Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
1.
Mar Drugs ; 19(1)2021 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-33430011

RESUMEN

Okadaic acid (OA) and its main structural analogs dinophysistoxin-1 (DTX1) and dinophysistoxin-2 (DTX2) are marine lipophilic phycotoxins distributed worldwide that can be accumulated by edible shellfish and can cause diarrheic shellfish poisoning (DSP). In order to study their toxicokinetics, mice were treated with different doses of OA, DTX1, or DTX2 and signs of toxicity were recorded up to 24 h. Toxin distribution in the main organs from the gastrointestinal tract was assessed by liquid chromatography-mass spectrometry (LC/MS/MS) analysis. Our results indicate a dose-dependency in gastrointestinal absorption of these toxins. Twenty-four hours post-administration, the highest concentration of toxin was detected in the stomach and, in descending order, in the large intestine, small intestine, and liver. There was also a different toxicokinetic pathway between OA, DTX1, and DTX2. When the same toxin doses are compared, more OA than DTX1 is detected in the small intestine. OA and DTX1 showed similar concentrations in the stomach, liver, and large intestine tissues, but the amount of DTX2 is much lower in all these organs, providing information on DSP toxicokinetics for human safety assessment.


Asunto(s)
Toxinas Marinas/farmacocinética , Intoxicación por Mariscos , Animales , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Femenino , Intestinos , Toxinas Marinas/toxicidad , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Ácido Ocadaico/análogos & derivados , Ácido Ocadaico/farmacocinética , Mariscos/análisis , Estómago , Distribución Tisular , Toxicocinética
2.
Toxins (Basel) ; 12(12)2020 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-33371280

RESUMEN

The hepatotoxin microcystin-LR (MC-LR) represents one of the most toxic cyanotoxins for human health. Considering its harmful effect, the World Health Organization recommended a limit in drinking water (DW) of 1 µg L-1. Due to the ineffectiveness of conventional treatments present in DW treatment plants against MC-LR, advanced oxidation processes (AOPs) are gaining interest due to the high redox potential of the OH• radicals. In this work UV/H2O2 was applied to a real lake water to remove MC-LR. The kinetics of the UV/H2O2 were compared with those of UV and H2O2 showing the following result: UV/H2O2 > UV > H2O2. Within the range of H2O2 tested (0-0.9 mM), the results showed that H2O2 concentration and the removal kinetics followed an increasing quadratic relation. By increasing the initial concentration of H2O2, the consumption of oxidant also increased but, in terms of MC-LR degraded for H2O2 dosed, the removal efficiency decreased. As the initial MC-LR initial concentration increased, the removal kinetics increased up to a limit concentration (80 µg L-1) in which the presence of high amounts of the toxin slowed down the process. Operating with UV fluence lower than 950 mJ cm-2, UV alone minimized the specific energy consumption required. UV/H2O2 (0.3 mM) and UV/H2O2 (0.9 mM) were the most advantageous combination when operating with UV fluence of 950-1400 mJ cm-2 and higher than 1400 mJ cm-2, respectively.


Asunto(s)
Agua Potable/análisis , Peróxido de Hidrógeno/farmacología , Lagos/análisis , Toxinas Marinas/análisis , Microcistinas/análisis , Rayos Ultravioleta , Purificación del Agua/métodos , Conservación de los Recursos Hídricos/métodos , Agua Potable/microbiología , Humanos , Toxinas Marinas/farmacocinética , Microcistinas/farmacocinética
3.
Toxins (Basel) ; 11(8)2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31404969

RESUMEN

Bivalve molluscs represent an important food source within the Philippines, but the health of seafood consumers is compromised through the accumulation of harmful algal toxins in edible shellfish tissues. In order to assess the dynamics of toxin risk in shellfish, this study investigated the uptake, depuration, assimilation, and analogue changes of paralytic shellfish toxins in Perna viridis. Tank experiments were conducted where mussels were fed with the toxic dinoflagellate Alexandrium minutum. Water and shellfish were sampled over a six day period to determine toxin concentrations in the shellfish meat and water, as well as algal cell densities. The maximum summed toxin concentration determined was 367 µg STX eq./100 g shellfish tissue, more than six times higher than the regulatory action limit in the Philippines. Several uptake and depuration cycles were observed during the study, with the first observed within the first 24 h coinciding with high algal cell densities. Toxin burdens were assessed within different parts of the shellfish tissue, with the highest levels quantified in the mantle during the first 18 h period but shifting towards the gut thereafter. A comparison of toxin profile data evidenced the conversion of GTX1,4 in the source algae to the less potent GTX2,3 in the shellfish tissue. Overall, the study illustrated the temporal variability in Perna viridis toxin concentrations during a modelled algal bloom event, and the accumulation of toxin from the water even after toxic algae were removed.


Asunto(s)
Bivalvos/metabolismo , Toxinas Marinas/metabolismo , Mariscos/análisis , Animales , Transporte Biológico , Floraciones de Algas Nocivas , Toxinas Marinas/farmacocinética
4.
Toxins (Basel) ; 10(11)2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30400229

RESUMEN

Several species of the dinoflagellate genus Dinophysis produce toxins that accumulate in bivalves when they feed on populations of these organisms. The accumulated toxins can lead to intoxication in consumers of the affected bivalves. The risk of intoxication depends on the amount and toxic power of accumulated toxins. In this review, current knowledge on the main processes involved in toxin accumulation were compiled, including the mechanisms and regulation of toxin acquisition, digestion, biotransformation, compartmentalization, and toxin depuration. Finally, accumulation kinetics, some models to describe it, and some implications were also considered.


Asunto(s)
Alveolados/metabolismo , Bivalvos/metabolismo , Toxinas Marinas/metabolismo , Alveolados/fisiología , Animales , Compartimento Celular , Digestión , Toxinas Marinas/farmacocinética
5.
Arch Toxicol ; 92(1): 143-156, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29110038

RESUMEN

Palytoxin, isolated from a zoanthid of the genus Palythoa, is the most potent marine toxin known. Intoxication by palytoxin leads to vasoconstriction, hemorrhage, ataxia, muscle weakness, ventricular fibrillation, pulmonary hypertension, ischemia and death. Palytoxin and its numerous derivatives (congeners) may enter the food chain and accumulate mainly in fishes and crabs, causing severe human intoxication and death following ingestion of contaminated products. Furthermore, toxic effects in individuals exposed via inhalation or skin contact to marine aerosol in coincidence with Ostreopsis blooms, have been reported. Blooms of the benthic dinoflagellate Ostreopsis cf. ovata are a concern in the Mediterranean Sea, since this species produces a wide range of palytoxin-like compounds listed among the most potent marine toxins. Thus, the formerly unsuspected broad distribution of the benthic dinoflagellate Ostreopsis spp. has recently posed a problem of risk assessment for human health. Palytoxin has a strong potential for toxicity in humans and animals, and currently this toxin is of great concern worldwide. This review summarized and discussed the pharmacology and toxicology data of palytoxin and its congeners, including their cytotoxicity, human and animal toxicities. Moreover, the risk assessment and their control strategies including prevention and treatment assays were evaluated.


Asunto(s)
Acrilamidas/química , Acrilamidas/toxicidad , Venenos de Cnidarios/química , Venenos de Cnidarios/toxicidad , Acrilamidas/farmacocinética , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Compuestos Bicíclicos Heterocíclicos con Puentes/toxicidad , Venenos de Cnidarios/farmacocinética , Humanos , Toxinas Marinas/química , Toxinas Marinas/farmacocinética , Toxinas Marinas/toxicidad , Piranos/química , Piranos/toxicidad , Medición de Riesgo/métodos
6.
Toxicol Lett ; 282: 136-146, 2018 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-29107028

RESUMEN

Azaspiracids (AZAs) are marine algal toxins that can be accumulated by edible shellfish to cause a foodborne gastrointestinal poisoning in humans. In the European Union, only AZA1, -2 and -3 are currently regulated and their concentration in shellfish is determined through their toxic equivalency factors (TEFs) derived from the intraperitoneal lethal potency in mice. Nevertheless, considering the potential human exposure by oral route, AZAs TEFs should be calculated by comparative oral toxicity data. Thus, the acute oral toxicity of AZA1, -2 and -3 was investigated in female CD-1 mice treated with different doses (AZA1: 135-1100µg/kg; AZA2 and AZA3: 300-1100µg/kg) and sacrificed after 24h or 14days. TEFs derived from the median lethal doses (LD50) were 1.0, 0.7 and 0.5, respectively for AZA1, -2 and -3. In fact, after 24h from gavage administration, LD50s were 443µg/kg (AZA1; 95% CL: 350-561µg/kg), 626µg/kg (AZA2; 95% CL: 430-911µg/kg) and 875µg/kg (AZA3; 95% CL: 757-1010µg/kg). Mice dead more than 5h after the treatment or those sacrificed after 24h (doses: ≥175µg AZA1/kg, ≥500µg AZA2/kg and ≥600µg AZA3/kg) showed enlarged pale liver, while increased serum markers of liver alteration were recorded even at the lowest doses. Blood chemistry revealed significantly increased serum levels of K+ ions (≥500mg/kg), whereas light microscopy showed tissue changes in the gastrointestinal tract, liver and spleen. No lethality, macroscopic, tissue or haematological changes were recorded two weeks post exposure, indicating reversible toxic effects. LC-MS/MS analysis of the main organs showed a dose-dependency in gastrointestinal absorption of these toxins: at 24h, the highest levels were detected in the stomach and, in descending order, in the intestinal content, liver, small intestine, kidneys, lungs, large intestine, heart as well as detectable traces in the brain. After 14days, AZA1 and AZA2 were still detectable in almost all the organs and intestinal content.


Asunto(s)
Furanos/toxicidad , Toxinas Marinas/toxicidad , Piranos/toxicidad , Compuestos de Espiro/toxicidad , Administración Oral , Animales , Relación Dosis-Respuesta a Droga , Femenino , Furanos/farmacocinética , Dosificación Letal Mediana , Toxinas Marinas/farmacocinética , Ratones Endogámicos , Mytilus edulis/química , Especificidad de Órganos , Piranos/farmacocinética , Compuestos de Espiro/farmacocinética , Distribución Tisular , Pruebas de Toxicidad Aguda
7.
Cell Physiol Biochem ; 43(1): 136-146, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28848202

RESUMEN

BACKGROUND: Azaspiracids (AZAs) are marine biotoxins produced by the dinoflagellates genera Azadinium and Amphidoma. These toxins cause azaspiracid poisoning (AZP), characterized by severe gastrointestinal illness in humans after the consumption of bivalve molluscs contaminated with AZAs. The main aim of the present study was to examine the consequences of human exposure to AZA1 by the study of absorption and effects of the toxin on Caco-2 cells, a reliable model of the human intestine. METHODS: The ability of AZA1 to cross the human intestinal epithelium has been evaluated by the Caco-2 transepithelial permeability assay. The toxin has been detected and quantified using a microsphere-based immunoassay. Cell alterations and ultrastructural effects has been observed with confocal and transmission electron microscopy Results: AZA1 was absorbed by Caco-2 cells in a dose-dependent way without affecting cell viability. However, modifications on occludin distribution detected by confocal microscopy imaging indicated a possible monolayer integrity disruption. Nevertheless, transmission electron microscopy imaging revealed ultrastructural damages at the nucleus and mitochondria with autophagosomes in the cytoplasm, however, tight junctions and microvilli remained unaffected. CONCLUSION: After the ingestion of molluscs with the AZA1, the toxin will be transported through the human intestinal barrier to blood causing damage on epithelial cells.


Asunto(s)
Toxinas Marinas/farmacología , Permeabilidad/efectos de los fármacos , Compuestos de Espiro/farmacología , Autofagosomas/efectos de los fármacos , Autofagosomas/ultraestructura , Células CACO-2 , Núcleo Celular/efectos de los fármacos , Núcleo Celular/ultraestructura , Supervivencia Celular/efectos de los fármacos , Dinoflagelados/metabolismo , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Toxinas Marinas/farmacocinética , Microscopía Confocal , Microscopía Electrónica de Transmisión , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Ocludina/metabolismo , Compuestos de Espiro/farmacocinética
8.
Aquat Toxicol ; 187: 29-37, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28363127

RESUMEN

Harmful algal blooms (HABs) occur nearly annually off the west coast of Florida and can impact both humans and wildlife, resulting in morbidity and increased mortality of marine animals including sea turtles. The key organism in Florida red tides is the dinoflagellate Karenia brevis that produces a suite of potent neurotoxins referred to as the brevetoxins (PbTx). Despite recent mortality events and rehabilitation efforts, still little is known about how the toxin directly impacts sea turtles, as they are not amenable to experimentation and what is known about toxin levels and distribution comes primarily from post-mortem data. In this study, we utilized the freshwater turtle Trachemys scripta and the diamondback terrapin, Malaclemys terrapin as model organisms to determine the distribution, clearance, and routes of excretion of the most common form of the toxin, brevetoxin-3, in turtles. Turtles were administered toxin via esophageal tube to mimic ingestion (33.48µg/kg PbTx-3, 3×/week for two weeks for a total of 7 doses) or by intratracheal instillation (10.53µg/kg, 3×/week for four weeks for a total of 12 doses) to mimic inhalation. Both oral and intratracheal administration of the toxin produced a suite of behavioral responses symptomatic of brevetoxicosis. The toxin distributed to all organ systems within 1h of administration but was rapidly cleared out over 24-48h, corresponding to a decline in clinical symptoms. Excretion appears to be primarily through conjugation to bile salts. Histopathological study revealed that the frequency of lesions varied within experimental groups with some turtles having no significant lesions at all, while similar lesions were found in a low number of control turtles suggesting another common factor(s) could be responsible. The overall goal of this research is better understand the impacts of brevetoxin on turtles in order to develop better treatment protocols for sea turtles exposed to HABs.


Asunto(s)
Toxinas Marinas/farmacocinética , Neurotoxinas/farmacocinética , Oxocinas/farmacocinética , Tortugas/metabolismo , Contaminantes Químicos del Agua/farmacocinética , Administración Oral , Animales , Conducta Animal/efectos de los fármacos , Dinoflagelados/metabolismo , Femenino , Florida , Agua Dulce/química , Floraciones de Algas Nocivas , Humanos , Exposición por Inhalación , Masculino , Toxinas Marinas/toxicidad , Tasa de Depuración Metabólica , Modelos Biológicos , Neurotoxinas/toxicidad , Especificidad de Órganos , Oxocinas/toxicidad , Distribución Tisular , Contaminantes Químicos del Agua/toxicidad
9.
Food Chem Toxicol ; 102: 166-175, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28223118

RESUMEN

Ingestion of shellfish with dinophysistoxin-2 (DTX2) can lead to diarrheic shellfish poisoning (DSP). The official control method of DSP toxins in seafood is the liquid chromatography-mass spectrometry analysis (LC-MS). However in order to calculate the total toxicity of shellfish, the concentration of each compound must be multiplied by individual Toxicity Equivalency Factor (TEF). Considering that TEFs caused some controversy and the scarce information about DTX2 toxicity, the aim of this study was to characterize the oral toxicity of DTX2 in mice. A 4-Level Up and Down Procedure allowed the characterization of DTX2 effects and the estimation of DTX2 oral TEF based on determination of the lethal dose 50 (LD50). DTX2 passed the gastrointestinal barrier and was detected in urine and feces. Acute toxicity symptoms include diarrhea and motionless, however anatomopathology study and ultrastructural images restricted the toxin effects to the gastrointestinal tract. Nevertheless enterocytes microvilli and tight junctions were not altered, disconnecting DTX2 diarrheic effects from paracellular epithelial permeability. This is the first report of DTX2 oral LD50 (2262 µg/kg BW) indicating that its TEF is about 0.4. This result suggests reevaluation of the present TEFs for the DSP toxins to better determine the actual risk to seafood consumers.


Asunto(s)
Piranos/administración & dosificación , Piranos/toxicidad , Pruebas de Toxicidad/métodos , Administración Oral , Animales , Peso Corporal/efectos de los fármacos , Cromatografía Liquida/métodos , Ingestión de Alimentos/efectos de los fármacos , Heces/química , Femenino , Intestinos/efectos de los fármacos , Intestinos/patología , Intestinos/ultraestructura , Dosificación Letal Mediana , Hígado/efectos de los fármacos , Hígado/patología , Hígado/ultraestructura , Toxinas Marinas/administración & dosificación , Toxinas Marinas/farmacocinética , Toxinas Marinas/toxicidad , Ratones , Ácido Ocadaico/análogos & derivados , Piranos/farmacocinética , Espectrometría de Masas en Tándem/métodos
10.
Food Chem Toxicol ; 101: 121-127, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28089692

RESUMEN

Okadaic acid (OA), Dinophysistoxins (DTX1 and DTX2) and their acyl-derivatives (DTX3) are marine toxins responsible for the human diarrhetic shellfish poisoning. To date the amount of toxins ingested from consumption of shellfish has been considered equal to the amount of toxins available for uptake by the human body. The aim of this study is to assess the OA, DTX2 and DTX3 fractions released from raw and steamed mussels and cockles into the digestive fluids (bioaccessibility) using a static in vitro digestion model. Higher bioaccessibility was found in mussels (86 ± 4%) than in cockles (59 ± 9%). A significant reduction of ester derivatives of OA and an increase of OA were observed in the bioaccessible fraction of mussel samples, suggesting that DTX3 undergo conversion into their more toxic parent compounds during human digestion. However, similar increase of DTX2 and reduction of the respective acyl derivatives was not observed. Steaming lead to significant reduction of OA and analogues bioaccessibility in both species even though increased concentrations of toxins are obtained after this treatment. Risk assessment based solely on DSP toxins occurrence in seafood can conduct to an overestimation of the exposure and lead to more conservative regulatory measures.


Asunto(s)
Bivalvos/efectos de los fármacos , Toxinas Marinas/farmacocinética , Ácido Ocadaico/farmacocinética , Piranos/farmacocinética , Mariscos/análisis , Vapor , Animales , Carcinógenos/farmacocinética , Cromatografía Liquida/métodos , Ésteres/química , Ácidos Grasos/química , Humanos , Técnicas In Vitro , Espectrometría de Masas/métodos
11.
Food Chem Toxicol ; 89: 54-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26783635

RESUMEN

Okadaic acid (OA) and their derivatives are marine toxins responsible for the human diarrhetic shellfish poisoning (DSP). To date the amount of toxins ingested in food has been considered equal to the amount of toxins available for uptake by the human body. In this study, the OA fraction released from the food matrix into the digestive fluids (bioaccessibility) was assessed using a static in vitro digestion model. Naturally contaminated mussels (Mytilus galloprovincialis) and donax clams (Donax sp.), collected from the Portuguese coast, containing OA and dinophysistoxin-3 (DTX3) were used in this study. Bioaccessibility of OA total content was 88% and 75% in mussels and donax clams, respectively. Conversion of DTX3 into its parent compound was verified during the simulated digestive process and no degradation of these toxins was found during the process. This is the first study assessing the bioaccessibility of OA-group toxins in naturally contaminated seafood. This study provides relevant new data that can improve and lead to more accurate food safety risk assessment studies concerning these toxins.


Asunto(s)
Toxinas Marinas/farmacocinética , Ácido Ocadaico/farmacocinética , Mariscos/análisis , Animales , Disponibilidad Biológica , Cromatografía Liquida , Técnicas In Vitro , Espectrometría de Masas en Tándem
12.
Toxicon ; 98: 1-3, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25702960

RESUMEN

Seasonal dynamics and the distribution of dinophysistoxin-3 (DTX-3) in organs of mussel Crenomytilus grayanus, collected in 2013 in Peter the Great Bay, Sea of Japan, were determined. Active toxin forms (OA, DTX-1/2) concentrated in digestive glands (60-80%), whereas non-active 7-O-acyl derivatives (DTX-3) were more abundant in edible soft tissues (more than 80%). Possible mechanism of fast removing of DTX-3 from the digestive gland and accumulation of DTX-3 in the soft tissues is supposed.


Asunto(s)
Toxinas Marinas/farmacocinética , Mytilidae/química , Piranos/farmacocinética , Mariscos/análisis , Animales , Cromatografía Líquida de Alta Presión , Japón , Ácido Ocadaico/análogos & derivados , Distribución Tisular
13.
Toxicon ; 97: 46-52, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25681577

RESUMEN

Perna viridis is a recently introduced species to US coastal waters and have vigorously spread throughout the southeastern seaboard since their invasion. Little information regarding their response to local environmental factors has been reported including responses to the local HAB species, Karenia brevis. This study monitored the tissue toxin concentration of brevetoxins in P. viridis from existing populations throughout two consecutive natural K. brevis blooms. The results showed P. viridis to rapidly accumulate PbTx upon exposure to the bloom, far exceeding the peak tissue concentrations of oysters, Crassostrea virginica, sampled during the same period, 57,653 ± 15,937 and 33,462 ± 10,391 ng g(-1) PbTx-3 equivalent, respectively. Further, P. viridis retained high PbTx concentrations in their tissues post bloom remaining above the regulatory limit for human consumption for 4-5 months, significantly longer than the depuration time of 2-8 weeks for native oyster and clam species. In the second year, the bloom persisted at high cell concentrations resulting in prolonged exposure and higher PbTx tissue concentrations indicating increased bioaccumulation in green mussels. While this species is not currently harvested for human consumption, the threat for post bloom trophic transfer could pose negative impacts on other important fisheries and higher food web implications.


Asunto(s)
Cadena Alimentaria , Floraciones de Algas Nocivas , Especies Introducidas , Toxinas Marinas/farmacocinética , Oxocinas/farmacocinética , Perna/metabolismo , Animales , Área Bajo la Curva , Crassostrea/metabolismo , Dinoflagelados/química , Ensayo de Inmunoadsorción Enzimática , Florida , Toxinas Marinas/metabolismo , Oxocinas/metabolismo , Especificidad de la Especie , Factores de Tiempo
14.
Chem Res Toxicol ; 27(7): 1166-75, 2014 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-24949875

RESUMEN

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.


Asunto(s)
Cisteína/química , Lípidos/química , Toxinas Marinas/farmacocinética , Neurotoxinas/farmacocinética , Oxocinas/farmacocinética , Administración Intravenosa , Animales , Encéfalo/metabolismo , Sistema Digestivo/metabolismo , Heces/química , Riñón/metabolismo , Pulmón/metabolismo , Masculino , Toxinas Marinas/sangre , Toxinas Marinas/química , Toxinas Marinas/orina , Ratones Endogámicos C57BL , Músculos/metabolismo , Miocardio/metabolismo , Neurotoxinas/sangre , Neurotoxinas/química , Neurotoxinas/orina , Oxocinas/sangre , Oxocinas/química , Oxocinas/orina , Bazo/metabolismo , Testículo/metabolismo , Distribución Tisular
15.
J Clin Pharmacol ; 54(11): 1239-46, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24922179

RESUMEN

Pediatric drug development is challenging when a product is studied for a pediatric disease that has a different underlying etiology and pathophysiology compared to the adult disease. Neurogenic bladder dysfunction (NBD) is such a therapeutic area with multiple unsuccessful development programs. The objective of this study was to critically evaluate clinical trial design elements that may have contributed to unsuccessful drug development programs for pediatric NBD. Trial design elements of drugs tested for pediatric NBD were identified from trials submitted to the U.S. Food and Drug Administration. Data were extracted from publically available FDA reviews and labeling and included trial design, primary endpoints, enrollment eligibilities, and pharmacokinetic data. A total of four products were identified. Although all four programs potentially provided clinically useful information, only one drug (oxybutynin) demonstrated efficacy in children with NBD. The lack of demonstrable efficacy for the remainder of the products illustrates that future trials should give careful attention to testing a range of doses, using objectively measured, clinically meaningful endpoints, and selecting clinical trial designs that are both interpretable and feasible. Compiling the drug development experience with pediatric NBD will facilitate an improved approach for future drug development for this, and perhaps other, therapeutic areas.


Asunto(s)
Compuestos de Bencidrilo/uso terapéutico , Cresoles/uso terapéutico , Ácidos Mandélicos/uso terapéutico , Toxinas Marinas/uso terapéutico , Oxocinas/uso terapéutico , Fenilpropanolamina/uso terapéutico , Quinazolinas/uso terapéutico , Vejiga Urinaria Neurogénica/tratamiento farmacológico , Adolescente , Antagonistas de Receptores Adrenérgicos alfa 1/administración & dosificación , Antagonistas de Receptores Adrenérgicos alfa 1/farmacocinética , Antagonistas de Receptores Adrenérgicos alfa 1/uso terapéutico , Área Bajo la Curva , Compuestos de Bencidrilo/administración & dosificación , Compuestos de Bencidrilo/farmacocinética , Niño , Preescolar , Cresoles/administración & dosificación , Cresoles/farmacocinética , Preparaciones de Acción Retardada , Humanos , Lactante , Ácidos Mandélicos/administración & dosificación , Ácidos Mandélicos/farmacocinética , Toxinas Marinas/administración & dosificación , Toxinas Marinas/farmacocinética , Antagonistas Muscarínicos/administración & dosificación , Antagonistas Muscarínicos/farmacocinética , Antagonistas Muscarínicos/uso terapéutico , Oxocinas/administración & dosificación , Oxocinas/farmacocinética , Fenilpropanolamina/administración & dosificación , Fenilpropanolamina/farmacocinética , Quinazolinas/administración & dosificación , Quinazolinas/farmacocinética , Comprimidos , Tartrato de Tolterodina
16.
J Am Soc Nephrol ; 25(6): 1187-97, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24511141

RESUMEN

Domoic acid (DA), an excitatory amino acid produced by diatoms belonging to the genus Pseudo-nitzschia, is a glutamate analog responsible for the neurologic condition referred to as amnesic shellfish poisoning. To date, the renal effects of DA have been underappreciated, although renal filtration is the primary route of systemic elimination and the kidney expresses ionotropic glutamate receptors. To characterize the renal effects of DA, we administered either a neurotoxic dose of DA or doses below the recognized limit of toxicity to adult Sv128/Black Swiss mice. DA preferentially accumulated in the kidney and elicited marked renal vascular and tubular damage consistent with acute tubular necrosis, apoptosis, and renal tubular cell desquamation, with toxic vacuolization and mitochondrial swelling as hallmarks of the cellular damage. Doses≥0.1 mg/kg DA elevated the renal injury biomarkers kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin, and doses≥0.005 mg/kg induced the early response genes c-fos and junb. Coadministration of DA with the broad spectrum excitatory amino acid antagonist kynurenic acid inhibited induction of c-fos, junb, and neutrophil gelatinase-associated lipocalin. These findings suggest that the kidney may be susceptible to excitotoxic agonists, and renal effects should be considered when examining glutamate receptor activation. Additionally, these results indicate that DA is a potent nephrotoxicant, and potential renal toxicity may require consideration when determining safe levels for human exposure.


Asunto(s)
Ácido Kaínico/análogos & derivados , Toxinas Marinas/toxicidad , Fármacos Neuromusculares Despolarizantes/toxicidad , Unión Neuromuscular/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Ácido Kaínico/farmacocinética , Ácido Kaínico/toxicidad , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Toxinas Marinas/farmacocinética , Ratones Endogámicos , Microscopía Electrónica de Transmisión , Dilatación Mitocondrial/efectos de los fármacos , Miocardio/metabolismo , Miocardio/patología , Fármacos Neuromusculares Despolarizantes/farmacocinética , Unión Neuromuscular/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de Ácido Kaínico/genética , Receptores de Ácido Kaínico/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vacuolas/patología , Vacuolas/ultraestructura , Receptor de Ácido Kaínico GluK2
17.
Toxins (Basel) ; 6(1): 211-28, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24394641

RESUMEN

Okadaic acid (OA) and its analogues, dinophysistoxin 1 (DTX1) and dinophysistoxin 2 (DTX2), are lipophilic and heat-stable marine toxins produced by dinoflagellates, which can accumulate in filter-feeding bivalves. These toxins cause diarrheic shellfish poisoning (DSP) in humans shortly after the ingestion of contaminated seafood. Studies carried out in mice indicated that DSP poisonous are toxic towards experimental animals with a lethal oral dose 2-10 times higher than the intraperitoneal (i.p.) lethal dose. The focus of this work was to study the absorption of OA, DTX1 and DTX2 through the human gut barrier using differentiated Caco-2 cells. Furthermore, we compared cytotoxicity parameters. Our data revealed that cellular viability was not compromised by toxin concentrations up to 1 µM for 72 h. Okadaic acid and DTX2 induced no significant damage; nevertheless, DTX1 was able to disrupt the integrity of Caco-2 monolayers at concentrations above 50 nM. In addition, confocal microscopy imaging confirmed that the tight-junction protein, occludin, was affected by DTX1. Permeability assays revealed that only DTX1 was able to significantly cross the intestinal epithelium at concentrations above 100 nM. These data suggest a higher oral toxicity of DTX1 compared to OA and DTX2.


Asunto(s)
Toxinas Marinas/toxicidad , Ácido Ocadaico/toxicidad , Piranos/toxicidad , Células CACO-2 , Humanos , Absorción Intestinal/efectos de los fármacos , Dosificación Letal Mediana , Toxinas Marinas/farmacocinética , Microscopía Confocal , Ácido Ocadaico/análogos & derivados , Ácido Ocadaico/farmacocinética , Piranos/farmacocinética
18.
Environ Toxicol Pharmacol ; 37(1): 1-6, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24280256

RESUMEN

There are increasing concerns about health risk of human exposure to microcystin-LR (MC-LR) over the past few decades. Nevertheless, its female mammalian reproductive toxicity has not been addressed yet. In the present study, we firstly reported MC-LR could impact female reproductive function of mammals. After 28 days MC-LR exposure, relative ovary weight significantly reduced in 20 µg/kg MC-LR group and this reduction may be relative with pathomorphological changes of ovary. The result of histological evaluation of follicles showed that primordial follicles decreased roughly in half at high dose level compared with control. Since serum hormone assay indicated that MC-LR induced decrease of progesterone but not FSH or LH, disturbance of estrus cycle was seemed to result from direct impact of ovary rather than indirectly from hypothalamus or pituitary. As expected, MC-LR was detected in the ovaries of MC-LR exposure mice by immunoblot analysis.


Asunto(s)
Toxinas Bacterianas/toxicidad , Toxinas Marinas/toxicidad , Microcistinas/toxicidad , Ovario/efectos de los fármacos , Animales , Toxinas Bacterianas/farmacocinética , Ciclo Estral/efectos de los fármacos , Femenino , Toxinas Marinas/farmacocinética , Ratones , Ratones Endogámicos BALB C , Microcistinas/farmacocinética , Tamaño de los Órganos/efectos de los fármacos , Ovario/metabolismo , Ovario/patología , Progesterona/sangre , Reproducción
19.
Toxicol Lett ; 225(2): 311-7, 2014 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-24374049

RESUMEN

The consumption of okadaic acid (OA) contaminated shellfish can induce acute toxic symptoms in humans such as diarrhea, nausea, vomiting and abdominal pain; carcinogenic and embryotoxic effects have also been described. Toxicokinetic studies with mice have shown that high cytotoxic doses of OA can pass the gastrointestinal barrier presumably by paracellular passage. However, in vitro studies using human intestinal Caco-2 cell monolayers to represent the intestinal barrier have shown that at low-dose exposure OA is transported against a concentration gradient suggesting an active efflux mechanism. Since P-glycoprotein (P-gp) transports a wide variety of substrates, we investigated its possible influence on the observed elimination of OA. We used two different cellular transwell models: (i) Caco-2 cell monolayer endogenously expressing human P-gp and simulating the intestinal barrier and (ii) MDCK-II cell monolayer stably over-expressing P-gp. Our study demonstrates clearly that OA at non-cytotoxic concentrations passes the monolayer barrier only to a low degree, and that it is actively eliminated by P-gp over the apical membrane. Therefore, our in vitro data indicate that humans appear to have efficient defense mechanisms to protect themselves against low-dose contaminated shellfish by exhibiting a low bioavailability as a result of active elimination of OA by P-gp.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Carcinógenos/farmacocinética , Intestinos/efectos de los fármacos , Ácido Ocadaico/farmacocinética , Animales , Disponibilidad Biológica , Transporte Biológico , Células CACO-2 , Carcinógenos/toxicidad , Cromatografía Líquida de Alta Presión , Perros , Relación Dosis-Respuesta a Droga , Humanos , Inactivación Metabólica , Mucosa Intestinal/metabolismo , Células de Riñón Canino Madin Darby , Toxinas Marinas/farmacocinética , Toxinas Marinas/toxicidad , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ácido Ocadaico/toxicidad , Extracción en Fase Sólida , Espectrometría de Masas en Tándem
20.
Toxicon ; 75: 16-26, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23726853

RESUMEN

Following a review of official control data on shellfish in France, Ingril Lagoon had been identified as a site where positive mouse bioassays for lipophilic toxins had been repeatedly observed. These unexplained mouse bioassays, also called atypical toxicity, coincided with an absence of regulated toxins and rapid death times in mice observed in the assay. The present study describes pinnatoxin G as the main compound responsible for the toxicity observed using the mouse bioassay for lipophilic toxins. Using a well-characterised standard for pinnatoxin G, LC-MS/MS analysis of mussel samples collected from 2009 to 2012 revealed regular occurrences of pinnatoxin G at levels sufficient to account for the toxicity in the mouse bioassays. Baseline levels of pinnatoxin G from May to October usually exceeded 40 µg kg(-1) in whole flesh, with a maximum in September 2010 of around 1200 µg kg(-1). These concentrations were much greater than those at the other 10 sites selected for vigilance testing, where concentrations did not exceed 10 µg kg(-1) in a 3-month survey from April to July 2010, and where rapid mouse deaths were not typically observed. Mussels were always more contaminated than clams, confirming that mussel is a good sentinel species for pinnatoxins. Profiles in mussels and clams were similar, with the concentration of pinnatoxin A less than 2% that of pinnatoxin G, and pteriatoxins were only present in non-quantifiable traces. Esters of pinnatoxin G could not be detected by analysis of extracts before and after alkaline hydrolysis. Analysis with a receptor-binding assay showed that natural pinnatoxin G was similarly active on the nicotinic acetylcholine receptor as chemically synthesized pinnatoxin G. Culture of Vulcanodinium rugosum, previously isolated from Ingril lagoon, confirmed that this alga is a pinnatoxin G producer (4.7 pg cell(-1)). Absence of this organism from the water column during prolonged periods of shellfish contamination and the dominance of non-motile life stages of V. rugosum both suggest that further studies will be required to fully describe the ecology of this organism and the accumulation of pinnatoxins in shellfish.


Asunto(s)
Alcaloides/toxicidad , Bivalvos/efectos de los fármacos , Toxinas Marinas/toxicidad , Mytilus/efectos de los fármacos , Compuestos de Espiro/toxicidad , Alcaloides/química , Alcaloides/farmacocinética , Animales , Bioensayo , Cromatografía Líquida de Alta Presión , Cromatografía Liquida , Dinoflagelados/metabolismo , Francia , Sedimentos Geológicos/química , Toxinas Marinas/química , Toxinas Marinas/farmacocinética , Ratones , Compuestos de Espiro/química , Compuestos de Espiro/farmacocinética , Espectrometría de Masas en Tándem , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...