Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Toxins (Basel) ; 15(11)2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37999510

RESUMEN

The cyanobacterial non-protein amino acid (AA) ß-Methylamino-L-alanine (BMAA) is considered to be a neurotoxin. BMAA caused histopathological changes in brains and spinal cords of primates consistent with some of those seen in early motor neuron disease; however, supplementation with L-serine protected against some of those changes. We examined the impact of BMAA on AA concentrations in human neuroblastoma cells in vitro. Cells were treated with 1000 µM BMAA and intracellular free AA concentrations in treated and control cells were compared at six time-points over a 48 h culture period. BMAA had a profound effect on intracellular AA levels at specific time points but in most cases, AA homeostasis was re-established in the cell. The most heavily impacted amino acid was serine which was depleted in BMAA-treated cells from 9 h onwards. Correction of serine depletion could be a factor in the observation that supplementation with L-serine protects against BMAA toxicity in vitro and in vivo. AAs that could potentially be involved in protection against BMAA-induced oxidation such as histidine, tyrosine, and phenylalanine were depleted in cells at later time points.


Asunto(s)
Aminoácidos Diaminos , Neuroblastoma , Animales , Humanos , Aminoácidos , Aminoácidos Diaminos/toxicidad , Aminoácidos Diaminos/metabolismo , Serina/farmacología , Neurotoxinas/toxicidad
2.
J Chromatogr A ; 1655: 462530, 2021 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-34517165

RESUMEN

LC-MS/MS method development for native amino acid detection can be problematic due to low ionisation efficiencies, in source fragmentation, potential for cluster ion formation and incorrect application of chromatography techniques. This has led to the majority of the scientific community derivatising amino acids for more sensitive analysis. Derivatisation has several benefits including reduced signal-to-noise ratios, more efficient ionisation, and a change in polarity, allowing the use of reverse phase chromatography. However, derivatisation of amino acids can be expensive, requires additional sample preparation steps, is more time consuming and increases sample instability, due to the most derivatised amino acids only be stable for finite amount of time. While showing initial promise, development of reliable hydrophilic interaction liquid chromatography (HILIC) separation methods has presented difficulties for the analyst including irreproducible separation and poor sensitivity. This study aimed to find a means to improve the detection sensitivity of the 20 protein amino acids by HILIC-MS/MS. We describe the use of previously undescribed amino acid-acetonitrile (ACN) adducts to improve detection of 16 out of the 20 amino acids. While all amino acids examined did form an ACN adduct, 4 had low intensity adduct formation compared to their protonated state, 3 of which are classified as basic amino acids. For 15 of the 20 amino acids tested, we used the ACN adduct for both quantification and qualification ions and demonstrated a significant enhancement in signal-to-noise ratio, ranging from 23 to 1762% improvement. Lower LODs, LOQs and lower ranges of linearity were also achieved for these amino acids. The optimised method was applied to a human neuroblastoma cell line (SH-SY5Y) with the potential to be applied to other complex sample types. The improved sensitivity this method offers simplifies sample preparation and reduces the costs of amino acid analysis compared to those methods that rely on derivatisation for sensitivity.


Asunto(s)
Aminoácidos , Espectrometría de Masas en Tándem , Acetonitrilos , Cromatografía Liquida , Humanos , Interacciones Hidrofóbicas e Hidrofílicas
3.
Amino Acids ; 53(9): 1351-1359, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34283312

RESUMEN

The cyanobacterial non-protein amino acid α-amino-ß-methylaminopropionic acid, more commonly known as BMAA, was first discovered in the seeds of the ancient gymnosperm Cycad circinalis (now Cycas micronesica Hill). BMAA was linked to the high incidence of neurological disorders on the island of Guam first reported in the 1950s. BMAA still attracts interest as a possible causative factor in amyotrophic lateral sclerosis (ALS) following the identification of ALS disease clusters associated with living in proximity to lakes with regular cyanobacterial blooms. Since its discovery, BMAA toxicity has been the subject of many in vivo and in vitro studies. A number of mechanisms of toxicity have been proposed including an agonist effect at glutamate receptors, competition with cysteine for transport system xc_ and other mechanisms capable of generating cellular oxidative stress. In addition, a wide range of studies have reported effects related to disturbances in proteostasis including endoplasmic reticulum stress and activation of the unfolded protein response. In the present studies we examine the effects of BMAA on the ubiquitin-proteasome system (UPS) and on chaperone-mediated autophagy (CMA) by measuring levels of ubiquitinated proteins and lamp2a protein levels in a differentiated neuronal cell line exposed to BMAA. The BMAA induced increases in oxidised proteins and the increase in CMA activity reported could be prevented by co-administration of L-serine but not by the two antioxidants examined. These data provide further evidence of a protective role for L-serine against the deleterious effects of BMAA.


Asunto(s)
Aminoácidos Diaminos/efectos adversos , Autofagia Mediada por Chaperones , Toxinas de Cianobacterias/efectos adversos , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Neuroblastoma/tratamiento farmacológico , Agregado de Proteínas/efectos de los fármacos , Serina/farmacología , Ubiquitina/metabolismo , Antioxidantes/farmacología , Diferenciación Celular , Agonistas de Aminoácidos Excitadores/efectos adversos , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Estrés Oxidativo , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Células Tumorales Cultivadas
4.
Res Microbiol ; 172(6): 103852, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34246779

RESUMEN

In contrast to mammalian cells, bacteria such as Escherichia coli have been shown to display tolerance towards the neurotoxin ß-methylamino-l-alanine (BMAA) suggesting that these prokaryotes possess a way to metabolise BMAA or its products, resulting in their export, degradation, or detoxification. Single gene deletion mutants of E. coli K-12 with inactivated amino acid biosynthesis pathways were treated with 500 µg/ml BMAA and the resulting growth was monitored. Wild type E. coli and most of the gene deletion mutants displayed unaltered growth in the presence of BMAA over 12 h. Conversely, deletion of genes in the cysteine biosynthesis pathway, cysE, cysK or cysM resulted in a BMAA dose-dependent growth delay in minimal medium. Through further studies of the ΔcysE strain, we observed increased susceptibility to oxidative stress from H2O2 in minimal medium, and disruptions in glutathione levels and oxidation state. The cysteine biosynthesis pathway is therefore linked to the tolerance of BMAA and oxidative stress in E. coli, which potentially represents a mechanism of BMAA detoxification.


Asunto(s)
Aminoácidos Diaminos/farmacología , Toxinas de Cianobacterias/farmacología , Cisteína/biosíntesis , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Aminoácidos Diaminos/metabolismo , Aminoácidos Diaminos/toxicidad , Medios de Cultivo , Toxinas de Cianobacterias/metabolismo , Toxinas de Cianobacterias/toxicidad , Cisteína Sintasa/genética , Tolerancia a Medicamentos , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Proteínas de Escherichia coli/genética , Eliminación de Gen , Glutatión/metabolismo , Peróxido de Hidrógeno/farmacología , Redes y Vías Metabólicas , Oxidación-Reducción , Estrés Oxidativo , Serina O-Acetiltransferasa/genética
5.
Int J Biochem Cell Biol ; 117: 105624, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31654750

RESUMEN

In Parkinson's disease (PD), as in many other neurodegenerative disorders, mitochondrial dysfunction, protein misfolding, and proteotoxic stress underly the disease process. For decades, the primary symptomatic treatment for PD has been the dopamine precursor L-DOPA (Levodopa). L-DOPA however can initiate protein misfolding through its ability to mimic the protein amino acid L-tyrosine, resulting in random errors in aminoacylation and L-DOPA becoming mistakenly inserted into the polypeptide chain of proteins in place of L-tyrosine. In the present study we examined the impact that the generation of DOPA-containing proteins had on human neuroblastoma cell (SH-SY5Y) function in vitro. We showed that even in the presence of antioxidants there was a significant accumulation of cytosolic ubiquitin in DOPA-treated cells, an upregulation in the endosomal-lysosomal degradation system, deleterious changes to mitochondrial morphology and a marked decline in mitochondrial function.The effects of L-DOPA on mitochondrial function were not observed with D-DOPA, the stereoisomer of L-DOPA that cannot be inserted into proteins so did not result from oxidative stress. We could fully protect against these effects by co-treatment with L-tyrosine, supporting the view that misincorporation of L-DOPA into proteins contributed to these cytotoxic effects, leading us to suggest that co-treatment with L-tyrosine could be beneficial therapeutically.


Asunto(s)
Levodopa/toxicidad , Mitocondrias/patología , Enfermedad de Parkinson/tratamiento farmacológico , Humanos , Levodopa/farmacología
6.
Amino Acids ; 51(8): 1221-1232, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31302779

RESUMEN

In addition to the 20 protein amino acids that are vital to human health, hundreds of naturally occurring amino acids, known as non-proteinogenic amino acids (NPAAs), exist and can enter the human food chain. Some NPAAs are toxic through their ability to mimic protein amino acids and this property is utilised by NPAA-containing plants to inhibit the growth of other plants or kill herbivores. The NPAA L-azetidine-2-carboxylic acid (Aze) enters the food chain through the use of sugar beet (Beta vulgaris) by-products as feed in the livestock industry and may also be found in sugar beet by-product fibre supplements. Aze mimics the protein amino acid L-proline and readily misincorporates into proteins. In light of this, we examined the toxicity of Aze to mammalian cells in vitro. We showed decreased viability in Aze-exposed cells with both apoptotic and necrotic cell death. This was accompanied by alterations in endosomal-lysosomal activity, changes to mitochondrial morphology and a significant decline in mitochondrial function. In summary, the results show that Aze exposure can lead to deleterious effects on human neuron-like cells and highlight the importance of monitoring human Aze consumption via the food chain.


Asunto(s)
Ácido Azetidinocarboxílico/farmacología , Muerte Celular , Dieta , Mitocondrias/patología , Neuroblastoma/patología , Humanos , Mitocondrias/efectos de los fármacos , Neuroblastoma/tratamiento farmacológico , Células Tumorales Cultivadas
7.
Toxicol In Vitro ; 56: 163-171, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30703532

RESUMEN

In addition to the 20 protein amino acids that are encoded for protein synthesis, hundreds of other naturally occurring amino acids, known as non-proteinogenic amino acids (NPAAs) exist. It is well known that some NPAAs are toxic through their ability to mimic protein amino acids, either in protein synthesis or in other metabolic pathways, and this property is utilised by some plants to inhibit the growth of other plants or kill herbivores. L-norvaline is an NPAA readily available for purchase as a dietary supplement. In light of previous evidence of l-norvaline's antifungal, antimicrobial and herbicidal activity, we examined the toxicity of l-norvaline to mammalian cells in vitro and showed that l-norvaline decreased cell viability at concentrations as low as 125 µM, caused necrotic cell death and significant changes to mitochondrial morphology and function. Furthermore, toxicity was reduced in the presence of structurally similar 'protein' amino acids, suggesting l-norvaline's cytotoxicity could be attributed to protein amino acid mimicry.


Asunto(s)
Suplementos Dietéticos/toxicidad , Valina/análogos & derivados , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ácido Glutámico/metabolismo , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Valina/toxicidad
8.
Amino Acids ; 50(3-4): 397-407, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29235019

RESUMEN

There is a strong body of evidence linking the non-protein amino acid (NPAA) ß-methylamino-L-alanine (BMAA) to the development of a number of neurodegenerative diseases. BMAA has been found globally, is produced by a number of organisms including cyanobacteria, diatoms, and dinoflagellates; and has been shown to biomagnify through trophic levels. The role of BMAA in neurodegenerative disease is highlighted by its presence in the brains of a number of neurodegenerative disease patients, where it was found in a protein-bound form. We have previously shown that BMAA is bound to cell proteins, and results in the upregulation of the unfolded protein response, an endoplasmic reticulum stress response activated by the presence of misfolded proteins within the cell. Structurally aberrant proteins are features of a number of neurodegenerative diseases, and further investigation of how BMAA interacts with proteins is crucial to our understanding of its toxicity. Here we use radiolabelled BMAA to investigate the interaction and binding of BMAA to eukaryotic and prokaryotic proteins. We found differences in the presence and distribution of protein-bound BMAA between E. coli and neuroblastoma cells, with an increase in binding over time only seen in the eukaryotic cells. We also found that BMAA was unable to bind to pure proteins, or cell lysate in native or denaturing conditions, indicating that biological processing is required for BMAA to bind to proteins.


Asunto(s)
Aminoácidos Diaminos/metabolismo , Encéfalo/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Proteínas/metabolismo , Aminoácidos Diaminos/toxicidad , Animales , Encéfalo/patología , Cianobacterias/genética , Cianobacterias/metabolismo , Toxinas de Cianobacterias , Diatomeas/genética , Diatomeas/metabolismo , Dinoflagelados/genética , Dinoflagelados/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Células Eucariotas/metabolismo , Humanos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Células Procariotas/metabolismo
9.
Neurotox Res ; 33(1): 33-42, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28634653

RESUMEN

The non-protein amino acid (NPAA) ß-methylamino-L-alanine (BMAA) is produced by a diverse range of cyanobacteria, diatoms and dinoflagellates, and is present in both aquatic and terrestrial ecosystems globally. Exposure to BMAA has been implicated in the development of neurodegenerative diseases including amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD) and Parkinson's disease (PD). BMAA is often found in nature along with its structural isomers 2,4-diaminobutyric acid (2,4-DAB) and aminoethylglycine (AEG); however, the toxicity of these NPAAs in combination has not been examined. We have previously demonstrated that BMAA induces endoplasmic reticulum (ER) stress and increases caspase and cathepsin activity in human neuroblastoma cells (SH-SY5Y), effects consistent with proteotoxic stress due to disturbances in protein synthesis, folding or turnover. The current study investigates whether 2,4-DAB and AEG share a similar mechanism of toxicity to BMAA, and if simultaneous exposure of cells to BMAA and its isomers results in increased toxicity in vitro. We show that a 48-h treatment with both 500 µM BMAA and 2,4-DAB decreases cell viability in vitro whereas AEG was not cytotoxic under the same conditions. Treatment of SH-SY5Y cells with 2,4-DAB did not increase expression of ER stress markers. Combined treatment of cells with BMAA and 2,4-DAB resulted in increased caspase activity and increased apoptosis above that of BMAA or 2,4-DAB on their own. These results suggest that 2,4-DAB does not share the same mechanism of toxicity as BMAA but the presence of 2,4-DAB increases the toxicity of BMAA to human cells in vitro.


Asunto(s)
Aminoácidos Diaminos/farmacología , Aminobutiratos/farmacología , Apoptosis/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neurotoxinas/farmacología , Caspasas/metabolismo , Catepsinas/metabolismo , Línea Celular Transformada , Supervivencia Celular , Toxinas de Cianobacterias , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Complejo IV de Transporte de Electrones/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Neuroblastoma/patología , ARN Mensajero/metabolismo , Factores de Tiempo , Transaminasas/metabolismo , Factor de Transcripción CHOP/metabolismo
10.
Toxicon ; 109: 7-12, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26559613

RESUMEN

ß-methylamino-L-alanine (BMAA), a non-protein amino acid synthesised by cyanobacteria, has been linked to a complex neurological disorder on Guam and more recently to other cases of sporadic ALS (sALS), however the mechanisms of BMAA toxicity are not completely understood. We have previously demonstrated that BMAA is misincorporated into newly synthesised proteins by human neuroblastoma cells and fibroblasts, resulting in the formation of autofluorescent material and the induction of apoptotic cell death. In the present study we show that BMAA at low levels does not cause an acute toxicity in neuroblastoma cells but increases the expression of the ER stress marker, C/EBP homologous protein (CHOP) and increases the activity of the pro-apoptotic enzyme caspase-3. We also observed an increase in the activity of the lysosomal cysteine proteases cathepsin B and L, characteristic of the accumulation of proteins in the lysosomal system. We were able to prevent these proteotoxic effects in neuroblastoma cells through co-treatment with l-serine suggesting that they resulted from incorporation of BMAA into proteins. Misincorporation provides a possible mechanism whereby BMAA could initiate misfolding, and the accumulation of aggregate-prone proteins in neurons. This build-up of misfolded proteins could explain the long latency period of the disease previously reported on Guam.


Asunto(s)
Aminoácidos Diaminos/toxicidad , Serina/farmacología , Catepsina B/metabolismo , Línea Celular Tumoral , Toxinas de Cianobacterias , Humanos , Técnicas In Vitro
11.
Exp Neurol ; 238(1): 29-37, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22001774

RESUMEN

Levodopa (L-DOPA), a close structural analogue of the protein amino acid L-tyrosine, can substitute for L-tyrosine in protein synthesis and be mistakenly incorporated into newly synthesised proteins in vitro. We show that L-DOPA-containing proteins are present in the brain in L-DOPA-treated Parkinson's disease patients and accumulate in specific brain regions. In vitro studies demonstrate that substitution of L-tyrosine residues in proteins with L-DOPA causes protein misfolding and promotes protein aggregation in SH-SY5Y neuroblastoma cells resulting in the appearance of autofluorescent bodies. We show that the presence of L-DOPA-containing proteins causes profound changes in mitochondria and stimulates the formation of autophagic vacuoles in cells. Unlike L-DOPA, which is toxic to cells through its ability to generate radicals, proteins containing incorporated L-DOPA are toxic to SH-SY5Y cells by a mechanism independent of oxidative stress and resistant to antioxidants. These data suggest that the accumulation of L-DOPA-containing proteins in vulnerable cells might negatively impact on cell function.


Asunto(s)
Antiparkinsonianos/metabolismo , Antiparkinsonianos/toxicidad , Química Encefálica/fisiología , Levodopa/metabolismo , Levodopa/toxicidad , Proteínas del Tejido Nervioso/metabolismo , Neurotoxinas/toxicidad , Enfermedad de Parkinson/metabolismo , Apoptosis/efectos de los fármacos , Electroforesis de las Proteínas Sanguíneas , Química Encefálica/efectos de los fármacos , Caspasas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Fragmentación del ADN , Humanos , Hidrólisis , Indicadores y Reactivos , L-Lactato Deshidrogenasa/metabolismo , Microscopía Electrónica , Microscopía Fluorescente , Membranas Mitocondriales/efectos de los fármacos , Oxidación-Reducción , Enfermedad de Parkinson/tratamiento farmacológico , Quinolinas
12.
Cardiovasc Res ; 91(1): 37-44, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21354994

RESUMEN

AIMS: Plasma concentrations of high-density lipoprotein (HDL)-cholesterol correlate inversely with the incidence of myocardial infarction in humans. We investigated the effect of treatment with human apolipoprotein A-I (apoA-I), the principal protein of HDL, on plaque disruption in an animal model. METHODS AND RESULTS: Seventy apolipoprotein E knockout mice were induced to develop atherosclerotic lesions in the brachiocephalic artery by feeding a high-fat diet for 9 weeks. Mice then received twice-weekly treatment with human apoA-I (8 mg/kg) or vehicle, for 2 weeks. The incidence of acute plaque disruption was reduced by 65% in mice receiving apoA-I (P < 0.01). Plaques in treated mice had a more stable phenotype, with an increase in smooth muscle cell (SMC): macrophage ratio (P = 0.05), principally the consequence of an increase in the number of SMC in plaques. In the fibrous cap, there were reductions in matrix metalloproteinase-13 (-69%, P < 0.0001) and S100A4, a marker of SMC de-differentiation (-60%, P < 0.0001). These results indicate that 2 weeks of treatment with small amounts of human apoA-I produces more stable plaques in a mouse model. CONCLUSION: Treatment with apoA-I has the potential to stabilize plaques and prevent plaque rupture in humans.


Asunto(s)
Apolipoproteína A-I/administración & dosificación , Aterosclerosis/tratamiento farmacológico , Tronco Braquiocefálico/efectos de los fármacos , Fármacos Cardiovasculares/administración & dosificación , Placa Aterosclerótica/tratamiento farmacológico , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/sangre , Aterosclerosis/genética , Aterosclerosis/patología , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patología , Desdiferenciación Celular , HDL-Colesterol/sangre , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Humanos , Mediadores de Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Metaloproteinasa 13 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Placa Aterosclerótica/sangre , Placa Aterosclerótica/genética , Placa Aterosclerótica/patología , Rotura Espontánea , Proteína de Unión al Calcio S100A4 , Proteínas S100/metabolismo
13.
Free Radic Biol Med ; 50(2): 389-99, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21111806

RESUMEN

Proteins are major biological targets for oxidative damage within cells because of their high abundance and rapid rates of reaction with radicals and singlet oxygen. These reactions generate high yields of hydroperoxides. The turnover of both native and modified/damaged proteins is critical for maintaining cell homeostasis, with this occurring via the proteasomal and endosomal-lysosomal systems; the former is of particular importance for intracellular proteins. In this study we have examined whether oxidation products generated on amino acids, peptides, and proteins modulate 26S proteasome activity. We show that oxidation products, and particularly protein hydroperoxides, are efficient inhibitors of the 26S proteasome tryptic and chymotryptic activities, with this depending, at least in part, on the presence of hydroperoxide groups. Removal of these species by reduction significantly reduces proteasome inhibition. This loss of activity is accompanied by a loss of thiol residues, but an absence of radical formation, consistent with molecular, rather than radical, reactions being responsible for proteasome inhibition. Aldehydes also seem to play a role in the inhibition of chymotryptic activity, with this prevented by treatment with NaBH(4), which reduces these groups. Inhibition occurred at hydroperoxide concentrations of ≥1µM for oxidized amino acids and peptides and ≥10µM for oxidized proteins, compared with ca. 100µM for H(2)O(2), indicating that H(2)O(2) is a much less effective inhibitor. These data indicate that the formation of oxidized proteins within cells may modulate cell function by interfering with the turnover of native proteins and the clearance of modified materials.


Asunto(s)
Aminoácidos/metabolismo , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/farmacología , Fragmentos de Péptidos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas/metabolismo , Aminoácidos/química , Animales , Células Cultivadas , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Peróxido de Hidrógeno/química , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Oxidantes/farmacología , Oxidación-Reducción , Fragmentos de Péptidos/química , Proteínas/química
14.
Int J Biochem Cell Biol ; 40(8): 1452-66, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18329946

RESUMEN

Despite astounding diversity in their structure and function, proteins are constructed from 22 protein or 'canonical' amino acids. Hundreds of amino acid analogues exist; many occur naturally in plants, some are synthetically produced or can be produced in vivo by oxidation of amino acid side-chains. Certain structural analogues of the protein amino acids can escape detection by the cellular machinery for protein synthesis and become misincorporated into the growing polypeptide chain of proteins to generate non-native proteins. In this review we seek to provide a comprehensive overview of the current knowledge on the biosynthetic incorporation of amino acid analogues into proteins by mammalian cells. We highlight factors influencing their incorporation and how the non-native proteins generated can alter cell function. We examine the ability of amino acid analogues, representing those commonly found in damaged proteins in pathological tissues, to be misincorporated into proteins by cells in vitro, providing us with a useful tool in the laboratory to generate modified proteins representing those present in a wide-range of pathologies. We also discuss the evidence for amino acid analogue incorporation in vivo and its association with autoimmune symptoms. We confine the review to studies in which the synthetic machinery of cell has not been modified to accept non-protein amino acids.


Asunto(s)
Aminoácidos/metabolismo , Biosíntesis de Proteínas , Aminoácidos Diaminos/metabolismo , Aminoácidos Diaminos/toxicidad , Aminoacilación , Animales , Bacterias/metabolismo , Canavanina/metabolismo , Canavanina/toxicidad , Etionina/metabolismo , Humanos , Levodopa/metabolismo , Levodopa/uso terapéutico , Levodopa/toxicidad , Lupus Eritematoso Sistémico/inducido químicamente , Oxidación-Reducción , Enfermedad de Parkinson/tratamiento farmacológico , Plantas/metabolismo , Triptófano/análogos & derivados , Triptófano/metabolismo , Triptófano/toxicidad
15.
Biochem J ; 398(2): 197-206, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16671891

RESUMEN

Hyperglycaemia, triose phosphate decomposition and oxidation reactions generate reactive aldehydes in vivo. These compounds react non-enzymatically with protein side chains and N-terminal amino groups to give adducts and cross-links, and hence modified proteins. Previous studies have shown that free or protein-bound carbonyls inactivate glyceraldehyde-3-phosphate dehydrogenase with concomitant loss of thiol groups [Morgan, Dean and Davies (2002) Arch. Biochem. Biophys. 403, 259-269]. It was therefore hypothesized that modification of lysosomal cysteine proteases (and the structurally related enzyme papain) by free and protein-bound carbonyls may modulate the activity of these components of the cellular proteolytic machinery responsible for the removal of modified proteins and thereby contribute to a decreased removal of modified proteins from cells. It is shown that MGX (methylglyoxal), GO (glyoxal) and glycolaldehyde, but not hydroxyacetone and glucose, inhibit catB (cathepsin B), catL (cathepsin L) and catS (cathepsin S) activity in macrophage cell lysates, in a concentration-dependent manner. Protein-bound carbonyls produced similar inhibition with both cell lysates and intact macrophage cells. Inhibition was also observed with papain, with this paralleled by loss of the active site cysteine residue and formation of the adduct species S-carboxymethylcysteine, from GO, in a concentration-dependent manner. Inhibition of autolysis of papain by MGX, along with cross-link formation, was detected by SDS/PAGE. Treatment of papain and catS with the dialdehyde o-phthalaldehyde resulted in enzyme inactivation and an intra-molecular active site cysteine-lysine cross-link. These results demonstrate that reactive aldehydes inhibit cysteine proteases by modification of the active site cysteine residue. This process may contribute to the accumulation of modified proteins in tissues of people with diabetes and age-related pathologies, including atherosclerosis, cataract and Alzheimer's disease.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Animales , Sitios de Unión , Carbocisteína/farmacología , Catepsinas/aislamiento & purificación , Catepsinas/metabolismo , Bovinos , Línea Celular , Cisteína Endopeptidasas/genética , Activación Enzimática/efectos de los fármacos , Glicosilación , Glioxal/farmacología , Humanos , Ratones , Peso Molecular , Papaína/aislamiento & purificación , Papaína/metabolismo , Unión Proteica , o-Ftalaldehído/farmacología
16.
Free Radic Biol Med ; 40(9): 1539-48, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16632114

RESUMEN

Reaction of radicals in the presence of O2, and singlet oxygen, with some amino acids, peptides, and proteins yields hydroperoxides. These species are key intermediates in chain reactions and protein damage. Previously we have shown that peptide and protein hydroperoxides react rapidly with thiols, and that this can result in inactivation of thiol-dependent enzymes. The major route for the cellular removal of damaged proteins is via catabolism mediated by proteosomal and lysosomal pathways; cysteine proteases (cathepsins) play a key role in the latter system. We hypothesized that inactivation of cysteine proteases by hydroperoxide-containing oxidised proteins may contribute to the accumulation of modified proteins within cells. We show here that thiol-dependent cathepsins, either isolated or in cell lysates, are rapidly and efficiently inactivated by amino acid, peptide, and protein hydroperoxides in a time- and concentration-dependent manner; this occurs with similar efficacy to equimolar H2O2. Inactivation involves reaction of the hydroperoxide with Cys residues as evidenced by thiol loss and formation of sulfenic acid intermediates. Structurally related, non-thiol-dependent cathepsins are less readily inactivated by these hydroperoxides. This inhibition, by oxidized proteins, of the system designed to remove modified proteins, may contribute to the accumulation of damaged proteins in cells subject to oxidative stress.


Asunto(s)
Aminoácidos/metabolismo , Catepsinas/metabolismo , Peróxido de Hidrógeno/metabolismo , Péptido Hidrolasas/metabolismo , Péptidos/metabolismo , Aminoácidos/química , Animales , Línea Celular , Radicales Libres/química , Radicales Libres/metabolismo , Macrófagos/metabolismo , Ratones , Oxidación-Reducción , Péptido Hidrolasas/química , Péptidos/química
17.
Arterioscler Thromb Vasc Biol ; 26(4): 851-6, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16410454

RESUMEN

OBJECTIVE: Lysosomal proteinases have been implicated in a number of pathologies associated with extracellular matrix breakdown. Therefore, we investigated the possibility that the lysosomal proteinase cathepsin S may be involved in atherosclerotic plaque destabilization. METHODS AND RESULTS: Atherosclerotic plaques in the brachiocephalic arteries of fat-fed apolipoprotein E/cathepsin S double knockout mice had 73% fewer acute plaque ruptures (P=0.026) and were 46% smaller (P=0.025) than those in age-, strain-, and sex-matched apolipoprotein E single knockout controls. When the incidence of acute plaque rupture was normalized for plaque size, the reduction in the double knockouts was 72% (P=0.039). The number of buried fibrous layers, indicative of an unstable plaque phenotype, was reduced by 67% in the double knockouts (P=0.008). The cysteine proteinase inhibitor, egg white cystatin, was biotinylated and used as an active-site-directed probe for cathepsins. Biotinylated cystatin selectively detected cathepsin S in extracts of human carotid atherosclerotic plaque. Active cathepsin S was detectable in extracts of human atherosclerotic plaque but not in nondiseased carotid arteries. Active cathepsins were especially prominent in macrophages in the shoulder regions of plaques, areas considered to be vulnerable to rupture. Cathepsin S protein colocalized with regions of elastin degradation in human coronary plaques. CONCLUSIONS: These data provide direct evidence that an endogenous proteinase, cathepsin S, plays an important role in atherosclerotic plaque destabilization and rupture.


Asunto(s)
Apolipoproteínas E , Aterosclerosis/patología , Catepsinas , Animales , Apolipoproteínas E/deficiencia , Aterosclerosis/genética , Tronco Braquiocefálico/patología , Catepsinas/deficiencia , Catepsinas/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Rotura Espontánea/genética , Rotura Espontánea/patología
18.
FEBS J ; 272(12): 3162-71, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15955073

RESUMEN

An Escherichia coli cell-free transcription/translation system was used to explore the high-level incorporation of L-3,4-dihydroxyphenylalanine (DOPA) into proteins by replacing tyrosine with DOPA in the reaction mixtures. ESI-MS showed specific incorporation of DOPA in place of tyrosine. More than 90% DOPA incorporation at each tyrosine site was achieved, allowing the recording of clean 15N-HSQC NMR spectra. A redox-staining method specific for DOPA was shown to provide a sensitive and generally applicable method for assessing the cell-free production of proteins. Of four proteins produced in soluble form in the presence of tyrosine, two resulted in insoluble aggregates in the presence of high levels of DOPA. DOPA has been found in human proteins, often in association with various disease states that implicate protein aggregation and/or misfolding. Our results suggest that misfolded and aggregated proteins may result, in principle, from ribosome-mediated misincorporation of intracellular DOPA accumulated due to oxidative stress. High-yield cell-free protein expression systems are uniquely suited to obtain rapid information on solubility and aggregation of nascent polypeptide chains.


Asunto(s)
Dihidroxifenilalanina/metabolismo , Biosíntesis de Proteínas , Proteínas/metabolismo , Secuencia de Aminoácidos , Sistema Libre de Células , Cromatografía Líquida de Alta Presión , Ciclofilinas/genética , Ciclofilinas/metabolismo , Dihidroxifenilalanina/química , Escherichia coli , Histidina/genética , Espectroscopía de Resonancia Magnética , Datos de Secuencia Molecular , Péptidos/análisis , Isomerasa de Peptidilprolil/genética , Isomerasa de Peptidilprolil/metabolismo , Ingeniería de Proteínas/métodos , Proteínas/química , Espectrometría de Masa por Ionización de Electrospray , Tirosina/metabolismo
19.
Free Radic Biol Med ; 37(11): 1756-64, 2004 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-15528035

RESUMEN

Protein-bound 3,4-dihydroxyphenylalanine (PB-DOPA) is a major product of hydroxyl radical attack on tyrosine residues of proteins. Levels of PB-DOPA in cells and tissues have been shown to be greatly elevated in age-related diseases. We demonstrate for the first time that l-DOPA (levodopa) can be biosynthetically incorporated into cell proteins by human cells (THP-1 monocytes and monocyte-derived macrophages). The DOPA-containing proteins generated were selectively visualized on PVDF membranes using a redox-cycling staining method. Many cell proteins contained DOPA and seemed to be synthesized as their full-length forms. The cellular removal of DOPA-containing proteins by THP-1 cells was by proteolysis involving both the proteasomal and the lysosomal systems. The rate of cellular proteolysis of DOPA-containing proteins increased at lower levels of DOPA incorporation but decreased at higher levels of DOPA incorporation. The decreased rate of degradation was accompanied by an increase in the activity of cathepsins B and L but the activity of cathepsin S increased only at lower levels of DOPA incorporation. These data raise the possibility that PB-DOPA could be generated in vivo from l-DOPA, which is the most widely used treatment for Parkinson disease.


Asunto(s)
Dihidroxifenilalanina/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , Proteínas/metabolismo , Catepsinas/metabolismo , Catepsinas/fisiología , Línea Celular , Dihidroxifenilalanina/análisis , Humanos , Lisosomas/fisiología , Macrófagos/química , Monocitos/química , Enfermedad de Parkinson/metabolismo , Complejo de la Endopetidasa Proteasomal/fisiología , Biosíntesis de Proteínas , Proteínas/química
20.
Int J Biochem Cell Biol ; 35(5): 716-27, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12672463

RESUMEN

The ubiquitin-proteasome pathway is a major route of degradation of cell proteins. It also plays an essential role in maintaining cell homeostasis by degrading many rate-limiting enzymes and critical regulatory proteins. Alterations in proteasome activity have been implicated in a number of pathologies including Parkinson's disease, Alzheimer's disease and diabetes. The eukaryotic proteasome is a multicatalytic protease characterized by three activities with distinct specificities against peptide substrates. Although substrates were identified which could selectively measure the individual activities in the purified proteasome little data is available on how specific those substrates are for proteasomal activity when used with biological samples which may contain many other active peptidases. Here we examine the three major peptidase activities in lysates of two cell types and in a liver cytosol fraction in the presence of specific proteasome inhibitors and after fractionation by gel permeation chromatography. We demonstrate that other proteinases present in these preparations can degrade the commonly used proteasome substrates under the standard assay conditions. We develop a simple method for separating the proteasome from the lower molecular weight proteases using a 500kDa molecular weight cut-off membrane. This allows proteasome activity to be accurately measured in crude biological samples and may have quite broad applicability. We also identify low molecular weight tryptic activity in both the cell and tissue preparations which could not be inhibited by the proteasome inhibitor epoxomycin but was inhibitable by two cysteine proteinase inhibitors and by lactacystin suggesting that lactacystin may not be completely proteasome specific.


Asunto(s)
Acetilcisteína/análogos & derivados , Cisteína Endopeptidasas/metabolismo , Complejos Multienzimáticos/metabolismo , Péptido Hidrolasas/metabolismo , Ubiquitina/metabolismo , Acetilcisteína/farmacología , Animales , Línea Celular , Cromatografía en Gel , Inhibidores de Cisteína Proteinasa/farmacología , Humanos , Leupeptinas/farmacología , Ratones , Complejos Multienzimáticos/antagonistas & inhibidores , Oligopéptidos/farmacología , Complejo de la Endopetidasa Proteasomal , Ratas , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA