. In the present study, we adopted the acetonitrile extraction method and added sample clean-up in the sample preparation for the determination of Pacific ciguatoxins CTX1B (aka P-CTX-1), 52-epi-54-deoxyCTX1B (aka P-CTX-2), and CTX3C (aka P-CTX-3C) in blood plasma by LC-MS/MS. We investigated sample clean-up, LC mobile phases, LC solvent programming, and settings of the two mass spectrometers (4000 Q TRAP and AB SCIEX Triple Quad 5500) in order to improve the ability to detect the Pacific ciguatoxins at ppt level. Rat blood plasma was used for the method development. Average recoveries of the three toxins in the rat plasma samples ranged from 90% to 116% with relative standard deviations of less than 15%. The method detection limits were still not low enough for the determination of the Pacific ciguatoxins in individual blood samples from Hawaiian monk seals with the two LC-MS systems. The methods were applied to a pooled sample of blood plasma collected from Hawaiian monk seals for confirmation of toxin exposure. This study will benefit monitoring of Pacific ciguatoxins in marine mammals and potentially humans by LC-MS/MS.


Assuntos
Cromatografia Líquida/métodos , Ciguatoxinas/sangue , Espectrometria de Massas em Tandem/métodos , Acetonitrilas/química , Animais , Limite de Detecção , Ratos , Focas Verdadeiras/sangue , Solventes
2.
J Chromatogr A ; 1416: 22-30, 2015 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-26381568

RESUMO

Toxins produced by harmful algae are associated with detrimental health effects and mass mortalities of marine mammals. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) is generally used to confirm the presence of algal toxins in marine mammals. Sample preparation and LC-MS/MS methods for the determination of three diarrhetic shellfish poisoning (DSP) toxins (okadaic acid, OA; dinophysistoxin-1, DTX1; dinophysistoxin-2, DTX2) and pectenotoxin-2 (PTX2) in bottlenose dolphin (Tursiops truncatus) urine and tissue samples were evaluated using spike-and-recovery tests. Sample clean-up with either reversed-phase silica or polymeric solid-phase extraction (SPE) reduced interference of sample matrices and improved toxin recoveries, with polymeric SPE showing higher sample loading capacity. LC separation on Xbridge C18 columns using acetonitrile/water gradient elutions with ammonia as the additive was chosen for its high detectivity and sensitivity in the MS detection of DSP toxins in negative ion mode. The retention times of OA, DTX1, and DTX2, separated as negative ions, increased with LC column temperature while the retention time of PTX2, separated as the neutral molecule, was weakly affected. At the same column temperature, retention times of OA, DTX1, and DTX2 gradually increased as the mobile phases aged while the retention time of PTX2 remained unchanged; higher column temperatures resulted in a greater increase in the retention time of each DSP toxin with mobile phase aging. Average recoveries of the 4 toxins in bottlenose dolphin samples ranged from 80% to 130% with relative standard deviations of less than 15% using the LC mobile phases prepared within one week at a column temperature of 30°C or 40°C. The preferred column temperature was 30°C, as the retention times of DSP toxins were less affected by mobile phase aging at this temperature. The limit of detection of each toxin analyzed in bottlenose dolphin samples was 2.8 ng/g or less in tissue samples and 0.7 ng/ml or less in urine.


Assuntos
Golfinho Nariz-de-Garrafa/metabolismo , Cromatografia Líquida/métodos , Diarreia/induzido quimicamente , Furanos/análise , Toxinas Marinhas/análise , Piranos/análise , Intoxicação por Frutos do Mar/metabolismo , Espectrometria de Massas em Tandem/métodos , Animais , Macrolídeos , Ácido Okadáico/análise , Extração em Fase Sólida/métodos
3.
Chem Res Toxicol ; 27(7): 1166-75, 2014 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-24949875

RESUMO

Brevetoxins produced during algal blooms of the dinoflagellate Karenia are metabolized by shellfish into reduction, oxidation, and conjugation products. Brevetoxin metabolites comprising amino acid- and lipid conjugates account for a large proportion of the toxicity associated with the consumption of toxic shellfish. However, the disposition of these brevetoxin metabolites has not been established. Using intravenous exposure to C57BL/6 mice, we investigated the disposition in the body of three radiolabeled brevetoxin metabolites. Amino acid-brevetoxin conjugates represented by S-desoxy-BTX-B2 (cysteine-BTX-B) and lipid-brevetoxin conjugates represented by N-palmitoyl-S-desoxy-BTX-B2 were compared to dihydro-BTX-B. Tissue concentration profiles were unique to each of the brevetoxin metabolites tested, with dihydro-BTX-B being widely distributed to all tissues, S-desoxy-BTX-B2 concentrated in kidney, and N-palmitoyl-S-desoxy-BTX-B2 having the highest concentrations in spleen, liver, and lung. Elimination patterns were also unique: dihydro-BTX-B had a greater fecal versus urinary elimination, whereas urine was a more important elimination route for S-desoxy-BTX-B2, and N-palmitoyl-S-desoxy-BTX-B2 persisted in tissues and was eliminated equally in both urine and feces. The structures particular to each brevetoxin metabolite resulting from the reduction, amino acid conjugation, or fatty acid addition of BTX-B were likely responsible for these tissue-specific distributions and unique elimination patterns. These observed differences provide further insight into the contribution each brevetoxin metabolite class has to the observed potencies.


Assuntos
Cisteína/química , Lipídeos/química , Toxinas Marinhas/farmacocinética , Neurotoxinas/farmacocinética , Oxocinas/farmacocinética , Administração Intravenosa , Animais , Encéfalo/metabolismo , Sistema Digestório/metabolismo , Fezes/química , Rim/metabolismo , Pulmão/metabolismo , Masculino , Toxinas Marinhas/sangue , Toxinas Marinhas/química , Toxinas Marinhas/urina , Camundongos Endogâmicos C57BL , Músculos/metabolismo , Miocárdio/metabolismo , Neurotoxinas/sangue , Neurotoxinas/química , Neurotoxinas/urina , Oxocinas/sangue , Oxocinas/química , Oxocinas/urina , Baço/metabolismo , Testículo/metabolismo , Distribuição Tecidual
4.
Mar Drugs ; 12(3): 1185-207, 2014 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-24663110

RESUMO

Domoic acid epileptic disease is characterized by spontaneous recurrent seizures weeks to months after domoic acid exposure. The potential for this disease was first recognized in a human case study of temporal lobe epilepsy after the 1987 amnesic shellfish-poisoning event in Quebec, and was characterized as a chronic epileptic syndrome in California sea lions through investigation of a series of domoic acid poisoning cases between 1998 and 2006. The sea lion study provided a breadth of insight into clinical presentations, unusual behaviors, brain pathology, and epidemiology. A rat model that replicates key observations of the chronic epileptic syndrome in sea lions has been applied to identify the progression of the epileptic disease state, its relationship to behavioral manifestations, and to define the neural systems involved in these behavioral disorders. Here, we present the concept of domoic acid epileptic disease as a delayed manifestation of domoic acid poisoning and review the state of knowledge for this disease state in affected humans and sea lions. We discuss causative mechanisms and neural underpinnings of disease maturation revealed by the rat model to present the concept for olfactory origin of an epileptic disease; triggered in dendodendritic synapases of the olfactory bulb and maturing in the olfactory cortex. We conclude with updated information on populations at risk, medical diagnosis, treatment, and prognosis.


Assuntos
Doenças dos Animais/induzido quimicamente , Doenças dos Animais/fisiopatologia , Epilepsia/induzido quimicamente , Epilepsia/veterinária , Ácido Caínico/análogos & derivados , Toxinas Marinhas/intoxicação , Fármacos Neuromusculares Despolarizantes/intoxicação , Neurotoxinas/intoxicação , Leões-Marinhos/fisiologia , Convulsões/veterinária , Intoxicação por Frutos do Mar/fisiopatologia , Intoxicação por Frutos do Mar/veterinária , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/fisiologia , Amnésia/induzido quimicamente , Amnésia/psicologia , Doenças dos Animais/diagnóstico , Animais , Comportamento Animal/efeitos dos fármacos , Bivalves , Epilepsia/diagnóstico , Epilepsia do Lobo Temporal/induzido quimicamente , Epilepsia do Lobo Temporal/fisiopatologia , Feminino , Contaminação de Alimentos , Hipocampo/fisiopatologia , Humanos , Ácido Caínico/intoxicação , Masculino , Pessoa de Meia-Idade , Condutos Olfatórios/fisiopatologia , Ratos , Recidiva , Convulsões/induzido quimicamente , Convulsões/diagnóstico , Intoxicação por Frutos do Mar/diagnóstico
5.
J Zoo Wildl Med ; 44(2): 364-75, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23805555

RESUMO

In 2005 and 2006, the central west Florida coast experienced two intense Karenia brevis red tide events lasting from February 2005 through December 2005 and August 2006 through December 2006. Strandings of sea turtles were increased in the study area with 318 turtles (n = 174, 2005; n = 144, 2006) stranding between 1 January 2005 and 31 December 2006 compared to the 12-yr average of 43 +/- 23 turtles. Live turtles (n = 61) admitted for rehabilitation showed clinical signs including unresponsiveness, paresis, and circling. Testing of biological fluids and tissues for the presence of brevetoxin activity by enzyme-linked immunosorbent assay found toxin present in 93% (52 of 56) of live stranded sea turtles, and 98% (42 of 43) of dead stranded sea turtles tested. Serial plasma samples were taken from several live sea turtles during rehabilitation and toxin was cleared from the blood within 5-80 days postadmit depending upon the species tested. Among dead animals the highest brevetoxin levels were found in feces, stomach contents, and liver. The lack of significant pathological findings in the majority of animals necropsied supports toxin-related mortality.


Assuntos
Líquidos Corporais/química , Dinoflagellida/metabolismo , Eutrofização , Toxinas Marinhas/sangue , Oxocinas/sangue , Tartarugas/sangue , Animais , Feminino , Florida , Masculino , Toxinas Marinhas/química , Toxinas Marinhas/metabolismo , Oxocinas/química , Oxocinas/metabolismo , Fatores de Tempo
6.
Chem Res Toxicol ; 26(6): 868-77, 2013 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-23642029

RESUMO

Brevetoxin B (BTX-B), produced by dinoflagellates of the species Karenia, is a highly reactive molecule, due in part to an α,ß-unsaturated aldehyde group at the terminal side chain, leading to the production of metabolites in shellfish by reduction, oxidation, and conjugation. We have investigated in mice the blood elimination of three common bioactive brevetoxin metabolites found in shellfish, which have been semisynthesized from BTX-B in radioactive forms. BTX-B was reduced at C42 to yield [(3)H] dihydro-BTX-B. [(3)H] S-desoxy-BTX-B2 (cysteine brevetoxin B) was semisynthesized from BTX-B by the conjugation of cysteine at the C50 olefinic group then [(3)H] radiolabeled by C42 aldehyde reduction. [(14)C] N-Palmitoyl-S-desoxy-BTX-B2 was prepared using S-desoxy-BTX-B2 as the starting material with addition of the [(14)C] radiolabeled fatty acid via cysteine-amide linkage. The elimination of intravenously administered [(3)H] S-desoxy-BTX-B2, [(14)C] N-palmitoyl-S-desoxy-BTX-B2, or [(3)H] dihydro-BTX-B was measured in blood collected from C57BL/6 mice over a 48 h period. Each brevetoxin metabolite tested exhibited biexponential elimination kinetics and fit a two-compartment model of elimination that was applied to generate toxicokinetic parameters. The rate of transfer between the central compartment (i.e., blood) and the peripheral compartment (e.g., tissue) for each brevetoxin differed substantially, with dihydro-BTX-B exchanging rapidly with the peripheral compartment, S-desoxy-BTX-B2 eliminating rapidly from the central compartment, and N-palmitoyl-S-desoxy-BTX-B2 eliminating slowly from the central compartment. Toxicokinetic parameters were analyzed in the context of the unique structure of each brevetoxin metabolite resulting from a reduction, amino acid conjugation, or fatty acid addition to BTX-B.


Assuntos
Cisteína/sangue , Toxinas Marinhas/sangue , Toxinas Marinhas/metabolismo , Oxocinas/sangue , Oxocinas/metabolismo , Trítio/sangue , Animais , Cisteína/química , Cisteína/metabolismo , Cisteína/farmacocinética , Cinética , Dose Letal Mediana , Masculino , Toxinas Marinhas/farmacocinética , Toxinas Marinhas/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , Oxocinas/farmacocinética , Oxocinas/toxicidade , Toxicocinética , Trítio/química , Trítio/farmacocinética
7.
J Wildl Dis ; 49(2): 246-60, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23568900

RESUMO

Harmful algal bloom events caused by the dinoflagellate Karenia brevis occurred along the central west Florida, USA, coast from February 2005 through December 2005 and from August 2006 through December 2006. During these events, from 4 February 2005 through 28 November 2006, live, debilitated seabirds admitted for rehabilitation showed clinical signs that included disorientation, inability to stand, ataxia, and seizures. Testing of blood, biologic fluids, and tissues for brevetoxin by enzyme-linked immunosorbent assay found toxin present in 69% (n=95) of rehabilitating seabirds. Twelve of the 19 species of birds had evidence of brevetoxin exposure. Commonly affected species included Double-crested Cormorants (Phalacrocorax auritus), Brown Pelicans (Pelecanus occidentalis), Great Blue Herons (Ardea herodias), and Common Loons (Gavia immer). Serial blood and fecal samples taken from several live seabirds during rehabilitation showed that brevetoxin was cleared within 5-10 days after being admitted to the rehabilitation facility, depending on the species tested. Among seabirds that died or were euthanized, the highest brevetoxin concentrations were found in bile, stomach contents, and liver. Most dead birds had no significant pathologic findings at necropsy, thereby supporting brevetoxin-related mortality.


Assuntos
Doenças das Aves/epidemiologia , Dinoflagellida/patogenicidade , Surtos de Doenças/veterinária , Toxinas Marinhas/análise , Oxocinas/análise , Infecções Protozoárias em Animais/epidemiologia , Animais , Doenças das Aves/mortalidade , Doenças das Aves/patologia , Aves , Causas de Morte , Dinoflagellida/metabolismo , Feminino , Florida/epidemiologia , Masculino , Toxinas Marinhas/toxicidade , Oxocinas/toxicidade , Infecções Protozoárias em Animais/mortalidade , Infecções Protozoárias em Animais/patologia , Especificidade da Espécie
8.
Toxicol Sci ; 133(1): 133-43, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23457124

RESUMO

The harmful alga Pseudo-nitzschia sp. is the cause of human amnesic shellfish poisoning and the stranding of thousands of sea lions with seizures as a hallmark symptom. A human case study and epidemiological report of hundreds of stranded sea lions found individuals presenting months after recovery with a neurological disease similar to temporal lobe epilepsy. A rat model developed to establish and better predict how epileptic disease results from domoic acid poisoning demonstrated that a single episode of status epilepticus (SE), after a latent period, leads to a progressive state of spontaneous recurrent seizure (SRS) and expression of atypical aggressive behaviors. Structural damage associated with domoic acid-induced SE is prominent in olfactory pathways. Here, we examine structural damage in seven rats that progressed to epileptic disease. Diseased animals show progressive neuronal loss in the piriform cortex and degeneration of terminal fields in these layers and the posteromedial cortical amygdaloid nucleus. Animals that display aggressive behavior had additional neuronal damage to the anterior olfactory cortex. This study provides insight into the structural basis for the progression of domoic acid epileptic disease and relates to the California sea lion, where poisoned animals progress to a disease characterized by SRS and aggressive behaviors.


Assuntos
Agressão/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Ácido Caínico/análogos & derivados , Neurônios/efeitos dos fármacos , Convulsões/psicologia , Estado Epiléptico/psicologia , Animais , Encéfalo/patologia , Contagem de Células , Modelos Animais de Doenças , Ácido Caínico/intoxicação , Masculino , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Recidiva , Convulsões/induzido quimicamente , Convulsões/patologia , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/patologia
9.
Toxicon ; 64: 81-6, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23319077

RESUMO

Ciguatoxins are sodium channel activator toxins responsible for ciguatera fish poisoning. In this study, we determined the toxicokinetic parameters of the Pacific ciguatoxin P-CTX-1 in rats after an intravenous (iv) dose of 0.13 ng P-CTX-1 per g of body weight. The ciguatoxin activity was assessed over time in blood using the sensitive functional Neuro2a assay. The data were analyzed with a two-compartmental model. After exposure, the ciguatoxin activity exhibited a rapid (alpha half-life of 6 min) and extensive distribution into tissues (apparent steady state volume of distribution of 7.8 L). Ciguatoxin elimination from blood was slower with a beta half-life estimated at 35.5 h. The toxicokinetic parameters determined from this study were compared to data previously obtained after oral and intraperitoneal exposure of rats to 0.26 ng P-CTX-1 per g of body weight. Maximal bioavailability was determined by the area under the concentration curve, and was used to calculate the absolute P-CTX-1 bioavailabilities for oral and intraperitoneal routes of exposures of 39% and 75%, respectively.


Assuntos
Ciguatera/metabolismo , Ciguatoxinas/farmacocinética , Ciguatoxinas/toxicidade , Administração Oral , Animais , Área Sob a Curva , Disponibilidade Biológica , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ciguatera/induzido quimicamente , Ciguatoxinas/administração & dosagem , Meia-Vida , Injeções Intraperitoneais , Injeções Intravenosas , Masculino , Camundongos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/patologia , Ratos , Ratos Sprague-Dawley , Canais de Sódio/efeitos dos fármacos
10.
Toxicol Pathol ; 41(3): 454-69, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22821366

RESUMO

The amnesic shellfish toxin, domoic acid, interferes with glutamatergic pathways leading to neuronal damage, most notably causing memory loss and seizures. In this study, the authors utilized a recently developed rat model for domoic acid-induced epilepsy, an emerging disease appearing in California sea lions weeks to months after poisoning, to identify structural damage that may lead to a permanent epileptic state. Sprague Dawley rats were kindled with several low hourly intraperitoneal doses of domoic acid until a state of status epilepticus (SE) appears. This kindling approach has previously been shown to induce a permanent state of epileptic disease in 96% animals within 6 months. Three animals were selected for neurohistology a week after the initial SE. An amino cupric silver staining method using neutral red counterstain was used on every eighth 40 µm coronal section from each brain to highlight neural degeneration from the olfactory bulb through the brain stem. The most extensive damage was found in the olfactory bulb and related olfactory pathways, including the anterior/medial olfactory cortices, endopiriform nucleus, and entorhinal cortex. These findings indicate that damage to olfactory pathways is prominent in a rat model for domoic acid-induced chronic recurrent spontaneous seizures and aggressive behavior.


Assuntos
Ácido Caínico/análogos & derivados , Condutos Olfatórios/efeitos dos fármacos , Condutos Olfatórios/patologia , Coloração pela Prata/métodos , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/patologia , Agressão/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Cobre/química , Modelos Animais de Doenças , Histocitoquímica/métodos , Ácido Caínico/toxicidade , Masculino , Condutos Olfatórios/química , Ratos , Ratos Sprague-Dawley , Compostos de Prata/química
11.
Chem Res Toxicol ; 25(12): 2805-9, 2012 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-23134453

RESUMO

Domoic acid (DA) causes neurological effects in multiple species upon exposure, including status epilepticus in pregnant sea lions and an epileptic disease state that commonly develops in juveniles. This study aims to define brain toxicokinetic parameters in the pregnant rat in the larger context of maternal-fetal toxin transfer. Specifically, Sprague-Dawley rats were exposed to a low observable effect level of 1.0 mg DA/kg intravenously at gestational day 20, and plasma, brain, and cerebrospinal fluid (CSF) samples were taken at discrete time points over 24 h. Domoic acid concentrations were determined by a tandem LC/MS method recently optimized for brain tissue and CSF. Data showed that 6.6% of plasma DA reached the brain, 5.3% reached the CSF, and DA levels were nearly identical in both brain and CSF for 12 h, remaining above the threshold to activate isolated hippocampal neurons for 2 h. The calculated terminal half-life of CSF was 4 h, consistent with the time for complete CSF regeneration, suggesting that CSF acts as a mechanism to clear DA from the brain.


Assuntos
Encéfalo/metabolismo , Ácido Caínico/análogos & derivados , Toxinas Marinhas/farmacocinética , Neurotoxinas/farmacocinética , Animais , Feminino , Ácido Caínico/sangue , Ácido Caínico/líquido cefalorraquidiano , Ácido Caínico/farmacocinética , Toxinas Marinhas/sangue , Toxinas Marinhas/líquido cefalorraquidiano , Neurotoxinas/sangue , Neurotoxinas/líquido cefalorraquidiano , Gravidez , Ratos , Ratos Sprague-Dawley
12.
Sci Total Environ ; 427-428: 98-105, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22560748

RESUMO

Ciguatera in humans is typically caused by the consumption of reef fish that have accumulated Ciguatoxins (CTXs) in their flesh. Over a six month period, we captured 38 wild adult great barracuda (Sphyraena barracuda), a species commonly associated with ciguatera in The Bahamas. We sampled three tissues (i.e., muscle, liver, and blood) and analysed them for the presence of ciguatoxins using a functional in vitro N2A bioassay. Detectable concentrations of ciguatoxins found in the three tissue types ranged from 2.51 to 211.74pg C-CTX-1 equivalents/g. Blood and liver toxin concentrations were positively correlated (ρ=0.86, P=0.003), indicating that, for the first time, blood sampling provides a non-lethal method of detecting ciguatoxin in wild fish. Non-lethal blood sampling also presents opportunities to couple this approach with biotelemetry and biologging techniques that enable the study of fish distribution and movement. To demonstrate the potential for linking ciguatoxin occurrence with barracuda spatial ecology, we also present a proof-of-concept case study where blood samples were obtained from 20 fish before releasing them with acoustic transmitters and tracking them in the coastal waters using a fixed acoustic telemetry array covering 44km(2). Fish that tested positive for CTX may have smaller home ranges than non-toxic fish (median distance travelled, U=2.21, P=0.03). Results presented from this study may help identify high risk areas and source-sink dynamics of toxins, potentially reducing the incidence and human health risk of ciguatera fish poisoning. Moreover, development of the non-lethal sampling approach and measurement of ciguatera from blood provide future opportunities to understand the mechanistic relationship between toxins and the spatial ecology of a broad range of marine fish species.


Assuntos
Comportamento Animal , Coleta de Amostras Sanguíneas/métodos , Ciguatoxinas/sangue , Monitoramento Ambiental/métodos , Peixes/fisiologia , Telemetria/métodos , Acústica , Animais , Bahamas , Fígado/química , Músculos/química , Estações do Ano , Estatísticas não Paramétricas
13.
Toxicology ; 294(1): 36-41, 2012 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-22306965

RESUMO

Domoic acid (DA) is a potent neurotoxin that has both marine wildlife and human health impacts, including developmental effects during prenatal exposure in rodent models. However, little is known regarding DA toxicokinetics in the fetal unit during maternal-fetal transfer. Tissue distribution and toxicokinetics of DA were investigated in pregnant rats and their pups just prior to birth at gestational day 20. Pregnant Sprague Dawley rats were given an intravenous dose of 1.0 mg DA/kg and samples of maternal plasma, fetal plasma, placenta, amniotic fluid and fetal brain were taken at intervals over 24 h. Toxicokinetic parameters were determined using WinNonLin software analysis. Maternal plasma DA log concentration-time curves fit a two compartment pharmacokinetic profile, with alpha and beta half-lives of elimination of 26.9 and 297 min, respectively. Placenta had a C(max) of 752 ng/mL and a terminal half-life of 577 min. Maternal-fetal transfer between the plasma compartments was 31% with a fetal plasma C(max) of 86 ng/mL at 60 min and terminal half-life of 553 min. Amniotic fluid and fetal brain had overall averages of 27±12 ng/mL and 8.12 ng/g, respectively, and did not show evidence of elimination over 24 h. The longer fetal retention of DA, particularly in amniotic fluid, indicates that the fetus may be continually re-exposed during gestation, which could potentially lead to a disease state even at small exposure dose. This has implications for the California sea lions (Zalophus californianus), which exhibit an epilepsy-like disease that arises months after DA producing blooms.


Assuntos
Ácido Caínico/análogos & derivados , Líquido Amniótico/química , Animais , Encéfalo/embriologia , Química Encefálica , Feminino , Feto/química , Feto/efeitos dos fármacos , Feto/metabolismo , Meia-Vida , Ácido Caínico/análise , Ácido Caínico/sangue , Ácido Caínico/farmacocinética , Ácido Caínico/toxicidade , Troca Materno-Fetal/efeitos dos fármacos , Placenta/química , Gravidez , Ratos/embriologia , Ratos Sprague-Dawley , Leões-Marinhos
14.
Anal Chim Acta ; 715: 71-9, 2012 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-22244169

RESUMO

We previously reported a solid-phase extraction (SPE) method for determination of the neurotoxin domoic acid (DA) in both seawater and phytoplankton by liquid chromatography-tandem mass spectrometry (LC-MS/MS) with the purpose of sample desalting without DA pre-concentration. In the present study, we optimized the SPE procedure with seawater and phytoplankton samples directly acidified with aqueous formic acid without addition of organic solvents, which allowed sample desalting and also 20-fold pre-concentration of DA in seawater and phytoplankton samples. In order to reduce MS contamination, a diverter valve was installed between LC and MS to send the LC eluant to waste, except for the 6-min elution window bracketing the DA retention time, which was sent to the MS. Reduction of the MS turbo gas temperature also helped to maintain the long-term stability of MS signal. Recoveries exceeded 90% for the DA-negative seawater and the DA-positive cultured phytoplankton samples spiked with DA. The SPE method for DA extraction and sample clean-up in seawater was extended to mammalian fluids and tissues with modification in order to accommodate the fluid samples with limited available volumes and the tissue extracts in aqueous methanol. Recoveries of DA from DA-exposed laboratory mammalian samples (amniotic fluid, cerebrospinal fluid, plasma, placenta, and brain) were above 85%. Recoveries of DA from samples (urine, feces, intestinal contents, and gastric contents) collected from field stranded marine mammals showed large variations and were affected by the sample status. The optimized SPE-LC-MS method allows determination of DA at trace levels (low pg mL(-1)) in seawater with/without the presence of phytoplankton. The application of SPE clean-up to mammalian fluids and tissue extracts greatly reduced the LC column degradation and MS contamination, which allowed routine screening of marine mammalian samples for confirmation of DA exposure and determination of fluid and tissue DA concentrations in experimental laboratory animals.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Ácido Caínico/análogos & derivados , Toxinas Marinhas/análise , Fitoplâncton/química , Água do Mar/química , Extração em Fase Sólida/métodos , Espectrometria de Massas em Tandem/métodos , Líquido Amniótico/química , Animais , Líquidos Corporais/química , Golfinhos/urina , Fezes/química , Feminino , Ácido Caínico/análise , Ratos , Ratos Sprague-Dawley , Leões-Marinhos/urina , Sensibilidade e Especificidade , Baleias/urina
15.
Physiol Behav ; 105(2): 315-20, 2012 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-21875611

RESUMO

Domoic acid (DA), a naturally occurring environmental toxin, has been observed to induce status epilepticus in humans, sea lions and pelicans. In a recent Sprague Dawley rat model, domoic acid dosing induced a state of status epilepticus which, after a symptom-free latent period without further dosing, progressed to recurrent spontaneous seizures, a hallmark of epilepsy. Certain individuals in this study also developed unusual behavioral changes, in particular an atypical aggression towards conspecifics. In this report we characterized the progression of aggressive behaviors after DA-induced status epilepticus and explored the relationship between aggressive behavior and recurrent spontaneous seizures. Experimental studies in this laboratory rat model are particularly relevant to California sea lions (Zapholus californianus), which show a spectrum of both epileptic and unusual behaviors, including aggression towards conspecifics in rehabilitation facilities, weeks to months after suspected exposure to domoic acid in the wild.


Assuntos
Agressão/fisiologia , Ácido Caínico/análogos & derivados , Neurotoxinas/toxicidade , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/fisiopatologia , Agressão/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Emoções/fisiologia , Ácido Caínico/toxicidade , Masculino , Ratos , Ratos Sprague-Dawley , Estatísticas não Paramétricas , Estresse Psicológico/fisiopatologia , Fatores de Tempo
16.
Environ Sci Technol ; 45(12): 5403-9, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21591690

RESUMO

Ciguatoxins are potent algal neurotoxins that concentrate in fish preyed upon by the critically endangered Hawaiian monk seal (Monachus schauinslandi). The only report for Hawaiian monk seal exposure to ciguatoxins occurred during a 1978 mortality event when two seal liver extracts tested positive by mouse bioassay. Ciguatoxins were thus proposed as a potential threat to the Hawaiian monk seal population. To reinvestigate monk seal exposure to ciguatoxins we utilized more selective detection methods, the Neuro-2A cytotoxicity assay, to quantify ciguatoxin activity and an analytical method LC-MS/MS to confirm the molecular structure. Tissue analysis from dead stranded animals revealed ciguatoxin activity in brain, liver, and muscle, whereas analysis of blood samples from 55 free-ranging animals revealed detectable levels of ciguatoxin activity (0.43 to 5.49 pg/mL P-CTX-1 equiv) in 19% of the animals. Bioassay-guided LC fractionation of two monk seal liver extracts identified several ciguatoxin-like peaks of activity including a peak corresponding to the P-CTX-3C which was confirmed present by LC-MS/MS. In conclusion, this work provides first confirmation that Hawaiian monk seals are exposed to significant levels of ciguatoxins and first evidence of transfer of ciguatoxin to marine mammals. This threat could pose management challenges for this endangered marine mammal species.


Assuntos
Ciguatoxinas/metabolismo , Monitoramento Ambiental , Geografia , Focas Verdadeiras/metabolismo , Animais , Encéfalo/metabolismo , Fracionamento Químico , Cromatografia Líquida de Alta Pressão , Ciguatoxinas/sangue , Ciguatoxinas/química , Havaí , Fígado/metabolismo , Espectrometria de Massas , Camundongos , Músculos/metabolismo , Especificidade de Órgãos , Focas Verdadeiras/sangue
17.
Toxicology ; 284(1-3): 1-6, 2011 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-21349314

RESUMO

Ciguatoxins are voltage-gated selective algal toxins responsible for ciguatera fish poisoning. In this study we evaluate the toxicokinetics of one of the most common ciguatoxins found in the Pacific, the P-CTX-1, in rat after an oral or intraperitoneal (ip) dose of 0.26 µg/kg body weight. We report levels of ciguatoxin activity assessed over time in blood, urine and feces, and at 4 days in liver, muscle and brain, using the functional in vitro N2A cytotoxicity assay. Following exposure, the ciguatoxin activity exhibited a rapid systemic absorption that was followed by a bi-exponential decline, and data best fit a two-compartment model analysis. Maximum blood concentrations were reached at 1.97 and 0.43 h after the oral and ip dose, respectively. Ciguatoxin elimination from blood was slow with terminal half lives (t(½)ß) estimated at 82 h for oral and 112 h for ip dosing. Ciguatoxin activity remained in liver, muscle and brain 96 h after ip and oral administration. While smaller amounts appeared in the urine, the main excretion route was feces, with peak rates reaching > 10 pg P-CTX-1 equivalents/h in both routes of administration. Assay guided fractionation showed the presence in the feces and liver of peaks of activity corresponding to the P-CTX-1 and to other less polar metabolites. In conclusion, biologically active ciguatoxins are detectable in blood, liver, muscle and brain, and continued to be excreted in urine and feces 4 days following exposure. Blood, as well as urine and feces may be useful matrices for low-invasive testing methods for ciguatera clinical cases.


Assuntos
Ciguatoxinas/farmacocinética , Ciguatoxinas/toxicidade , Administração Oral , Animais , Ciguatera/induzido quimicamente , Ciguatera/metabolismo , Ciguatoxinas/administração & dosagem , Enguias , Injeções Intraperitoneais , Masculino , Farmacocinética , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual/efeitos dos fármacos , Distribuição Tecidual/fisiologia
18.
Chem Res Toxicol ; 24(1): 54-64, 2011 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-21142195

RESUMO

Brevetoxins are neurotoxins produced by marine dinoflagellates, primarily Karenia brevis, and can cause intoxication and even mortality of marine species, affect human health through the consumption of brevetoxin-contaminated shellfish, and effect respiratory irritation through aerosol exposure at coastal areas. Brevetoxin-A and brevetoxin-B, the major brevetoxins produced in algae, are metabolized to a series of amino acid and peptide-related derivatives in shellfish through the reactions of the amino acid residue cysteine with an α,ß-unsaturated aldehyde group. In this paper, covalent interactions between brevetoxin and proteins were investigated using brevetoxin-B and human serum albumin (HSA) as a model. It is demonstrated that both noncovalent and covalent interactions can occur between brevetoxin-B and HSA with in vitro experiments. Covalent adducts of brevetoxin-B and HSA were generated under physiological conditions and reduced with sodium borohydride based on the reaction conditions of single amino acid residues with brevetoxin-B. LC/MS analysis of toxin-treated HSA recognized the formation of the intact protein adducts with primarily one and two toxin molecules attached to one HSA molecule. HSA treated with/without brevetoxin-B was digested with trypsin, trypsin following chymotrypsin, and Pronase, respectively, for LC/MS analysis of adduction sites. Brevetoxin-B was found to react primarily with Cys(34) and His(3) and with His and Lys at other sites of HSA with variable reactivity and with Lys in general the least reactive.


Assuntos
Toxinas Marinhas/química , Oxocinas/química , Albumina Sérica/química , Cromatografia Líquida de Alta Pressão , Quimotripsina/metabolismo , Cisteína/química , Dinoflagellida/metabolismo , Histidina/química , Humanos , Toxinas Marinhas/toxicidade , Espectrometria de Massas , Oxocinas/toxicidade , Pronase/metabolismo , Ligação Proteica , Albumina Sérica/metabolismo , Tripsina/metabolismo
19.
Toxicon ; 57(1): 168-71, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20688097

RESUMO

The emergence of an epilepsy syndrome in sea lions poisoned by domoic acid (DA) draws striking parallels to the single case study of temporal lobe epilepsy (TLE) that developed in an 84 yr old man one year after being poisoned by DA. To establish a basis for understanding this disease in sea lions and humans that appears to progress from DA poisoning, we have investigated the potential for a single incident of DA poisoning in rats to progress to spontaneous recurrent seizures (SRS), the hallmark of epilepsy. We have developed a DA administration protocol to induce a nonlethal status epilepticus (SE) and monitored the animals for SRS by 6 h/week of video recording. We demonstrate that a single episode of SE leads to SRS in 94% of rats (n = 23) in 6 months. These findings indicate that DA induced SE can efficiently translate to epileptic disease.


Assuntos
Epilepsia/induzido quimicamente , Ácido Caínico/análogos & derivados , Ácido Caínico/toxicidade , Toxinas Marinhas/toxicidade , Neurotoxinas/toxicidade , Convulsões/induzido quimicamente , Animais , Modelos Animais de Doenças , Progressão da Doença , Epilepsia/fisiopatologia , Masculino , Ratos , Ratos Sprague-Dawley , Convulsões/fisiopatologia , Gravação de Videoteipe
20.
BMC Neurosci ; 11: 107, 2010 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-20796285

RESUMO

BACKGROUND: Ciguatoxins (CTXs) are polyether marine neurotoxins and potent activators of voltage-gated sodium channels. This toxin is carried by multiple reef-fish species and human consumption of ciguatoxins can result in an explosive gastrointestinal/neurologic illness. This study characterizes the global transcriptional response in mouse brain to a symptomatic dose of the highly toxic Pacific ciguatoxin P-CTX-1 and additionally compares this data to transcriptional profiles from liver and whole blood examined previously. Adult male C57/BL6 mice were injected with 0.26 ng/g P-CTX-1 while controls received only vehicle. Animals were sacrificed at 1, 4 and 24 hrs and transcriptional profiling was performed on brain RNA with Agilent whole genome microarrays. RT-PCR was used to independently validate gene expression and the web tool DAVID was used to analyze gene ontology (GO) and molecular pathway enrichment of the gene expression data. RESULTS: A pronounced 4°C hypothermic response was recorded in these mice, reaching a minimum at 1 hr and lasting for 8 hrs post toxin exposure. Ratio expression data were filtered by intensity, fold change and p-value, with the resulting data used for time course analysis, K-means clustering, ontology classification and KEGG pathway enrichment. Top GO hits for this gene set included acute phase response and mono-oxygenase activity. Molecular pathway analysis showed enrichment for complement/coagulation cascades and metabolism of xenobiotics. Many immediate early genes such as Fos, Jun and Early Growth Response isoforms were down-regulated although others associated with stress such as glucocorticoid responsive genes were up-regulated. Real time PCR confirmation was performed on 22 differentially expressed genes with a correlation of 0.9 (Spearman's Rho, p < 0.0001) with microarray results. CONCLUSIONS: Many of the genes differentially expressed in this study, in parallel with the hypothermia, figure prominently in protection against neuroinflammation. Pathologic activity of the complement/coagulation cascade has been shown in patients suffering from a chronic form of ciguatera poisoning and is of particular interest in this model. Anti-inflammatory processes were at work not only in the brain but were also seen in whole blood and liver of these animals, creating a systemic anti-inflammatory environment to protect against the initial cellular damage caused by the toxin.


Assuntos
Anti-Inflamatórios , Química Encefálica/efeitos dos fármacos , Química Encefálica/genética , Ciguatoxinas/farmacologia , Fármacos Neuroprotetores , Animais , Sangue/metabolismo , Coagulação Sanguínea/efeitos dos fármacos , Perfilação da Expressão Gênica , Genes Precoces/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Análise em Microsséries , RNA/biossíntese , RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Canais de Sódio/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA