Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Mol Neurosci ; 17: 1222935, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38495551

RESUMO

This study reports on biallelic homozygous and monoallelic de novo variants in SLITRK3 in three unrelated families presenting with epileptic encephalopathy associated with a broad neurological involvement characterized by microcephaly, intellectual disability, seizures, and global developmental delay. SLITRK3 encodes for a transmembrane protein that is involved in controlling neurite outgrowth and inhibitory synapse development and that has an important role in brain function and neurological diseases. Using primary cultures of hippocampal neurons carrying patients' SLITRK3 variants and in combination with electrophysiology, we demonstrate that recessive variants are loss-of-function alleles. Immunostaining experiments in HEK-293 cells showed that human variants C566R and E606X change SLITRK3 protein expression patterns on the cell surface, resulting in highly accumulating defective proteins in the Golgi apparatus. By analyzing the development and phenotype of SLITRK3 KO (SLITRK3-/-) mice, the study shows evidence of enhanced susceptibility to pentylenetetrazole-induced seizure with the appearance of spontaneous epileptiform EEG as well as developmental deficits such as higher motor activities and reduced parvalbumin interneurons. Taken together, the results exhibit impaired development of the peripheral and central nervous system and support a conserved role of this transmembrane protein in neurological function. The study delineates an emerging spectrum of human core synaptopathies caused by variants in genes that encode SLITRK proteins and essential regulatory components of the synaptic machinery. The hallmark of these disorders is impaired postsynaptic neurotransmission at nerve terminals; an impaired neurotransmission resulting in a wide array of (often overlapping) clinical features, including neurodevelopmental impairment, weakness, seizures, and abnormal movements. The genetic synaptopathy caused by SLITRK3 mutations highlights the key roles of this gene in human brain development and function.

2.
Sci Rep ; 12(1): 3114, 2022 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-35210470

RESUMO

On 11th March 2020, the UK government announced plans for the scaling of COVID-19 testing, and on 27th March 2020 it was announced that a new alliance of private sector and academic collaborative laboratories were being created to generate the testing capacity required. The Cambridge COVID-19 Testing Centre (CCTC) was established during April 2020 through collaboration between AstraZeneca, GlaxoSmithKline, and the University of Cambridge, with Charles River Laboratories joining the collaboration at the end of July 2020. The CCTC lab operation focussed on the optimised use of automation, introduction of novel technologies and process modelling to enable a testing capacity of 22,000 tests per day. Here we describe the optimisation of the laboratory process through the continued exploitation of internal performance metrics, while introducing new technologies including the Heat Inactivation of clinical samples upon receipt into the laboratory and a Direct to PCR protocol that removed the requirement for the RNA extraction step. We anticipate that these methods will have value in driving continued efficiency and effectiveness within all large scale viral diagnostic testing laboratories.


Assuntos
SARS-CoV-2
3.
Sci Rep ; 12(1): 2883, 2022 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-35190592

RESUMO

We report the development of a large scale process for heat inactivation of clinical COVID-19 samples prior to laboratory processing for detection of SARS-CoV-2 by RT-qPCR. With more than 266 million confirmed cases, over 5.26 million deaths already recorded at the time of writing, COVID-19 continues to spread in many parts of the world. Consequently, mass testing for SARS-CoV-2 will remain at the forefront of the COVID-19 response and prevention for the near future. Due to biosafety considerations the standard testing process requires a significant amount of manual handling of patient samples within calibrated microbiological safety cabinets. This makes the process expensive, effects operator ergonomics and restricts testing to higher containment level laboratories. We have successfully modified the process by using industrial catering ovens for bulk heat inactivation of oropharyngeal/nasopharyngeal swab samples within their secondary containment packaging before processing in the lab to enable all subsequent activities to be performed in the open laboratory. As part of a validation process, we tested greater than 1200 clinical COVID-19 samples and showed less than 1 Cq loss in RT-qPCR test sensitivity. We also demonstrate the bulk heat inactivation protocol inactivates a murine surrogate of human SARS-CoV-2. Using bulk heat inactivation, the assay is no longer reliant on containment level 2 facilities and practices, which reduces cost, improves operator safety and ergonomics and makes the process scalable. In addition, heating as the sole method of virus inactivation is ideally suited to streamlined and more rapid workflows such as 'direct to PCR' assays that do not involve RNA extraction or chemical neutralisation methods.


Assuntos
Teste de Ácido Nucleico para COVID-19/métodos , COVID-19/diagnóstico , Contenção de Riscos Biológicos/métodos , Temperatura Alta , Reação em Cadeia da Polimerase em Tempo Real/métodos , SARS-CoV-2/genética , Manejo de Espécimes/métodos , Inativação de Vírus , Animais , COVID-19/virologia , Linhagem Celular , Humanos , Camundongos , Vírus da Hepatite Murina/genética , RNA Viral/genética , RNA Viral/isolamento & purificação , Sensibilidade e Especificidade
4.
Sci Rep ; 9(1): 17293, 2019 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-31754242

RESUMO

A possible role for calcium signalling in the autosomal dominant form of dementia, familial encephalopathy with neuroserpin inclusion bodies (FENIB), has been proposed, which may point towards a mechanism by which cells could sense and respond to the accumulation of mutant serpin polymers in the endoplasmic reticulum (ER). We therefore explored possible defects in Ca2+-signalling, which may contribute to the pathology associated with another serpinopathy, α1-antitrypsin (AAT) deficiency. Using CHO K1 cell lines stably expressing a wild type human AAT (MAAT) and a disease-causing polymer-forming variant (ZAAT) and the truncated variant (NHK AAT), we measured basal intracellular free Ca2+, its responses to thapsigargin (TG), an ER Ca2+-ATPase blocker, and store-operated Ca2+-entry (SOCE). Our fura2 based Ca2+ measurements detected no differences between these 3 parameters in cell lines expressing MAAT and cell lines expressing ZAAT and NHK AAT mutants. Thus, in our cell-based models of α1-antitrypsin (AAT) deficiency, unlike the case for FENIB, we were unable to detect defects in calcium signalling.


Assuntos
Sinalização do Cálcio/genética , Cálcio/metabolismo , Epilepsias Mioclônicas/genética , Transtornos Heredodegenerativos do Sistema Nervoso/genética , alfa 1-Antitripsina/metabolismo , Animais , Células CHO , Cricetulus , Epilepsias Mioclônicas/patologia , Transtornos Heredodegenerativos do Sistema Nervoso/patologia , Humanos , Microscopia Confocal , Mutação , Imagem Óptica , alfa 1-Antitripsina/genética
5.
Am J Hum Genet ; 104(4): 721-730, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30929742

RESUMO

VAMP2 encodes the vesicular SNARE protein VAMP2 (also called synaptobrevin-2). Together with its partners syntaxin-1A and synaptosomal-associated protein 25 (SNAP25), VAMP2 mediates fusion of synaptic vesicles to release neurotransmitters. VAMP2 is essential for vesicular exocytosis and activity-dependent neurotransmitter release. Here, we report five heterozygous de novo mutations in VAMP2 in unrelated individuals presenting with a neurodevelopmental disorder characterized by axial hypotonia (which had been present since birth), intellectual disability, and autistic features. In total, we identified two single-amino-acid deletions and three non-synonymous variants affecting conserved residues within the C terminus of the VAMP2 SNARE motif. Affected individuals carrying de novo non-synonymous variants involving the C-terminal region presented a more severe phenotype with additional neurological features, including central visual impairment, hyperkinetic movement disorder, and epilepsy or electroencephalography abnormalities. Reconstituted fusion involving a lipid-mixing assay indicated impairment in vesicle fusion as one of the possible associated disease mechanisms. The genetic synaptopathy caused by VAMP2 de novo mutations highlights the key roles of this gene in human brain development and function.


Assuntos
Deficiência Intelectual/genética , Hipotonia Muscular/genética , Transtornos do Neurodesenvolvimento/genética , Neurônios/metabolismo , Sinapses/metabolismo , Proteína 2 Associada à Membrana da Vesícula/genética , Adolescente , Transtorno Autístico/genética , Transtorno Autístico/metabolismo , Encéfalo/diagnóstico por imagem , Criança , Pré-Escolar , Epilepsia/metabolismo , Exocitose , Feminino , Heterozigoto , Humanos , Lipídeos/química , Imageamento por Ressonância Magnética , Masculino , Fusão de Membrana , Transtornos dos Movimentos/genética , Mutação , Transtornos do Neurodesenvolvimento/metabolismo , Neurotransmissores/metabolismo , Fenótipo , Domínios Proteicos , Proteínas R-SNARE/metabolismo , Proteína 2 Associada à Membrana da Vesícula/fisiologia
6.
Cell Rep ; 21(1): 97-109, 2017 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-28978487

RESUMO

Mutations in the Golgi SNARE (SNAP [soluble NSF attachment protein] receptor) protein Membrin (encoded by the GOSR2 gene) cause progressive myoclonus epilepsy (PME). Membrin is a ubiquitous and essential protein mediating ER-to-Golgi membrane fusion. Thus, it is unclear how mutations in Membrin result in a disorder restricted to the nervous system. Here, we use a multi-layered strategy to elucidate the consequences of Membrin mutations from protein to neuron. We show that the pathogenic mutations cause partial reductions in SNARE-mediated membrane fusion. Importantly, these alterations were sufficient to profoundly impair dendritic growth in Drosophila models of GOSR2-PME. Furthermore, we show that Membrin mutations cause fragmentation of the presynaptic cytoskeleton coupled with transsynaptic instability and hyperactive neurotransmission. Our study highlights how dendritic growth is vulnerable even to subtle secretory pathway deficits, uncovers a role for Membrin in synaptic function, and provides a comprehensive explanatory basis for genotype-phenotype relationships in GOSR2-PME.


Assuntos
Dendritos/metabolismo , Mutação , Epilepsias Mioclônicas Progressivas/genética , Proteínas Qb-SNARE/genética , Via Secretória/genética , Sinapses/metabolismo , Animais , Dendritos/ultraestrutura , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Expressão Gênica , Estudos de Associação Genética , Complexo de Golgi/metabolismo , Humanos , Masculino , Fusão de Membrana , Epilepsias Mioclônicas Progressivas/metabolismo , Epilepsias Mioclônicas Progressivas/patologia , Fenótipo , Cultura Primária de Células , Proteínas Qb-SNARE/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Sinapses/patologia , Adulto Jovem
7.
Cell Rep ; 9(1): 206-218, 2014 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-25284778

RESUMO

Munc18-1 is a critical component of the core machinery controlling neuroexocytosis. Recently, mutations in Munc18-1 leading to the development of early infantile epileptic encephalopathy have been discovered. However, which degradative pathway controls Munc18-1 levels and how it impacts on neuroexocytosis in this pathology is unknown. Using neurosecretory cells deficient in Munc18, we show that a disease-linked mutation, C180Y, renders the protein unstable at 37°C. Although the mutated protein retains its function as t-SNARE chaperone, neuroexocytosis is impaired, a defect that can be rescued at a lower permissive temperature. We reveal that Munc18-1 undergoes K48-linked polyubiquitination, which is highly increased by the mutation, leading to proteasomal, but not lysosomal, degradation. Our data demonstrate that functional Munc18-1 levels are controlled through polyubiquitination and proteasomal degradation. The C180Y disease-causing mutation greatly potentiates this degradative pathway, rendering Munc18-1 unable to facilitate neuroexocytosis, a phenotype that is reversed at a permissive temperature.


Assuntos
Proteínas Munc18/genética , Proteínas Munc18/metabolismo , Mutação Puntual , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Exocitose/fisiologia , Humanos , Modelos Moleculares , Proteínas Munc18/química , Ligação Proteica , Estrutura Secundária de Proteína , Ratos , Proteínas SNARE/metabolismo , Ubiquitinação
8.
J Cell Sci ; 126(Pt 11): 2353-60, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23761923

RESUMO

Munc18-1 plays a dual role in transporting syntaxin-1A (Sx1a) to the plasma membrane and regulating SNARE-mediated membrane fusion. As impairment of either function leads to a common exocytic defect, assigning specific roles for various Munc18-1 domains has proved difficult. Structural analyses predict that a loop region in Munc18-1 domain 3a could catalyse the conversion of Sx1a from a 'closed', fusion-incompetent to an 'open', fusion-competent conformation. As this conversion occurs at the plasma membrane, mutations in this loop could potentially separate the chaperone and exocytic functions of Munc18-1. Expression of a Munc18-1 deletion mutant lacking 17 residues of the domain 3a loop (Munc18-1(Δ317-333)) in PC12 cells deficient in endogenous Munc18 (DKD-PC12 cells) fully rescued transport of Sx1a to the plasma membrane, but not exocytic secretory granule fusion. In vitro binding of Munc18-1(Δ317-333) to Sx1a was indistinguishable from that of full-length Munc18-1, consistent with the critical role of the closed conformation in Sx1a transport. However, in DKD-PC12 cells, Munc18-1(Δ317-333) binding to Sx1a was greatly reduced compared to that of full-length Munc18-1, suggesting that closed conformation binding contributes little to the overall interaction at the cell surface. Furthermore, we found that Munc18-1(Δ317-333) could bind SNARE complexes in vitro, suggesting that additional regulatory factors underpin the exocytic function of Munc18-1 in vivo. Together, these results point to a defined role for Munc18-1 in facilitating exocytosis linked to the loop region of domain 3a that is clearly distinct from its function in Sx1a transport.


Assuntos
Membrana Celular/metabolismo , Exocitose/fisiologia , Proteínas Munc18/metabolismo , Sintaxina 1/metabolismo , Animais , Membrana Celular/genética , Humanos , Proteínas Munc18/genética , Células PC12 , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Transporte Proteico/fisiologia , Ratos , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Sintaxina 1/genética
9.
J Cell Biol ; 200(3): 301-20, 2013 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-23382463

RESUMO

Before undergoing neuroexocytosis, secretory granules (SGs) are mobilized and tethered to the cortical actin network by an unknown mechanism. Using an SG pull-down assay and mass spectrometry, we found that myosin VI was recruited to SGs in a Ca(2+)-dependent manner. Interfering with myosin VI function in PC12 cells reduced the density of SGs near the plasma membrane without affecting their biogenesis. Myosin VI knockdown selectively impaired a late phase of exocytosis, consistent with a replenishment defect. This exocytic defect was selectively rescued by expression of the myosin VI small insert (SI) isoform, which efficiently tethered SGs to the cortical actin network. These myosin VI SI-specific effects were prevented by deletion of a c-Src kinase phosphorylation DYD motif, identified in silico. Myosin VI SI thus recruits SGs to the cortical actin network, potentially via c-Src phosphorylation, thereby maintaining an active pool of SGs near the plasma membrane.


Assuntos
Actinas/metabolismo , Exocitose , Cadeias Pesadas de Miosina/metabolismo , Vesículas Secretórias/metabolismo , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Bovinos , Técnicas de Silenciamento de Genes , Humanos , Espectrometria de Massas , Dados de Sequência Molecular , Cadeias Pesadas de Miosina/química , Neurônios/citologia , Neurônios/metabolismo , Células PC12 , Peptídeos/química , Fosforilação , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Ratos , Vesículas Secretórias/ultraestrutura , Quinases da Família src/metabolismo
10.
Mol Biol Cell ; 22(21): 4134-49, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21900502

RESUMO

Munc18-1 plays pleiotropic roles in neurosecretion by acting as 1) a molecular chaperone of syntaxin-1, 2) a mediator of dense-core vesicle docking, and 3) a priming factor for soluble N-ethylmaleimide-sensitive factor attachment protein receptor-mediated membrane fusion. However, how these functions are executed and whether they are correlated remains unclear. Here we analyzed the role of the domain-1 cleft of Munc18-1 by measuring the abilities of various mutants (D34N, D34N/M38V, K46E, E59K, K46E/E59K, K63E, and E66A) to bind and chaperone syntaxin-1 and to restore the docking and secretion of dense-core vesicles in Munc18-1/-2 double-knockdown cells. We identified striking correlations between the abilities of these mutants to bind and chaperone syntaxin-1 with their ability to restore vesicle docking and secretion. These results suggest that the domain-1 cleft of Munc18-1 is essential for binding to syntaxin-1 and thereby critical for its chaperoning, docking, and secretory functions. Our results demonstrate that the effect of the alleged priming mutants (E59K, D34N/M38V) on exocytosis can largely be explained by their reduced syntaxin-1-chaperoning functions. Finally, our data suggest that the intracellular expression and distribution of syntaxin-1 determines the level of dense-core vesicle docking.


Assuntos
Membrana Celular/metabolismo , Proteínas Munc18/metabolismo , Transporte Proteico , Vesículas Secretórias/metabolismo , Sintaxina 1/metabolismo , Substituição de Aminoácidos , Animais , Calorimetria , Expressão Gênica , Humanos , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteínas Munc18/genética , Norepinefrina/metabolismo , Células PC12 , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Ratos , Vesículas Secretórias/ultraestrutura , Sintaxina 1/genética , Termodinâmica , Titulometria
11.
J Neurochem ; 115(1): 1-10, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20681955

RESUMO

Munc18-1 plays essential roles in neurosecretion by interacting with syntaxin-1 and controlling the formation of the soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNARE) complex. At least three important functions of Munc18-1 have been proposed: (i) molecular chaperone of syntaxin-1 for appropriate localization and expression of syntaxin-1, (ii) priming/stimulation of the SNARE-mediated membrane fusion, and (iii) docking of large dense-core vesicles to the plasma membrane. Similarly, at least two different binding modes have been proposed for the interaction between Munc18-1 and syntaxin-1: (i) binary binding to a 'closed' conformation of syntaxin-1, and (ii) binding to the N-terminal peptide of syntaxin-1, which is thought to enable an interaction with the quaternary SNARE complex and/or further stabilize the binary interaction between Munc18-1 and closed syntaxin-1. Recent structural analyses have identified critical Munc18-1 residues implicated in these different modes of binding. These have recently been tested functionally in rescue experiments using Munc18-1 null neurons, chromaffin cells and Munc18-1/-2 knockdown PC12 cells, allowing remarkable progress to be made in the structural/functional understanding of Munc18-1. In this review, we summarize these recent advances and attempt to propose an updated model of the pleiotropic functions of Munc18-1 in neuroexocytosis.


Assuntos
Proteínas Munc18/genética , Proteínas Munc18/fisiologia , Neurossecreção/genética , Neurossecreção/fisiologia , Animais , Exocitose/genética , Exocitose/fisiologia , Humanos , Proteínas Munc18/química , Ligação Proteica , Conformação Proteica , Proteínas SNARE/genética , Proteínas SNARE/fisiologia , Sintaxina 1/genética , Sintaxina 1/fisiologia
12.
Mol Biol Cell ; 20(23): 4962-75, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19812250

RESUMO

Munc18-1 binds to syntaxin-1A via two distinct sites referred to as the "closed" conformation and N terminus binding. The latter has been shown to stimulate soluble N-ethylmaleimide-sensitive factor attachment protein receptor-mediated exocytosis, whereas the former is believed to be inhibitory or dispensable. To precisely define the contributions of each binding mode, we have engineered Munc18-1/-2 double knockdown neurosecretory cells and show that not only syntaxin-1A and -1B but also syntaxin-2 and -3 are significantly reduced as a result of Munc18-1 and -2 knockdown. Syntaxin-1 was mislocalized and the regulated secretion was abolished. We next examined the abilities of Munc18-1 mutants to rescue the defective phenotypes. Mutation (K46E/E59K) of Munc18-1 that selectively prevents binding to closed syntaxin-1 was unable to restore syntaxin-1 expression, localization, or secretion. In contrast, mutations (F115E/E132A) of Munc18-1 that selectively impair binding to the syntaxin-1 N terminus could still rescue the defective phenotypes. Our results indicate that Munc18-1 and -2 act in concert to support the expression of a broad range of syntaxins and to deliver syntaxin-1 to the plasma membrane. Our studies also indicate that the binding to the closed conformation of syntaxin is essential for Munc18-1 stimulatory action, whereas the binding to syntaxin N terminus plays a more limited role in neurosecretory cells.


Assuntos
Proteínas Munc18/química , Proteínas Munc18/metabolismo , Estrutura Terciária de Proteína , Proteínas Qa-SNARE/química , Proteínas Qa-SNARE/metabolismo , Animais , Sítios de Ligação , Técnicas de Silenciamento de Genes , Humanos , Modelos Moleculares , Proteínas Munc18/genética , Mutação , Células PC12 , Fenótipo , Ligação Proteica , Proteínas/genética , Proteínas/metabolismo , Proteínas Qa-SNARE/genética , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Vesículas Secretórias/metabolismo , Termodinâmica , Técnicas do Sistema de Duplo-Híbrido
13.
J Biol Chem ; 284(32): 21637-46, 2009 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-19483085

RESUMO

Neuronal communication relies on the fusion of neurotransmitter-containing vesicles with the plasma membrane. The soluble N-ethylmaleimide-sensitive fusion protein attachment protein receptor (SNARE) proteins initiate membrane fusion through the formation of the SNARE complex, a process tightly regulated by Sec1/Munc18-1 (SM) proteins. The emerging trend is that SM proteins promote SNARE-mediated membrane fusion by binding to a Syntaxin N-terminal motif. Here we report that mutations in the hydrophobic pocket of Munc18-1 (F115E and E132A), predicted to disrupt the N-terminal Sx1a interaction have a modest effect on binding to Sx1a in its free state, but abolish binding to the SNARE complex. Overexpression of the Munc18-1 mutant in PC12 cells lacking Munc18-1 rescues both neuroexocytosis and the plasma membrane localization of Syntaxin. However, total internal reflection fluorescence microscopy analysis reveals that expression of a Munc18-1 double mutant reduces the rate of vesicle fusion, an effect only detectable at the onset of stimulation. The Munc18-1 hydrophobic pocket is therefore critical for SNARE complex binding. However, mutations abrogating this interaction have a limited impact on Ca(2+)-dependent exocytosis in PC12 cells.


Assuntos
Proteínas Munc18/fisiologia , Proteínas SNARE/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Exocitose , Modelos Biológicos , Conformação Molecular , Dados de Sequência Molecular , Células PC12 , Estrutura Terciária de Proteína , Proteínas Qa-SNARE/metabolismo , Ratos , Proteínas Recombinantes/química
14.
J Chromatogr A ; 1127(1-2): 154-60, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16806241

RESUMO

An analytical HPLC method for the simultaneous determination of eight sulfonamides in swine wastewater was developed. The samples were collected from three states in Malaysia. Sample clean up was carried out by employing solid-phase extraction using a 60 mg Oasis HLB (Waters) cartridge with 3 ml reservoir. The HPLC column used was Supelcosil C18 (250 mm x 4.6mm I.D.) and elution was carried out using gradient mode. The mobile phases used were acetonitrile and 0.5% acetic acid in purified water. Antibiotics were detected using UV absorbance at 272 nm. Recoveries obtained for sulphanilamide ranged from 31.9+/-5.1% to 36.2+/-1.0%, while recoveries for other sulfa drugs studied were from 91.9+/-5.0% to 106.0+/-1.1%. The limit of quantitation (LOQ) for sulfamerazine, sulfamethazine and sulfamethoxypyridazine was 7.5 ng/L, while the LOQ for the other studied antibiotics was 5.0 ng/L. The method was used to analyse sulfonamides in wastewater collected from selected Malaysian swine facilities.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Sulfonamidas/análise , Poluentes Químicos da Água/análise , Criação de Animais Domésticos , Animais , Malásia , Reprodutibilidade dos Testes , Suínos , Eliminação de Resíduos Líquidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...