Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Cell Sci ; 129(20): 3792-3802, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27566163

RESUMO

Membrane-associated glycoprotein neural cell adhesion molecule (NCAM) and its polysialylated form (PSA-NCAM) play an important role in brain plasticity by regulating cell-cell interactions. Here, we demonstrate that the cytosolic serine protease prolyl endopeptidase (PREP) is able to regulate NCAM and PSA-NCAM. Using a SH-SY5Y neuroblastoma cell line with stable overexpression of PREP, we found a remarkable loss of PSA-NCAM, reduced levels of NCAM180 and NCAM140 protein species, and a significant increase in the NCAM immunoreactive band migrating at an apparent molecular weight of 120 kDa in PREP-overexpressing cells. Moreover, increased levels of NCAM fragments were found in the concentrated medium derived from PREP-overexpressing cells. PREP overexpression selectively induced an activation of matrix metalloproteinase-9 (MMP-9), which could be involved in the observed degradation of NCAM, as MMP-9 neutralization reduced the levels of NCAM fragments in cell culture medium. We propose that increased PREP levels promote epidermal growth factor receptor (EGFR) signaling, which in turn activates MMP-9. In conclusion, our findings provide evidence for newly-discovered roles for PREP in mechanisms regulating cellular plasticity through NCAM and PSA-NCAM.


Assuntos
Moléculas de Adesão de Célula Nervosa/metabolismo , Proteólise , Serina Endopeptidases/metabolismo , Animais , Anticorpos Neutralizantes/metabolismo , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células Cultivadas , Meios de Cultura , Receptores ErbB/metabolismo , Técnicas de Silenciamento de Genes , Imuno-Histoquímica , Metaloproteinase 9 da Matriz/metabolismo , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neuroblastoma/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos , Prolil Oligopeptidases , Proteólise/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes/farmacologia , Ácidos Siálicos/metabolismo , Sialiltransferases/metabolismo
2.
Biol Chem ; 397(1): 45-55, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26351917

RESUMO

Secretory peptides and proteins are frequently modified by pyroglutamic acid (pE, pGlu) at their N-terminus. This modification is catalyzed by the glutaminyl cyclases QC and isoQC. Here, we decipher the roles of the isoenzymes by characterization of IsoQC-/- mice. These mice show a significant reduction of glutaminyl cyclase activity in brain and peripheral tissue, suggesting ubiquitous expression of the isoQC enzyme. An assay of substrate conversion in vivo reveals impaired generation of the pGlu-modified C-C chemokine ligand 2 (CCL2, MCP-1) in isoQC-/- mice. The pGlu-formation was also impaired in primary neurons, which express significant levels of QC. Interestingly, however, the formation of the neuropeptide hormone thyrotropin-releasing hormone (TRH), assessed by immunohistochemistry and hormonal analysis of hypothalamic-pituitary-thyroid axis, was not affected in isoQC-/-, which contrasts to QC-/-. Thus, the results reveal differential functions of isoQC and QC in the formation of the pGlu-peptides CCL2 and TRH. Substrates requiring extensive prohormone processing in secretory granules, such as TRH, are primarily converted by QC. In contrast, protein substrates such as CCL2 appear to be primarily converted by isoQC. The results provide a new example, how subtle differences in subcellular localization of enzymes and substrate precursor maturation might influence pGlu-product formation.


Assuntos
Aminoaciltransferases/metabolismo , Administração Oral , Aminoaciltransferases/deficiência , Animais , Células Cultivadas , Glucose/administração & dosagem , Teste de Tolerância a Glucose , Inflamação/induzido quimicamente , Inflamação/metabolismo , Isoenzimas/metabolismo , Lipopolissacarídeos/administração & dosagem , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Ácido Pirrolidonocarboxílico/metabolismo , Especificidade por Substrato
3.
Acta Neuropathol ; 129(4): 565-83, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25666182

RESUMO

The brains of Alzheimer's disease (AD) patients are characterized by deposits of Abeta peptides and by accompanying chronic inflammation. Here, we provide evidence that the enzyme isoglutaminyl cyclase (isoQC) is a novel factor contributing to both aspects of AD pathology. Two putative substrates of isoQC, N-truncated Abeta peptides and the monocyte chemoattractant chemokine CCL2, undergo isoQC-catalyzed pyroglutamate (pGlu) modification. This triggers Abeta aggregation and facilitates the biological activity of CCL2, which collectively results in the formation of high molecular weight Abeta aggregates, glial cell activation, neuroinflammation and neuronal cell death. In mouse brain, we found isoQC to be neuron-specifically expressed in neocortical, hippocampal and subcortical structures, localized to the endoplasmic reticulum and Golgi apparatus as well as co-expressed with its substrate CCL2. In aged APP transgenic Tg2576 mice, both isoQC and CCL2 mRNA levels are up-regulated and isoQC and CCL2 proteins were found to be co-induced in Abeta plaque-associated reactive astrocytes. Also, in mouse primary astrocyte culture, a simultaneous up-regulation of isoQC and CCL2 expression was revealed upon Abeta and pGlu-Abeta stimulation. In brains of AD patients, the expression of isoQC and CCL2 mRNA and protein is up-regulated compared to controls and correlates with pGlu-Abeta load and with the decline in mini-mental state examination. Our observations provide evidence for a dual involvement of isoQC in AD pathogenesis by catalysis of pGlu-Abeta and pGlu-CCL2 formation which mutually stimulate inflammatory events and affect cognition. We conclude that isoQC inhibition may target both major pathological events in the development of AD.


Assuntos
Doença de Alzheimer/patologia , Aminoaciltransferases/metabolismo , Encéfalo/metabolismo , Quimiocina CCL2/metabolismo , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Aminoaciltransferases/genética , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Encéfalo/patologia , Células Cultivadas , Quimiocina CCL2/genética , Modelos Animais de Doenças , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fragmentos de Peptídeos/metabolismo , Fatores de Tempo , Regulação para Cima/genética
4.
Biochim Biophys Acta ; 1852(1): 146-55, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25446989

RESUMO

Recently, Aß peptide variants with an N-terminal truncation and pyroglutamate modification were identified and shown to be highly neurotoxic and prone to aggregation. This modification of Aß is catalyzed by glutaminyl cyclase (QC) and pharmacological inhibition of QC diminishes Aß deposition and accompanying gliosis and ameliorates memory impairment in transgenic mouse models of Alzheimer's disease (AD). QC expression was initially described in the hypothalamus, where thyrotropin-releasing hormone (TRH) is one of its physiological substrates. In addition to its hormonal role, a novel neuroprotective function of TRH following excitotoxicity and Aß-mediated neurotoxicity has been reported in the hippocampus. Functionally matching this finding, we recently demonstrated QC expression by hippocampal interneurons in mouse brain. Here, we detected neuronal co-expression of QC and TRH in the hippocampus of young adult wild type mice using double immunofluorescence labeling. This provides evidence for TRH being a physiological QC substrate in hippocampus. Additionally, in neocortex of aged but not of young mice transgenic for amyloid precursor protein an increase of QC mRNA levels was found compared to wild type littermates. This phenomenon was not observed in hippocampus, which is later affected by Aß pathology. However, in hippocampus of transgenic - but not of wild type mice - a correlation between QC and TRH mRNA levels was revealed. This co-regulation of the enzyme QC and its substrate TRH was reflected by a co-induction of both proteins in reactive astrocytes in proximity of Aß deposits. Also, in primary mouse astrocytes a co-induction of QC and TRH was demonstrated upon Aß stimulation.


Assuntos
Aminoaciltransferases/metabolismo , Astrócitos/enzimologia , Hipocampo/enzimologia , Neurônios/enzimologia , Hormônio Liberador de Tireotropina/metabolismo , Aminoaciltransferases/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Sequência de Bases , Primers do DNA , Hipocampo/citologia , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , Especificidade por Substrato , Hormônio Liberador de Tireotropina/genética
5.
Int J Dev Neurosci ; 36: 64-73, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24886834

RESUMO

Glutaminyl cyclases (QCs) catalyze the formation of pyroglutamate (pGlu) from glutamine precursors at the N-terminus of a number of peptide hormones, neuropeptides and chemokines. This post-translational modification stabilizes these peptides, protects them from proteolytical degradation or is important for their biological activity. However, QC is also involved in a pathogenic pGlu modification of peptides accumulating in protein aggregation disorders such as Alzheimer's disease and familial Danish and familial British dementia. Its isoenzyme (isoQC) was shown to contribute to aspects of inflammation by pGlu-modifying and thereby stabilizing the monocyte chemoattractant protein CCL2. For the generation of respective animal models and for pharmacological treatment studies the characterization of the mouse strain and brain region-specific expression of QC and isoQC is indispensible. In order to address this issue, we used enzymatic activity assays and specific antibodies to detect both QC variants by immunohistochemistry in nine different mouse strains. Comparing different brain regions, the highest enzymatic QC/isoQC activity was detected in ventral brain, followed by cortex and hippocampus. Immunohistochemical stainings revealed that QC/isoQC activity in cortex mostly arises from isoQC expression. For most brain regions, the highest QC/isoQC activity was detected in C3H and FVB mice, whereas low QC/isoQC activity was present in CD1, SJL and C57 mice. Quantification of QC- and isoQC-immunoreactive cells by unbiased stereology revealed a higher abundance of isoQC- than of QC-immunoreactive neurons in Edinger-Westphal nucleus and in substantia nigra. In the locus coeruleus, however, there were comparable densities of QC- and of isoQC-immunoreactive neurons. These observations are of considerable importance with regard to the selection of appropriate mouse strains for the study of QC/isoQC relevance in mouse models of neurodegeneration and neuroinflammation and for the testing of therapeutical interventions in these models.


Assuntos
Aminoaciltransferases/metabolismo , Encéfalo/enzimologia , Animais , Encéfalo/anatomia & histologia , Camundongos , Camundongos Knockout , Especificidade da Espécie , Especificidade por Substrato
6.
J Alzheimers Dis ; 39(2): 385-400, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24164736

RESUMO

Brains of Alzheimer's disease (AD) patients are characterized in part by the formation of high molecular weight aggregates of amyloid-ß (Aß) peptides, which interfere with neuronal function and provoke neuronal cell death. The pyroglutamate (pGlu) modification of Aß was demonstrated to be catalyzed by the enzyme glutaminyl cyclase (QC) and to enhance pathogenicity and neurotoxicity. Here, we addressed the role of QC in AD pathogenesis in human cortex. Two sets of human postmortem brain tissue from a total of 13 non-demented controls and 11 AD cases were analyzed by immunohistochemistry and unbiased stereology, quantitative RT-PCR, and enzymatic activity assays for the expression level of QC in temporal and entorhinal cortex. Additionally, cortical Aß and pGlu-Aß concentrations were quantified by ELISA. Data on QC expression and Aß peptide concentrations were correlated with each other and with the Mini-Mental State Examination (MMSE) of individual cases. In control cases, QC expression was higher in the more vulnerable entorhinal cortex than in temporal cortex. In AD brains, QC mRNA expression and the immunoreactivity of QC were increased in both cortical regions and frequently associated with pGlu-Aß deposits. The analyses of individual cases revealed significant correlations between QC mRNA levels and the concentration of insoluble pGlu-Aß aggregates, but not of unmodified Aß peptides. Elevated pGlu-Aß load showed a better correlation with the decline in MMSE than elevated concentration of unmodified Aß. Our observations provide evidence for an involvement of QC in AD pathogenesis and cognitive decline by QC-catalyzed pGlu-Aß formation.


Assuntos
Doença de Alzheimer/metabolismo , Aminoaciltransferases/metabolismo , Peptídeos beta-Amiloides/metabolismo , Transtornos Cognitivos/metabolismo , Córtex Entorrinal/metabolismo , Ácido Pirrolidonocarboxílico/metabolismo , Lobo Temporal/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/complicações , Doença de Alzheimer/diagnóstico , Aminoaciltransferases/genética , Transtornos Cognitivos/diagnóstico , Transtornos Cognitivos/etiologia , Feminino , Humanos , Masculino , Neurônios/metabolismo , Escalas de Graduação Psiquiátrica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
7.
Int J Dev Neurosci ; 31(1): 36-45, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23070049

RESUMO

The amyloid precursor protein (APP) can be proteolytically degraded via non-amyloidogenic α-secretase and amyloidogenic ß-secretase pathways. Previously, we have identified the presynaptic protein Munc13-1 as a diacylglycerol/phorbolester (DAG/PE) receptor that contributes to secretory, non-amyloidogenic APP processing after PE stimulation. Here, we used organotypic brain slice cultures from wild-type mice and from Munc13-1 knock-out (KO), Munc13-2 KO and Munc13-1/2 double KO (DKO) mice for pharmacological stimulation experiments. First, we demonstrate that neuronal populations and synaptic components important for secretory APP processing develop normally in organotypic brain slice cultures of all genotypes analyzed. Blockade of voltage-gated Na(+) channels by tetrodotoxin reduced the PE-stimulated secretory APP processing, whereas depolarization by high extracellular K(+) concentration evoked APP secretion. Additionally, the PE-stimulated APP secretion from Munc13-1 KO brain slices was significantly lower than that from wild-type brain slices. This effect was not observed in brain slices from Munc13-2 KO mice, which is consistent with the lower abundance and subpopulation-specific distribution of Munc13-2 in presynaptic elements. In Munc13-1/2 DKO brain slices, the deficiency of Munc13-1 dominated the effect of APP processing. The Munc13-1 KO effect on APP processing could be rescued by the stimulation of postsynaptic glutamatergic receptors. This indicates that lack of postsynaptic glutamate receptor stimulation in Munc13-1 KO brain slice cultures but not presynaptic mechanisms account for compromised APP processing. We conclude that organotypic brain slices cultures are a valuable tool for studying APP processing pathways in intact neuronal circuits and that neuronal activity is important for maintenance of the non-amyloidogenic APP processing.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores de Glutamato/metabolismo , Sinapses/metabolismo , Anestésicos Locais/farmacologia , Animais , Encéfalo/citologia , Bloqueadores dos Canais de Cálcio/farmacologia , Relação Dose-Resposta a Droga , Antagonistas de Aminoácidos Excitatórios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Genótipo , Ácido Glutâmico/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Ácido Cinurênico/farmacologia , L-Lactato Desidrogenase/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Proteínas do Tecido Nervoso/deficiência , Neurônios/ultraestrutura , Técnicas de Cultura de Órgãos , Ésteres de Forbol/farmacologia , Cloreto de Potássio/farmacologia , Toxina Tetânica/farmacologia , Tetrodotoxina/farmacologia , Fatores de Tempo
8.
J Neurosci ; 31(36): 12790-801, 2011 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-21900558

RESUMO

Posttranslational amyloid-ß (Aß) modification is considered to play an important role in Alzheimer's disease (AD) etiology. An N-terminally modified Aß species, pyroglutamate-amyloid-ß (pE3-Aß), has been described as a major constituent of Aß deposits specific to human AD but absent in normal aging. Formed via cyclization of truncated Aß species by glutaminyl cyclase (QC; QPCT) and/or its isoenzyme (isoQC; QPCTL), pE3-Aß aggregates rapidly and is known to seed additional Aß aggregation. To directly investigate pE3-Aß toxicity in vivo, we generated and characterized transgenic TBA2.1 and TBA2.2 mice, which express truncated mutant human Aß. Along with a rapidly developing behavioral phenotype, these mice showed progressively accumulating Aß and pE3-Aß deposits in brain regions of neuronal loss, impaired long-term potentiation, microglial activation, and astrocytosis. Illustrating a threshold for pE3-Aß neurotoxicity, this phenotype was not found in heterozygous animals but in homozygous TBA2.1 or double-heterozygous TBA2.1/2.2 animals only. A significant amount of pE3-Aß formation was shown to be QC-dependent, because crossbreeding of TBA2.1 with QC knock-out, but not isoQC knock-out, mice significantly reduced pE3-Aß levels. Hence, lowering the rate of QC-dependent posttranslational pE3-Aß formation can, in turn, lower the amount of neurotoxic Aß species in AD.


Assuntos
Precursor de Proteína beta-Amiloide/biossíntese , Transtornos Heredodegenerativos do Sistema Nervoso/genética , Transtornos Heredodegenerativos do Sistema Nervoso/patologia , Hipocampo/patologia , Ácido Pirrolidonocarboxílico/metabolismo , Envelhecimento/patologia , Envelhecimento/psicologia , Doença de Alzheimer/patologia , Animais , Comportamento Animal , Encéfalo/patologia , Ensaio de Imunoadsorção Enzimática , Gliose/patologia , Transtornos Heredodegenerativos do Sistema Nervoso/psicologia , Humanos , Imuno-Histoquímica , Cinética , Potenciação de Longa Duração/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Microscopia Eletrônica , Plasticidade Neuronal/genética , Plasticidade Neuronal/fisiologia , Fenótipo , Equilíbrio Postural/fisiologia , Processamento de Proteína Pós-Traducional , Reflexo de Sobressalto/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Acta Neuropathol ; 121(6): 705-19, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21301857

RESUMO

In the hippocampal formation of Alzheimer's disease (AD) patients, both focal and diffuse deposits of Aß peptides appear in a subregion- and layer-specific manner. Recently, pyroglutamate (pGlu or pE)-modified Aß peptides were identified as a highly pathogenic and seeding Aß peptide species. Since the pE modification is catalyzed by glutaminyl cyclase (QC) this enzyme emerged as a novel pharmacological target for AD therapy. Here, we reveal the role of QC in the formation of different types of hippocampal pE-Aß aggregates. First, we demonstrate that both, focal and diffuse pE-Aß deposits are present in defined layers of the AD hippocampus. While the focal type of pE-Aß aggregates was found to be associated with the somata of QC-expressing interneurons, the diffuse type was not. To address this discrepancy, the hippocampus of amyloid precursor protein transgenic mice was analysed. Similar to observations made in AD, focal (i.e. core-containing) pE-Aß deposits originating from QC-positive neurons and diffuse pE-Aß deposits not associated with QC were detected in Tg2576 mouse hippocampus. The hippocampal layers harbouring diffuse pE-Aß deposits receive multiple afferents from QC-rich neuronal populations of the entorhinal cortex and locus coeruleus. This might point towards a mechanism in which pE-Aß and/or QC are being released from projection neurons at hippocampal synapses. Indeed, there are a number of reports demonstrating the reduction of diffuse, but not of focal, Aß deposits in hippocampus after deafferentation experiments. Moreover, we demonstrate in neurons by live cell imaging and by enzymatic activity assays that QC is secreted in a constitutive and regulated manner. Thus, it is concluded that hippocampal pE-Aß plaques may develop through at least two different mechanisms: intracellularly at sites of somatic QC activity as well as extracellularly through seeding at terminal fields of QC expressing projection neurons.


Assuntos
Doença de Alzheimer/patologia , Aminoaciltransferases/metabolismo , Peptídeos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Ácido Pirrolidonocarboxílico/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacocinética , Antagonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Antagonistas Adrenérgicos beta/farmacocinética , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Aminoaciltransferases/deficiência , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Autorradiografia/métodos , Células Cultivadas , Di-Hidroalprenolol/farmacocinética , Feminino , Proteínas de Fluorescência Verde/genética , Hipocampo/patologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Cloreto de Potássio/farmacologia , Prazosina/farmacocinética , Ligação Proteica/efeitos dos fármacos , Receptores Adrenérgicos/metabolismo , Fatores de Tempo , Trítio/farmacocinética , Ioimbina/farmacocinética
10.
Acta Neuropathol ; 120(2): 195-207, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20383514

RESUMO

Glutaminyl cyclase (QC) was discovered recently as the enzyme catalyzing the pyroglutamate (pGlu or pE) modification of N-terminally truncated Alzheimer's disease (AD) Abeta peptides in vivo. This modification confers resistance to proteolysis, rapid aggregation and neurotoxicity and can be prevented by QC inhibitors in vitro and in vivo, as shown in transgenic animal models. However, in mouse brain QC is only expressed by a relatively low proportion of neurons in most neocortical and hippocampal subregions. Here, we demonstrate that QC is highly abundant in subcortical brain nuclei severely affected in AD. In particular, QC is expressed by virtually all urocortin-1-positive, but not by cholinergic neurons of the Edinger-Westphal nucleus, by noradrenergic locus coeruleus and by cholinergic nucleus basalis magnocellularis neurons in mouse brain. In human brain, QC is expressed by both, urocortin-1 and cholinergic Edinger-Westphal neurons and by locus coeruleus and nucleus basalis Meynert neurons. In brains from AD patients, these neuronal populations displayed intraneuronal pE-Abeta immunoreactivity and morphological signs of degeneration as well as extracellular pE-Abeta deposits. Adjacent AD brain structures lacking QC expression and brains from control subjects were devoid of such aggregates. This is the first demonstration of QC expression and pE-Abeta formation in subcortical brain regions affected in AD. Our results may explain the high vulnerability of defined subcortical neuronal populations and their central target areas in AD as a consequence of QC expression and pE-Abeta formation.


Assuntos
Doença de Alzheimer/patologia , Aminoaciltransferases/metabolismo , Núcleo Basal de Meynert/enzimologia , Locus Cerúleo/enzimologia , Substância Cinzenta Periaquedutal/enzimologia , Ácido Pirrolidonocarboxílico/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Aminoaciltransferases/deficiência , Peptídeos beta-Amiloides/metabolismo , Animais , Colina O-Acetiltransferase/metabolismo , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Pessoa de Meia-Idade , Tirosina 3-Mono-Oxigenase/metabolismo , Urocortinas/metabolismo
11.
Int J Dev Neurosci ; 27(8): 825-35, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19699792

RESUMO

Glutaminyl cyclase (QC) converts N-terminal glutaminyl residues into pyroglutamate (pE), thereby stabilizing these peptides/proteins. Recently, we demonstrated that QC also plays a pathogenic role in Alzheimer's disease by generating the disease-associated pE-Abeta from N-terminally truncated Abeta peptides in vivo. This newly identified function makes QC an interesting pharmacological target for Alzheimer's disease therapy. However, the expression of QC in brain and peripheral organs, its cell type-specific and subcellular localization as well as developmental profiles in brain are not known. The present study was performed to address these issues in mice. In brain, QC mRNA expression was highest in hypothalamus, followed by hippocampus and cortex. In liver, QC mRNA concentration was almost as high as in brain while lower QC mRNA levels were detected in lung and heart and very low expression levels were found in kidney and spleen. In the developmental course, stable QC mRNA levels were detected in hypothalamus from postnatal day 5 to 370. On the contrary, in cortex and hippocampus QC mRNA levels were highest after birth and declined during ontogenesis by 20-25%. These results were corroborated by immunocytochemical analysis in mouse brain demonstrating a robust QC expression in a subpopulation of lateral and paraventricular hypothalamic neurons and the labeling of a significant number of small neurons in the hippocampal molecular layer, in the hilus of the dentate gyrus and in all layers of the neocortex. Hippocampal QC-immunoreactive neurons include subsets of parvalbumin-, calbindin-, calretinin-, cholecystokinin- and somatostatin-positive GABAergic interneurons. The density of QC labeled hippocampal neurons declined during postnatal development matching the decrease in QC mRNA expression levels. Subcellular double immunofluorescent analysis localized QC within the endoplasmatic reticulum, Golgi apparatus and secretory granules, consistent with a function of QC in protein maturation and/or modification. Our results are in compliance with a role of QC in hypothalamic hormone maturation and suggest additional, yet unidentified QC functions in brain regions relevant for learning and memory which are affected in Alzheimer's disease.


Assuntos
Aminoaciltransferases/metabolismo , Encéfalo/enzimologia , Encéfalo/crescimento & desenvolvimento , Doença de Alzheimer/metabolismo , Aminoaciltransferases/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/anatomia & histologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Neurônios/ultraestrutura , Organelas/metabolismo , Organelas/ultraestrutura , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA