Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Neuropathol Appl Neurobiol ; 48(4): e12796, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35141929

RESUMEN

AIMS: Alzheimer's disease (AD) is characterised by amyloid-beta (Aß) aggregates in the brain. Targeting Aß aggregates is a major approach for AD therapies, although attempts have had little to no success so far. A novel treatment option is to focus on blocking the actual formation of Aß multimers. The enzyme tissue transglutaminase (TG2) is abundantly expressed in the human brain and plays a key role in post-translational modifications in Aß resulting in covalently cross-linked, stable and neurotoxic Aß oligomers. In vivo absence of TG2 in the APP23 mouse model may provide evidence that TG2 plays a key role in development and/or progression of Aß-related pathology. METHODS: Here, we compared the effects on Aß pathology in the presence or absence of TG2 using 12-month-old wild type, APP23 and a crossbreed of the TG2-/- mouse model and APP23 mice (APP23/TG2-/-). RESULTS: Using immunohistochemistry, we found that the number of Aß deposits was significantly reduced in the absence of TG2 compared with age-matched APP23 mice. To pinpoint possible TG2-associated mechanisms involved in this observation, we analysed soluble brain Aß1-40 , Aß1-42 and/or Aß40/42 ratio, and mRNA levels of human APP and TG2 family members present in brain of the various mouse models. In addition, using immunohistochemistry, both beta-pleated sheet formation in Aß deposits and the presence of reactive astrocytes associated with Aß deposits were analysed. CONCLUSIONS: We found that absence of TG2 reduces the formation of Aß pathology in the APP23 mouse model, suggesting that TG2 may be a suitable therapeutic target for reducing Aß deposition in AD.


Asunto(s)
Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Proteína Glutamina Gamma Glutamiltransferasa 2
2.
Anal Biochem ; 592: 113578, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31923381

RESUMEN

Self-interaction, chaperone binding and posttranslational modification of amyloid-beta (Aß) is essential in the initiation and propagation of Aß aggregation. Aggregation results in insoluble Aß deposits characteristic of Alzheimer's disease (AD) brain lesions, i.e. senile plaques and cerebral amyloid angiopathy. Tissue transglutaminase (tTG) is a calcium-dependent enzyme that catalyzes posttranslational modifications including the formation of covalent ε-(γ-glutamyl)lysine isopeptide bonds (molecular crosslinks), and colocalizes with Aß deposits in AD. Two independent groups recently found that apart from the induction of Aß oligomerization, the blood-derived transglutaminase member FXIIIa forms stable protein-protein complexes with Aß independent of the transamidation reaction. Here, we investigated whether also tTG forms rigid protein complexes with Aß in the absence of catalytic activation. We found that both Aß1-40 and Aß1-42 are substrates for tTG-catalyzed crosslinking. In addition, in the absence of calcium or the presence of a peptidergic inhibitor of tTG, stable tTG-Aß1-40 complexes were found. Interestingly, the stable complexes between tTG and Aß1-40, were only found at 'physiological' concentrations of Aß1-40. Together, our data suggest that depending on the Aß species at hand, and on the concentration of Aß, rigid protein-complexes are formed between tTG and Aß1-40 without the involvement of the crosslinking reaction.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Proteínas de Unión al GTP/metabolismo , Fragmentos de Péptidos/metabolismo , Transglutaminasas/metabolismo , Anciano , Anciano de 80 o más Años , Encéfalo/patología , Humanos , Agregación Patológica de Proteínas , Unión Proteica , Proteína Glutamina Gamma Glutamiltransferasa 2
3.
J Neurochem ; 134(6): 1116-28, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26088696

RESUMEN

Cerebral amyloid angiopathy (CAA) is a pathological hallmark of Alzheimer's disease (AD) and characterized by deposition of amyloid-ß (Aß) protein and smooth muscle cell (SMC) death in cerebral vessel walls. Apolipoprotein E (ApoE) is of importance in both Aß accumulation and Aß-mediated toxicity towards SMCs in the cerebral vessel wall, although its exact role in CAA pathogenesis remains unclear. Tissue transglutaminase (tTG) is an enzyme capable of inducing both protein complexes and altered protein bioactivity via post-translational cross-linking. In CAA, tTG and its catalytic activity are associated with deposited Aß. Furthermore, several apolipoproteins are known substrates of tTG. We therefore investigated whether ApoE is a substrate for tTG and if this affects ApoE's bioactivity. We found strong binding of different ApoE isoforms with tTG and demonstrated tTG-catalysed ApoE multimers. In post-mortem human AD cases, ApoE colocalized with in situ active tTG in CAA. Moreover, human brain SMCs treated with Aß demonstrated enhanced secretion of both ApoE and tTG, and of TG cross-links in the extracellular matrix. Interestingly, tTG-catalysed cross-linked ApoE failed to protect SMCs against Aß-mediated cytotoxicity. Together, our data demonstrate a novel tTG-driven post-translational modification of ApoE that might play an important role in CAA. Cerebral amyloid angiopathy (CAA) is a pathological hallmark of Alzheimer's disease (AD) and characterized by amyloid-ß (Aß) protein deposition and cerebral smooth muscle cell (SMC) death. We found that, in contrast to normal vessels, in CAA apolipoprotein E (ApoE) is cross-linked by tissue transglutaminase (tTG) resulting in stable ApoE complexes. These complexes no longer protect cerebral SMC from Aß-mediated toxicity. Our findings demonstrate a novel mechanism explaining the Aß-mediated cerebral SMC cell death characteristic of CAA in AD cases.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/efectos adversos , Apolipoproteínas E/metabolismo , Proteínas de Unión al GTP/metabolismo , Transglutaminasas/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Western Blotting , Supervivencia Celular , Angiopatía Amiloide Cerebral/metabolismo , Angiopatía Amiloide Cerebral/patología , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Músculo Liso Vascular/patología , Proteína Glutamina Gamma Glutamiltransferasa 2 , Isoformas de Proteínas/metabolismo , Resonancia por Plasmón de Superficie
4.
Sci Rep ; 13(1): 3481, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36859481

RESUMEN

Midlife hypertension is an important risk factor for cognitive impairment and dementia, including Alzheimer's disease. We investigated the effects of long-term treatment with two classes of antihypertensive drugs to determine whether diverging mechanisms of blood pressure lowering impact the brain differently. Spontaneously hypertensive rats (SHR) were either left untreated or treated with a calcium channel blocker (amlodipine) or beta blocker (atenolol) until one year of age. The normotensive Wistar Kyoto rat (WKY) was used as a reference group. Both drugs lowered blood pressure equally, while only atenolol decreased heart rate. Cerebrovascular resistance was increased in SHR, which was prevented by amlodipine but not atenolol. SHR showed a larger carotid artery diameter with impaired pulsatility, which was prevented by atenolol. Cerebral arteries demonstrated inward remodelling, stiffening and endothelial dysfunction in SHR. Both treatments similarly improved these parameters. MRI revealed that SHR have smaller brains with enlarged ventricles. In addition, neurofilament light levels were increased in cerebrospinal fluid of SHR. However, neither treatment affected these parameters. In conclusion, amlodipine and atenolol both lower blood pressure, but elicit a different hemodynamic profile. Both medications improve cerebral artery structure and function, but neither drug prevented indices of brain damage in this model of hypertension.


Asunto(s)
Hipertensión , Hipotensión , Ratas , Animales , Antihipertensivos , Ratas Endogámicas SHR , Atenolol , Amlodipino , Ratas Endogámicas WKY , Arteria Carótida Común
5.
Neurobiol Dis ; 45(3): 839-50, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22051113

RESUMEN

Parkinson's disease (PD) is characterized by accumulation of α-synuclein aggregates and degeneration of melanized, catecholaminergic neurons. The tissue transglutaminase (tTG) enzyme catalyzes molecular protein cross-linking. In PD, tTG levels are increased and cross-linking has been identified as an important factor in α-synuclein aggregation. In our quest to link tTGs distribution in the human brain to the hallmarks of PD pathology, we recently reported that catecholaminergic neurons in PD disease-affected brain areas display typical endoplasmic reticulum (ER) granules showing tTG immunoreactivity. In the present study, we set out to elucidate the nature of the interaction between tTG and the ER in PD pathogenesis, using retinoic-acid differentiated SH-SY5Y cells exposed to the PD-mimetic 1-methyl-4-phenylpyridinium (MPP(+)). Alike our observations in PD brain, MPP(+)-treated cells displayed typical TG-positive granules, that were also induced by other PD mimetics and by ER-stress inducing toxins. Additional immunocytochemical and biochemical investigation revealed that tTG is indeed associated to the ER, in particular at the cytoplasmic face of the ER. Upon MPP(+) exposure, additional recruitment of tTG toward the ER was found. In addition, we observed that MPP(+)-induced tTG activity results in transamidation of ER membrane proteins, like calnexin. Our data provide strong evidence for a, so far unrecognized, localization of tTG at the ER, at least in catecholaminergic neurons, and suggests that in PD activation of tTG may have a direct impact on ER function, in particular via post-translational modification of ER membrane proteins.


Asunto(s)
Retículo Endoplásmico/enzimología , Neuronas/ultraestructura , Transglutaminasas/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Calnexina/metabolismo , Calreticulina/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Proteínas del Citoesqueleto/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Retículo Endoplásmico/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Microscopía Electrónica de Transmisión , Neuroblastoma/patología , Neuroblastoma/ultraestructura , Neuronas/efectos de los fármacos , Proteína Disulfuro Isomerasas/metabolismo , Tapsigargina/farmacología , Tretinoina/farmacología , Tunicamicina/metabolismo
6.
Cells ; 11(3)2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-35159198

RESUMEN

Amyloid-beta (Aß) deposition in the brain is closely linked with the development of Alzheimer's disease (AD). Unfortunately, therapies specifically targeting Aß deposition have failed to reach their primary clinical endpoints, emphasizing the need to broaden the search strategy for alternative targets/mechanisms. Transglutaminase-2 (TG2) catalyzes post-translational modifications, is present in AD lesions and interacts with AD-associated proteins. However, an unbiased overview of TG2 interactors is lacking in both control and AD brain. Here we aimed to identify these interactors using a crossbreed of the AD-mimicking APP23 mouse model with wild type and TG2 knock-out (TG2-/-) mice. We found that absence of TG2 had no (statistically) significant effect on Aß pathology, soluble brain levels of Aß1-40 and Aß1-42, and mRNA levels of TG family members compared to APP23 mice at 18 months of age. Quantitative proteomics and network analysis revealed a large cluster of TG2 interactors involved in synaptic transmission/assembly and cell adhesion in the APP23 brain typical of AD. Comparative proteomics of wild type and TG2-/- brains revealed a TG2-linked pathological proteome consistent with alterations in both pathways. Our data show that TG2 deletion leads to considerable network alterations consistent with a TG2 role in (dys)regulation of synaptic transmission and cell adhesion in APP23 brains.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Proteína Glutamina Gamma Glutamiltransferasa 2
7.
Acta Neuropathol Commun ; 7(1): 206, 2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31829283

RESUMEN

Multiple Sclerosis (MS) is the most common cause of acquired neurological disability in young adults, pathologically characterized by leukocyte infiltration of the central nervous system, demyelination of the white and grey matter, and subsequent axonal loss. Microglia are proposed to play a role in MS lesion formation, however previous literature has not been able to distinguish infiltrated macrophages from microglia. Therefore, in this study we utilize the microglia-specific, homeostatic markers TMEM119 and P2RY12 to characterize their immunoreactivity in MS grey matter lesions in comparison to white matter lesions. Furthermore, we assessed the immunological status of the white and grey matter lesions, as well as the responsivity of human white and grey matter derived microglia to inflammatory mediators. We are the first to show that white and grey matter lesions in post-mortem human material differ in their immunoreactivity for the homeostatic microglia-specific markers TMEM119 and P2RY12. In particular, whereas immunoreactivity for TMEM119 and P2RY12 is decreased in the center of WMLs, immunoreactivity for both markers is not altered in GMLs. Based on data from post-mortem human microglia cultures, treated with IL-4 or IFNγ+LPS and on  counts of CD3+ or CD20+ lymphocytes in lesions, we show that downregulation of TMEM119 and P2RY12  immunoreactivity in MS lesions corresponds with the presence of lymphocytes and lymphocyte-derived cytokines within the parenchyma but not in  the meninges. Furthermore, the presence of TMEM119+ and partly P2RY12+ microglia in pre-active lesions as well as in  the rim of active white and grey matter lesions, in addition to TMEM119+ and P2RY12+ rod-like microglia in subpial grey matter lesions suggest that blocking the entrance of lymphocytes into the CNS of MS patients may not interfere with all possible effects of TMEM119+ and P2RY12+ microglia in both white and grey matter MS lesions.


Asunto(s)
Sustancia Gris/metabolismo , Proteínas de la Membrana/metabolismo , Microglía/metabolismo , Esclerosis Múltiple/metabolismo , Receptores Purinérgicos P2Y12/metabolismo , Sustancia Blanca/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Sustancia Gris/química , Sustancia Gris/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Masculino , Proteínas de la Membrana/análisis , Microglía/química , Microglía/patología , Persona de Mediana Edad , Esclerosis Múltiple/patología , Receptores Purinérgicos P2Y12/análisis , Sustancia Blanca/química , Sustancia Blanca/patología
8.
PLoS One ; 13(4): e0196433, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29689097

RESUMEN

The neurodegenerative disease multiple sclerosis (MS) is pathologically characterized by the massive influx of immune cells into the central nervous system. This contributes to demyelination and axonal damage which causes symptoms such as motor and cognitive dysfunctions. The migration of leukocytes from the blood vessel is orchestrated by a multitude of factors whose determination is essential in reducing cellular influx in MS patients and the experimental autoimmune encephalomyelitis (EAE) animal model. The here studied enzyme tissue Transglutaminase (TG2) is present intracellularly, on the cell surface and extracellularly. There it contributes to cellular adhesion and migration via its transamidation activity and possibly by facilitating cellular interaction with the extracellular matrix. Previous data from our group showed reduced motor symptoms and cellular infiltration after using a pharmacological TG2 transamidation activity inhibitor in a rat EAE model. However, it remained elusive if the cross-linking activity of the enzyme resulted in the observed effects. To follow-up, we now characterized two new small molecule TG2 activity inhibitors, BJJF078 and ERW1041E. Both compounds are potent inhibitor of recombinant human and mouse Transglutaminase enzyme activity, mainly TG2 and the close related enzyme TG1. In addition they did not affect the binding of TG2 to the extracellular matrix substrate fibronectin, a process via which TG2 promotes cellular adhesion and migration. We found, that ERW1041E but not BJJF078 resulted in reduced EAE disease motor-symptoms while neither caused apparent changes in pathology (cellular influx), Transglutaminase activity or expression of inflammation related markers in the spinal cord, compared to vehicle treated controls. Although we cannot exclude issues on bioavailability and in vivo efficacy of the used compounds, we hypothesize that extracellular TG1/TG2 activity is of greater importance than (intra-)cellular activity in mouse EAE pathology.


Asunto(s)
Antiinflamatorios/farmacología , Benzamidas/farmacología , Encefalomielitis Autoinmune Experimental/patología , Inhibidores Enzimáticos/farmacología , Proteínas de Unión al GTP/antagonistas & inhibidores , Isoxazoles/farmacología , Monocitos/efectos de los fármacos , Esclerosis Múltiple/patología , Naftalenos/farmacología , Piperidinas/farmacología , Pirrolidinas/farmacología , Quinolinas/farmacología , Transglutaminasas/antagonistas & inhibidores , Animales , Antiinflamatorios/química , Antiinflamatorios/uso terapéutico , Benzamidas/química , Benzamidas/uso terapéutico , Células Cultivadas , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/uso terapéutico , Fibronectinas/metabolismo , Proteínas de Unión al GTP/metabolismo , Humanos , Isoxazoles/química , Isoxazoles/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Monocitos/patología , Esclerosis Múltiple/tratamiento farmacológico , Naftalenos/química , Naftalenos/uso terapéutico , Piperidinas/química , Piperidinas/uso terapéutico , Unión Proteica/efectos de los fármacos , Proteína Glutamina Gamma Glutamiltransferasa 2 , Pirrolidinas/química , Pirrolidinas/uso terapéutico , Quinolinas/química , Quinolinas/uso terapéutico , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/patología , Transglutaminasas/metabolismo
10.
Nucl Med Biol ; 44: 90-104, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27837727

RESUMEN

INTRODUCTION: The protein-protein crosslinking activity of the enzyme tissue transglutaminase (TG2; EC 2.3.2.13) is associated with the pathogenesis of various diseases, including celiac disease, lung-, liver- and kidney fibrosis, cancer and neurodegenerative diseases. This study aims at developing a TG2 PET tracer based on the peptidic irreversible TG2 inhibitor Z006. METHODS: Initially, the carbon-11 labeling of Z006 at the diazoketone position was explored. Subsequently, a set of analogues that allow for fluorine-18 labeling was synthesized. Two potent analogues, 6f and 6g, were radiolabeled with fluorine-18 and biodistribution and metabolite analysis in Wistar rats was performed. The identity of the main metabolite of [18F]6g was elucidated using LC-MS/MS. In vitro binding to isolated TG2 and in vitro autoradiography on MDA-MB-231 breast cancer tissue using [18F]6g was performed. RESULTS: [18F]6f and [18F]6g were obtained in 20 and 9% yields, respectively. Following administration to healthy Wistar rats, rapid metabolism of both tracers was observed. Remarkably, full conversion to just one single metabolite was observed for one of the tracers, [18F]6g. By LC-MS/MS analysis this metabolite was identified as C-terminally saponified [18F]6g. This metabolite was also found to be a potent TG2 inhibitor in vitro. In vitro binding to isolated TG2 and in vitro autoradiography on MDA-MB-231 tumor sections using [18F]6g demonstrated high specific and selective binding of [18F]6g to active TG2. CONCLUSIONS: Whereas based on the intensive metabolism [18F]6f seems unsuitable as a TG2 PET tracer, the results warrant further evaluation of [18F]6gin vivo.


Asunto(s)
Radioisótopos de Flúor , Proteínas de Unión al GTP/metabolismo , Péptidos/química , Tomografía de Emisión de Positrones/métodos , Transglutaminasas/metabolismo , Acilación , Animales , Proteínas de Unión al GTP/antagonistas & inhibidores , Marcaje Isotópico , Masculino , Péptidos/farmacocinética , Péptidos/farmacología , Proteína Glutamina Gamma Glutamiltransferasa 2 , Ratas , Ratas Wistar , Distribución Tisular , Transglutaminasas/antagonistas & inhibidores
11.
Nucl Med Biol ; 43(4): 232-42, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27067043

RESUMEN

INTRODUCTION: Tissue transglutaminase (TG2) is a ubiquitously expressed enzyme capable of forming metabolically and mechanically stable crosslinks between the γ-carboxamide of a glutamine acyl-acceptor substrate and the ε-amino functionality of a lysine acyl-donor substrate resulting in protein oligomers. High TG2 crosslinking activity has been implicated in the pathogenesis of various diseases including celiac disease, cancer and fibrotic and neurodegenerative diseases. Development of a PET tracer specific for active TG2 provides a novel tool to further investigate TG2 biology in vivo in disease states. Recently, potent irreversible active site TG2 inhibitors carrying an acrylamide warhead were synthesized and pharmacologically characterized. METHODS: Three of these inhibitors, compound 1, 2 and 3, were successfully radiolabeled with carbon-11 on the acrylamide carbonyl position using a palladium mediated [(11)C]CO aminocarbonylation reaction. Ex vivo biodistribution and plasma stability were evaluated in healthy Wistar rats. Autoradiography was performed on MDA-MB-231 tumor sections. RESULTS: [(11)C]1, -2 and -3 were obtained in decay corrected radiochemical yields of 38-55%. Biodistribution showed low uptake in peripheral tissues, with the exception of liver and kidney. Low brain uptake of <0.05% ID/g was observed. Blood plasma analysis demonstrated that [(11)C]1 and [(11)C]2 were rapidly metabolized, whereas [(11)C]3 was metabolized at a more moderate rate (63.2 ± 6.8 and 28.7 ± 10.8% intact tracer after 15 and 45 min, respectively). Autoradiography with [(11)C]3 on MDA-MB-231 tumor sections showed selective and specific binding of the radiotracer to the active state of TG2. CONCLUSIONS: Taken together, these results identify [(11)C]3 as the most promising of the three compounds tested for development as PET radiotracer for the in vivo investigation of TG2 activity.


Asunto(s)
Amidas/química , Radioisótopos de Carbono , Inhibidores Enzimáticos/química , Proteínas de Unión al GTP/análisis , Proteínas de Unión al GTP/metabolismo , Tomografía de Emisión de Positrones/métodos , Transglutaminasas/análisis , Transglutaminasas/metabolismo , Amidas/metabolismo , Amidas/farmacocinética , Animales , Línea Celular Tumoral , Estabilidad de Medicamentos , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Proteínas de Unión al GTP/antagonistas & inhibidores , Humanos , Marcaje Isotópico , Masculino , Proteína Glutamina Gamma Glutamiltransferasa 2 , Radioquímica , Ratas , Ratas Wistar , Distribución Tisular , Transglutaminasas/antagonistas & inhibidores
12.
Free Radic Res ; 36(6): 695-9, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12180195

RESUMEN

alpha-Lipoic acid (LA), an antioxidant with broad neuroprotective capacity, is thought to act by scavenging reactive oxygen species and stimulation of glutathione synthesis. LA shows structural resemblance to dithiolethiones, like anethole dithiolethione (ADT). ADT protects against oxidative damage, primarily by induction of phase II detoxication enzymes, in particular NAD(P)H:quinone oxidoreductase (NQO1) and glutathione-S-transferase (GST). Therefore, we investigated whether LA, like ADT, is capable also of inducing these protective enzymes. Our data show that LA, like ADT, induces a highly significant, time- and concentration dependent, increase in the activity of NQO1 and GST in C6 astroglial cells. The LA or ADT mediated induction of NQO1 was further confirmed by quantitative PCR and western blot analysis. This work for the first time unequivocally demonstrates LA mediated upregulation of phase II detoxication enzymes, which may highly contribute to the compounds' neuroprotective potential. Moreover, the data support the notion of a common mechanism of action of LA and ADT.


Asunto(s)
Antioxidantes/farmacología , Astrocitos/enzimología , Fármacos Neuroprotectores/farmacología , Ácido Tióctico/farmacología , Anetol Tritiona/farmacología , Animales , Astrocitos/efectos de los fármacos , Astrocitoma/enzimología , Neoplasias del Sistema Nervioso Central/enzimología , Glutatión Transferasa/efectos de los fármacos , Glutatión Transferasa/metabolismo , Inactivación Metabólica , NAD(P)H Deshidrogenasa (Quinona)/efectos de los fármacos , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Oxidorreductasas/efectos de los fármacos , Oxidorreductasas/metabolismo , Ratas , Transferasas/efectos de los fármacos , Transferasas/metabolismo , Células Tumorales Cultivadas
13.
Neurochem Int ; 62(4): 486-91, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23380076

RESUMEN

Tissue transglutaminase (tTG) is a cross-linking enzyme involved in protein aggregation during Parkinson's disease (PD) pathogenesis. Autophagy is inhibited by tTG activation via a mechanism in which cross-linking of beclin 1, an autophagy initiator at the level of the endoplasmic reticulum (ER), has been implicated. We reported increased tTG protein levels and activity at the ER in both PD brain and in a PD-mimicking cell system. Here we characterized the interaction between tTG and beclin 1 at the ER membrane and the role of tTG in reduced autophagy in an in vitro model of PD, using differentiated SH-SY5Y neurons treated with the PD-mimic MPP(+). We found that under PD-mimicking conditions, beclin 1 and tTG partially colocalized at the ER, beclin 1 levels increased at the ER, and tTG readily cross-linked beclin 1 which was prevented by enzymatic blockade of tTG. Under these conditions, accumulation of beclin 1 at the ER was enhanced by inhibition of tTG activity. In line with these observations and the role of beclin 1 in autophagy, levels of the autophagy marker protein LC3II in MPP(+)-treated cells, were significantly increased by inhibition of tTG activity. Our data provide first evidence for a role of tTG-mediated regulation of beclin 1 and autophagy in MPP(+)-treated human SH-SY5Y cells.


Asunto(s)
1-Metil-4-fenilpiridinio/toxicidad , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/fisiología , Proteínas de Unión al GTP/fisiología , Proteínas de la Membrana/metabolismo , Transglutaminasas/fisiología , Autofagia/efectos de los fármacos , Beclina-1 , Línea Celular Tumoral , Retículo Endoplásmico/metabolismo , Proteínas de Unión al GTP/metabolismo , Humanos , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transglutaminasas/metabolismo
14.
Neurochem Int ; 58(7): 785-93, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21440023

RESUMEN

Transamidation of α-synuclein by the Ca(2+)-dependent enzyme tissue transglutaminase (tTG, EC 2.3.2.13) is implicated in Parkinson's disease (PD). tTG may therefore offer a novel therapeutic target to intervene in PD. Here we first evaluated the potency and efficacy of three recently developed irreversible active-site inhibitors of tTG (B003, Z006 and KCC009) to inhibit tTG activity in vitro and in living cells. In vitro, all compounds were found to be full inhibitors of tTG activity showing a rank order of potency (defined by IC-50 values) of Z006>B003>KCC009. Upon Ca(2+) ionophore (A23187) induced activation of cellular tTG (measured by incorporation of the tTG-specific amine substrate 5-(biotinamido)pentylamine (BAP) into cellular proteins) in neuroblastoma SH-SY5Y cells, only Z006 (0.3-30 µM) retained the capacity to completely inhibit tTG activity. Under these conditions B003 (3-300 µM) only partially blocked tTG activity whereas KCC009 (3-100 µM) failed to affect tTG activity at any of the concentrations used. Z006 (30 µM) also blocked the tTG mediated incorporation of BAP into α-synuclein monomers and SDS-resistant multimers in vitro and in α-synuclein overexpressing SHSY5Y cells exposed to A23187 or the PD mimetic 1-methyl-4-phenylpyridine (MPP(+)). Moreover, Z006 (30 µM) substantially reduced formation of SDS-resistant α-synuclein multimers in SH-SY5Y cells exposed to A23187 or MPP(+) in the absence of BAP. We conclude that α-synuclein is a cellular substrate for tTG under conditions mimicking PD and blockade of tTG activity counteracts α-synuclein transamidation and aggregation in vitro and in living cells. Moreover, our cell model appears an excellent readout to identify candidate inhibitors of intracellular tTG.


Asunto(s)
Amidas/metabolismo , Proteínas de Unión al GTP/antagonistas & inhibidores , Modelos Biológicos , Enfermedad de Parkinson/metabolismo , Transglutaminasas/antagonistas & inhibidores , alfa-Sinucleína/metabolismo , Western Blotting , Línea Celular Tumoral , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Proteínas de Unión al GTP/metabolismo , Humanos , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transglutaminasas/metabolismo
15.
PLoS One ; 6(9): e25037, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21949843

RESUMEN

An important neuropathological feature of neuroinflammatory processes that occur during e.g. Multiple Sclerosis (MS) is the formation of an astroglial scar. Astroglial scar formation is facilitated by the interaction between astrocytes and extracellular matrix proteins (ECM) such as fibronectin. Since there is evidence indicating that glial scars strongly inhibit both axon growth and (re)myelination in brain lesions, it is important to understand the factors that contribute to the interaction between astrocytes and ECM proteins. Tissue Transglutaminase (TG2) is a multifunctional enzyme with an ubiquitous tissue distribution, being clearly present within the brain. It has been shown that inflammatory cytokines can enhance TG2 activity. In addition, TG2 can mediate cell adhesion and migration and it binds fibronectin with high affinity. We therefore hypothesized that TG2 is involved in astrocyte-fibronectin interactions. Our studies using primary rat astrocytes show that intracellular and cell surface expression and activity of TG2 is increased after treatment with pro-inflammatory cytokines. Astrocyte-derived TG2 interacts with fibronectin and is involved in astrocyte adhesion onto and migration across fibronectin. TG2 is involved in stimulating focal adhesion formation which is necessary for the interaction of astrocytes with ECM proteins. We conclude that astrocyte-derived TG2 contributes to the interaction between astrocytes and fibronectin. It might thereby regulate ECM remodeling and possibly glial scarring.


Asunto(s)
Astrocitos/metabolismo , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Fibronectinas/metabolismo , Transglutaminasas/metabolismo , Animales , Western Blotting , Membrana Celular/metabolismo , Proliferación Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Matriz Extracelular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteínas de Unión al GTP , Técnicas para Inmunoenzimas , Inmunoprecipitación , Proteína Glutamina Gamma Glutamiltransferasa 2 , ARN Mensajero/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transglutaminasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA