Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 247
Filtrar
1.
J Labelled Comp Radiopharm ; 66(4-6): 145-154, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36931890

RESUMO

The generation of amyloid beta peptides that aggregate in the brain is believed to play a major role in Alzheimer's disease. In theory, the inhibition of beta-site amyloid precursor protein-cleaving enzyme 1 (BACE1), which catalyzes the initial rate-limiting step in amyloid beta production, may slow or stop Alzheimer's disease. Herein, we report the preparation of two potent BACE1 inhibitors, BI 1147560 (1) and BI 1181181 (2), labeled with carbon-14 and with deuterium. The use of advanced key chiral intermediates like 3 and 5 shortened the carbon-14 syntheses of these two compounds to five and six steps, respectively, and helped in preparing them with very high chemical purity and enantiomeric excess without deviating from the process chemistry route. For the deuterium synthesis, oxetan-3-ylmethanamine [2 H6 ]-7 and 2-fluoro-2-methylpropan-1-amine [2 H6 ]-9 were prepared then used with the chiral intermediate 5 to furnish deuterium labeled 1 and 2, respectively.


Assuntos
Doença de Alzheimer , Humanos , Peptídeos beta-Amiloides , Secretases da Proteína Precursora do Amiloide/fisiologia , Precursor de Proteína beta-Amiloide , Ácido Aspártico Endopeptidases/química , Ácido Aspártico Endopeptidases/fisiologia , Radioisótopos de Carbono , Deutério , Inibidores Enzimáticos
2.
J Biol Chem ; 296: 100372, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33548223

RESUMO

Neural cell adhesion molecules 1 (NCAM1) and 2 (NCAM2) belong to the cell adhesion molecules of the immunoglobulin superfamily and have been shown to regulate formation, maturation, and maintenance of synapses. NCAM1 and NCAM2 undergo proteolysis, but the identity of all the proteases involved and how proteolysis is used to regulate their functions are not known. We report here that NCAM1 and NCAM2 are BACE1 substrates in vivo. NCAM1 and NCAM2 overexpressed in HEK cells were both cleaved by metalloproteinases or BACE1, and NCAM2 was also processed by γ-secretase. We identified the BACE1 cleavage site of NCAM1 (at Glu 671) and NCAM2 (at Glu 663) using mass spectrometry and site-directed mutagenesis. Next, we assessed BACE1-mediated processing of NCAM1 and NCAM2 in the mouse brain during aging. NCAM1 and NCAM2 were cleaved in the olfactory bulb of BACE1+/+ but not BACE1-/- mice at postnatal day 10 (P10), 4 and 12 months of age. In the hippocampus, a BACE1-specific soluble fragment of NCAM1 (sNCAM1ß) was only detected at P10. However, we observed an accumulation of full-length NCAM1 in hippocampal synaptosomes in 4-month-old BACE1-/- mice. We also found that polysialylated NCAM1 (PSA-NCAM1) levels were increased in BACE1-/- mice at P10 and demonstrated that BACE1 cleaves both NCAM1 and PSA-NCAM1 in vitro. In contrast, we did not find evidence for BACE1-dependent NCAM2 processing in the hippocampus at any age analyzed. In summary, our data demonstrate that BACE1 differentially processes NCAM1 and NCAM2 depending on the region of brain, subcellular localization, and age in vivo.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Antígeno CD56/metabolismo , Moléculas de Adesão de Célula Nervosa/metabolismo , Secretases da Proteína Precursora do Amiloide/fisiologia , Animais , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/fisiologia , Encéfalo/metabolismo , Antígeno CD56/fisiologia , Moléculas de Adesão Celular/metabolismo , Feminino , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Moléculas de Adesão de Célula Nervosa/fisiologia , Neurônios/metabolismo , Ácidos Siálicos/metabolismo , Análise Espaço-Temporal , Sinapses/metabolismo
3.
Eur J Immunol ; 50(8): 1209-1219, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32198923

RESUMO

Ankylosing spondylitis (AS) is associated with autoantibody production to class II MHC-associated invariant chain peptide, CD74/CLIP. In this study, we considered that anti-CD74/CLIP autoantibodies present in sera from AS might recognize CD74 degradation products that accumulate upon deficiency of the enzyme signal peptide peptidase-like 2A (SPPL2a). We analyzed monocytes from healthy controls (n = 42), psoriatic arthritis (n = 25), rheumatoid arthritis (n = 16), and AS patients (n = 15) for SPPL2a enzyme activity and complemented the experiments using SPPL2a-sufficient and -deficient THP-1 cells. We found defects in SPPL2a function and CD74 processing in a subset of AS patients, which culminated in CD74 and HLA class II display at the cell surface. These findings were verified in SPPL2a-deficient THP-1 cells, which showed expedited expression of MHC class II, total CD74 and CD74 N-terminal degradation products at the plasma membrane upon receipt of an inflammatory trigger. Furthermore, we observed that IgG anti-CD74/CLIP autoantibodies recognize CD74 N-terminal degradation products that accumulate upon SPPL2a defect. In conclusion, reduced activity of SPPL2a protease in monocytes from AS predisposes to endosomal accumulation of CD74 and CD74 N-terminal fragments, which, upon IFN-γ-exposure, is deposited at the plasma membrane and can be recognized by anti-CD74/CLIP autoantibodies.


Assuntos
Antígenos de Diferenciação de Linfócitos B/imunologia , Ácido Aspártico Endopeptidases/fisiologia , Autoanticorpos/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Proteólise , Espondilite Anquilosante/imunologia , Adulto , Idoso , Antígenos de Diferenciação de Linfócitos B/metabolismo , Feminino , Antígenos HLA-DR/análise , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Imunoglobulina G/imunologia , Interferon gama/farmacologia , Masculino , Pessoa de Meia-Idade , Células THP-1
4.
Mol Biol Cell ; 31(1): 27-44, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31746668

RESUMO

Processing of amyloid precursor protein (APP) by the ß-secretase BACE1 is the initial step of the amyloidogenic pathway to generate amyloid-ß (Aß). Although newly synthesized BACE1 and APP are transported along the secretory pathway, it is not known whether BACE1 and APP share the same post-Golgi trafficking pathways or are partitioned into different transport routes. Here we demonstrate that BACE1 exits the Golgi in HeLa cells and primary neurons by a pathway distinct from the trafficking pathway for APP. By using the Retention Using Selective Hooks system, we show that BACE1 is transported from the trans-Golgi network to the plasma membrane in an AP-1- and Arf1/4-dependent manner. Subsequently, BACE1 is endocytosed to early and recycling endosomes. Perturbation of BACE1 post-Golgi trafficking results in an increase in BACE1 cleavage of APP and increased production of both Aß40 and Aß42. These findings reveal that Golgi exit of BACE1 and APP in primary neurons is tightly regulated, resulting in their segregation along different transport routes, which limits APP processing.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Transporte Proteico/fisiologia , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/fisiologia , Peptídeos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidases/fisiologia , Membrana Celular/metabolismo , Movimento Celular , Endocitose , Endossomos/metabolismo , Complexo de Golgi/metabolismo , Células HeLa , Humanos , Neurônios/metabolismo , Rede trans-Golgi/metabolismo , Rede trans-Golgi/fisiologia
5.
PLoS One ; 13(12): e0207621, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30517136

RESUMO

The malaria parasite replicates within erythrocytes. The pathogenesis of clinical malaria is in large part due to the capacity of the parasite to remodel its host cell. To do this, intraerythrocytic stages of Plasmodium falciparum export more than 300 proteins that dramatically alter the morphology of the infected erythrocyte as well as its mechanical and adhesive properties. P. falciparum plasmepsin V (PfPMV) is an aspartic protease that processes proteins for export into the host erythrocyte and is thought to play a key role in parasite virulence and survival. However, although standard techniques for gene disruption as well as conditional protein knockdown have been previously attempted with the pfpmv gene, complete gene removal or knockdown was not achieved so direct genetic proof that PMV is an essential protein has not been established. Here we have used a conditional gene excision approach combining CRISPR-Cas9 gene editing and DiCre-mediated recombination to functionally inactivate the pfpmv gene. The resulting mutant parasites displayed a severe growth defect. Detailed phenotypic analysis showed that development of the mutant parasites was arrested early in the ring-to-trophozoite transition in the erythrocytic cycle following gene excision. Our findings are the first to elucidate the effects of PMV gene disruption, showing that it is essential for parasite viability in asexual blood stages. The mutant parasites can now be used as a platform to further dissect the Plasmodium protein export pathway.


Assuntos
Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/fisiologia , Plasmodium falciparum/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Sistemas CRISPR-Cas , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Regulação da Expressão Gênica , Humanos , Mutação/genética , Plasmodium falciparum/genética , Inibidores de Proteases , Processamento de Proteína Pós-Traducional , Proteínas de Protozoários/metabolismo
6.
eNeuro ; 5(4)2018.
Artigo em Inglês | MEDLINE | ID: mdl-30079376

RESUMO

ß-Site amyloid precursor protein cleaving enzyme 1 (BACE1) is required for the production of ß-amyloid (Aß), one of the major pathogenic molecules of Alzheimer's disease (AD), and is therefore being actively pursued as a drug target for AD. Adult hippocampal neurogenesis (AHN) is a lifelong process that is known to be important for learning and memory and may have the potential to regenerate damaged neural tissue. In this study, we examined whether BACE1 regulates AHN, which holds important implications for its suitability as a drug target in AD. Cohorts of 2-month-old wild-type (BACE1+/+), heterozygous, and homozygous BACE1 knockout mice (BACE1+/- and BACE1-/-, respectively) were injected with 5-bromo-2'-deoxyuridine (BrdU) and sacrificed 1 day later to examine the impact of loss of BACE1 on neural precursor cell (NPC) proliferation in the adult brain. Parallel cohorts of mice were sacrificed 4 weeks after BrdU injection to determine the effects of BACE1 on survival and differentiation of newborn NPCs. We found that NPC proliferation was increased in BACE1-/- mice compared to BACE1+/+ mice, while no difference was observed in NPC survival across genotypes. Differentiation of NPCs to neuronal lineage was impaired in BACE1-/- mice. However, no differences were observed in astrogenesis, the proportion of immature neurons, or the production of oligodendrocytes across genotypes. Importantly, corresponding with a decrease in neuronal differentiation in the absence of a complementary increase in an alternate cell fate, BACE1-/- mice were found to have a pool of undifferentiated NPCs in the hippocampus compared to BACE1+/+ and BACE1+/- mice.


Assuntos
Secretases da Proteína Precursora do Amiloide/fisiologia , Ácido Aspártico Endopeptidases/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Hipocampo/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Fatores Etários , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
7.
Mol Psychiatry ; 23(7): 1542-1554, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29703946

RESUMO

Potassium channel Kv2.1 regulates potassium current in cortical neurons and potassium efflux is necessary for cell apoptosis. As a major component of delayed rectifier current potassium channels, Kv2.1 forms clusters in the membrane of hippocampal neurons. BACE2 is an aspartyl protease to cleave APP to prevent the generation of Aß, a central component of neuritic plaques in Alzheimer's brain. We now identified Kv2.1 as a novel substrate of BACE2. We found that BACE2 cleaved Kv2.1 at Thr376, Ala717, and Ser769 sites and disrupted Kv2.1 clustering on cell membrane, resulting in decreased Ik of Kv2.1 and a hyperpolarizing shift in primary neurons. Furthermore, we discovered that the BACE2-cleaved Kv2.1 forms, Kv2.1-1-375, Kv2.1-1-716, and Kv2.1-1-768, depressed the delayed rectifier Ik surge and reduced neuronal apoptosis. Our study suggests that BACE2 plays a neuroprotective role by cleavage of Kv2.1 to prevent the outward potassium currents, a potential new target for Alzheimer's treatment.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Secretases da Proteína Precursora do Amiloide/fisiologia , Ácido Aspártico Endopeptidases/metabolismo , Ácido Aspártico Endopeptidases/fisiologia , Canais de Potássio Shab/fisiologia , Sequência de Aminoácidos , Animais , Apoptose/fisiologia , Membrana Celular/metabolismo , Células HEK293 , Hipocampo/metabolismo , Humanos , Neurônios/metabolismo , Técnicas de Patch-Clamp , Potássio/metabolismo , Cultura Primária de Células , Ratos , Canais de Potássio Shab/metabolismo , Especificidade por Substrato
8.
Neurobiol Aging ; 60: 129-140, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28946017

RESUMO

The cleavage of amyloid precursor protein (APP) by ß-site APP cleaving enzyme 1 (BACE1) is the rate-limiting step in beta amyloid generation during Alzheimer's disease (AD) pathogenesis. In AD brains, BACE1 is abnormally accumulated in endocytic compartments, where the acidic pH is optimal for its activity. However, mechanisms regulating the endosome-to-trans-Golgi network (TGN) retrieval of BACE1 remain unclear. Here, we show that partitioning defective 3 (Par3) facilitates BACE1 retrograde trafficking from endosomes to the TGN. Par3 functions through aPKC-mediated phosphorylation of BACE1 on Ser498, which in turn promotes the interaction between BACE1 and phosphofurin acidic cluster sorting protein 1 and facilitates the retrograde trafficking of BACE1 to the TGN. In human AD brains, there is a significant decrease in Ser498 phosphorylation of BACE1 suggesting that defective phosphorylation-dependent retrograde transport of BACE1 is important in AD pathogenesis. Together, our studies provide mechanistic insight into a novel role for Par3 and aPKC in regulating the retrograde endosome-to-TGN trafficking of BACE1 and shed light on the mechanisms of AD pathogenesis.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Proteínas de Transporte/fisiologia , Endossomos/metabolismo , Complexo de Golgi/metabolismo , Proteína Quinase C/fisiologia , Proteínas de Transporte Vesicular/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/fisiologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/fisiologia , Encéfalo/metabolismo , Células Cultivadas , Humanos , Proteínas do Tecido Nervoso , Fosforilação , Ratos
9.
Uirusu ; 67(1): 49-58, 2017.
Artigo em Japonês | MEDLINE | ID: mdl-29593153

RESUMO

Hepatitis C virus (HCV) is a blood-borne virus and causes chronic infection leading to development of steatosis, cirrhosis and hepatocellular carcinoma. However, molecular mechanisms of induction of liver diseases by HCV infection are still unclear. This review focuses on thevirological significance of processing of HCV core protein by signal peptide peptidase in propagation and pathogenesis of HCV.


Assuntos
Ácido Aspártico Endopeptidases/fisiologia , Hepacivirus/crescimento & desenvolvimento , Hepacivirus/patogenicidade , Proteínas do Core Viral/metabolismo , Animais , Estresse do Retículo Endoplasmático , Genoma Viral , Hepacivirus/genética , Hepacivirus/fisiologia , Interações Hospedeiro-Patógeno , Camundongos , Replicação Viral/genética
10.
Channels (Austin) ; 10(5): 365-378, 2016 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-27253079

RESUMO

ß-site APP-cleaving enzyme 1 (BACE1) has become infamous for its pivotal role in the pathogenesis of Alzheimer's disease (AD). Consequently, BACE1 represents a prime target in drug development. Despite its detrimental involvement in AD, it should be quite obvious that BACE1 is not primarily present in the brain to drive mental decline. In fact, additional functions have been identified. In this review, we focus on the regulation of ion channels, specifically voltage-gated sodium and KCNQ potassium channels, by BACE1. These studies provide evidence for a highly unexpected feature in the functional repertoire of BACE1. Although capable of cleaving accessory channel subunits, BACE1 exerts many of its physiologically significant effects through direct, non-enzymatic interactions with main channel subunits. We discuss how the underlying mechanisms can be conceived and develop scenarios how the regulation of ion conductances by BACE1 might shape electric activity in the intact and diseased brain and heart.


Assuntos
Secretases da Proteína Precursora do Amiloide/fisiologia , Ácido Aspártico Endopeptidases/fisiologia , Ativação do Canal Iônico , Doença de Alzheimer/fisiopatologia , Animais , Humanos , Neurônios/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Canais de Sódio Disparados por Voltagem/fisiologia
11.
Neurobiol Aging ; 39: 90-8, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26923405

RESUMO

ß-amyloid precursor protein (APP) and amyloid beta peptide (Aß) are strongly implicated in Alzheimer's disease (AD) pathogenesis, although recent evidence has linked APP-ßCTF generated by BACE1 (ß-APP cleaving enzyme 1) to the development of endocytic abnormalities and cholinergic neurodegeneration in early AD. We show that partial BACE1 genetic reduction prevents these AD-related pathological features in the Ts2 mouse model of Down syndrome. Partially reducing BACE1 by deleting one BACE1 allele blocked development of age-related endosome enlargement in the medial septal nucleus, cerebral cortex, and hippocampus and loss of choline acetyltransferase (ChAT)-positive medial septal nucleus neurons. BACE1 reduction normalized APP-ßCTF elevation but did not alter Aß40 and Aß42 peptide levels in brain, supporting a critical role in vivo for APP-ßCTF in the development of these abnormalities. Although ameliorative effects of BACE1 inhibition on ß-amyloidosis and synaptic proteins levels have been previously noted in AD mouse models, our results highlight the additional potential value of BACE1 modulation in therapeutic targeting of endocytic dysfunction and cholinergic neurodegeneration in Down syndrome and AD.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/fisiologia , Peptídeos beta-Amiloides/fisiologia , Precursor de Proteína beta-Amiloide/fisiologia , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/fisiologia , Neurônios Colinérgicos/patologia , Síndrome de Down/genética , Síndrome de Down/patologia , Endossomos/patologia , Deleção de Genes , Estudos de Associação Genética , Degeneração Neural/patologia , Envelhecimento/genética , Envelhecimento/patologia , Alelos , Animais , Colina O-Acetiltransferase/metabolismo , Modelos Animais de Doenças , Endossomos/genética , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Degeneração Neural/genética , Núcleos Septais/citologia , Núcleos Septais/enzimologia
12.
Biochim Biophys Acta ; 1860(6): 1192-201, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26947009

RESUMO

BACKGROUND: Cells have evolved the mechanisms to survive nutritional shortages in the environment. In Saccharomyces cerevisiae, α-mannosidase Ams1 is known to play a role in catabolism of N-linked free oligosaccharides in the cytosol. Although, this enzyme is also known to be transported selectively from the cytosol to the vacuoles by autophagy, the physiological significance of this transport has not been clarified. METHODS: To elucidate the regulatory mechanism of the activity of Ams1, we assessed the enzymatic activity of the cell free extract of the wild-type and various gene disruptants under different nutritional conditions. In addition, the regulation of Ams1 at both transcription and post-translation was examined. RESULTS: The activity of Ams1 was significantly increased upon the depletion of glucose in the medium. Interestingly, the activity of the enzyme was also stimulated by nitrogen starvation. Our data showed that the activity of Ams1 is regulated by the stress responsive transcriptional factors Msn2/4 through the protein kinase A and the target of rapamycin complex 1 pathways. In addition, Ams1 is post-translationally activated by Pep4-dependent processing in the vacuoles. CONCLUSION: Yeast cells monitor extracellular nutrients to regulate mannoside catabolism via the cellular signaling pathway. GENERAL SIGNIFICANCE: This study revealed that intracellular Ams1 activity is exquisitely upregulated in response to nutrient starvation by induced expression as well as by Pep4-dependent enhanced activity in the vacuoles. The signaling molecules responsible for regulation of Ams1 were also clarified.


Assuntos
Saccharomyces cerevisiae/enzimologia , Transdução de Sinais/fisiologia , alfa-Manosidase/metabolismo , Ácido Aspártico Endopeptidases/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Glucose/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos/fisiologia , Nitrogênio/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Regulação para Cima
13.
Mol Neurodegener ; 10: 44, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26336937

RESUMO

BACKGROUND: Alzheimer's disease (AD) is the most common form of dementia, the number of affected individuals is rising, with significant impacts for healthcare systems. Current symptomatic treatments delay, but do not halt, disease progression. Genetic evidence points to aggregation and deposition of amyloid-ß (Aß) in the brain being causal for the neurodegeneration and dementia typical of AD. Approaches to target Aß via inhibition of γ-secretase or passive antibody therapy have not yet resulted in substantial clinical benefits. Inhibition of BACE1 (ß-secretase) has proven a challenging concept, but recent BACE1inhibitors can enter the brain sufficiently well to lower Aß. However, failures with the first clinical BACE1 inhibitors have highlighted the need to generate compounds with appropriate efficacy and safety profiles, since long treatment periods are expected to be necessary in humans. RESULTS: Treatment with NB-360, a potent and brain penetrable BACE-1 inhibitor can completely block the progression of Aß deposition in the brains of APP transgenic mice, a model for amyloid pathology. We furthermore show that almost complete reduction of Aß was achieved also in rats and in dogs, suggesting that these findings are translational across species and can be extrapolated to humans. Amyloid pathology may be an initial step in a complex pathological cascade; therefore we investigated the effect of BACE-1 inhibition on neuroinflammation, a prominent downstream feature of the disease. NB-360 stopped accumulation of activated inflammatory cells in the brains of APP transgenic mice. Upon chronic treatment of APP transgenic mice, patches of grey hairs appeared. CONCLUSIONS: In a rapidly developing field, the data on NB-360 broaden the chemical space and expand knowledge on the properties that are needed to make a BACE-1 inhibitor potent and safe enough for long-term use in patients. Due to its excellent brain penetration, reasonable oral doses of NB-360 were sufficient to completely block amyloid-ß deposition in an APP transgenic mouse model. Data across species suggest similar treatment effects can possibly be achieved in humans. The reduced neuroinflammation upon amyloid reduction by NB-360 treatment supports the notion that targeting amyloid-ß pathology can have beneficial downstream effects on the progression of Alzheimer's disease.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Proteínas do Tecido Nervoso/antagonistas & inibidores , Fármacos Neuroprotetores/uso terapêutico , Ácidos Picolínicos/uso terapêutico , Tiazinas/uso terapêutico , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/fisiologia , Peptídeos beta-Amiloides/genética , Animais , Ácido Aspártico Endopeptidases/fisiologia , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Células CHO , Cricetinae , Cricetulus , Cães , Avaliação Pré-Clínica de Medicamentos , Feminino , Cor de Cabelo/efeitos dos fármacos , Humanos , Camundongos , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Microglia/patologia , Mutação , Proteínas do Tecido Nervoso/fisiologia , Fármacos Neuroprotetores/farmacocinética , Fragmentos de Peptídeos/metabolismo , Ácidos Picolínicos/química , Ácidos Picolínicos/farmacocinética , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Tiazinas/química , Tiazinas/farmacocinética
14.
Mol Neurodegener ; 10: 31, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-26202512

RESUMO

BACKGROUND: Accumulation of the ß-amyloid peptide (Aß) is a major pathological hallmark of Alzheimer's disease (AD). Recent studies have shown that synaptic Aß toxicity may directly impair synaptic function. However, proteins regulating Aß generation at the synapse have not been characterized. Here, we sought to identify synaptic proteins that interact with the extracellular domain of APP and regulate Aß generation. RESULTS: Affinity purification-coupled mass spectrometry identified members of the Synaptotagmin (Syt) family as novel interacting proteins with the APP ectodomain in mouse brains. Syt-1, -2 and -9 interacted with APP in cells and in mouse brains in vivo. Using a GST pull-down approach, we have further demonstrated that the Syt interaction site lies in the 108 amino acids linker region between the E1 and KPI domains of APP. Stable overexpression of Syt-1 or Syt-9 with APP in CHO and rat pheochromocytoma cells (PC12) significantly increased APP-CTF and sAPP levels, with a 2 to 3 fold increase in secreted Aß levels in PC12 cells. Moreover, using a stable knockdown approach to reduce the expression of endogenous Syt-1 in PC12 cells, we have observed a ~ 50% reduction in secreted Aß generation. APP processing also decreased in these cells, shown by lower CTF levels. Lentiviral-mediated knock down of endogenous Syt-1 in mouse primary neurons also led to a significant reduction in both Aß40 and Aß42 generation. As secreted sAPPß levels were significantly reduced in PC12 cells lacking Syt-1 expression, our results suggest that Syt-1 regulates Aß generation by modulating BACE1-mediated cleavage of APP. CONCLUSION: Altogether, our data identify the synaptic vesicle proteins Syt-1 and 9 as novel APP-interacting proteins that promote Aß generation and thus may play an important role in the pathogenesis of AD.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Sinaptotagminas/fisiologia , Secretases da Proteína Precursora do Amiloide/fisiologia , Peptídeos beta-Amiloides/biossíntese , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/química , Animais , Ácido Aspártico Endopeptidases/fisiologia , Células CHO , Cricetinae , Cricetulus , Camundongos , Neurônios/metabolismo , Células PC12 , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/metabolismo , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Especificidade da Espécie , Sinaptotagmina I/deficiência , Sinaptotagmina I/genética , Sinaptotagmina I/fisiologia , Sinaptotagmina II/fisiologia
15.
J Neurosci ; 35(9): 3806-14, 2015 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-25740511

RESUMO

Inhibition of ß-site APP cleaving enzyme 1 (BACE1) is being pursued as a therapeutic target for treating patients with Alzheimer's disease because BACE1 is the sole ß-secretase for generating ß-amyloid peptide. Knowledge regarding the other cellular functions of BACE1 is therefore critical for the safe use of BACE1 inhibitors in human patients. BACE1 deficiency in mice causes hypomyelination during development and impairs remyelination in injured sciatic nerves. Since BACE1 is expected to be ubiquitously expressed, we asked whether axonal or Schwann cell BACE1 is required for optimal remyelination. By swapping sciatic nerve segments from BACE1-null mice with the corresponding wild-type nerve segments or vice versa, we tested how a deficiency of BACE1 in Schwann cells or axons affects remyelination. Our results show that BACE1 in axons and Schwann cells is similarly important for remyelination of regenerated axons. Nerve injury induces BACE1 transcription and protein levels are elevated in Schwann cells. Expression of type I neuregulin 1 (Nrg1), rather than type III Nrg1, was induced by Schwann cells, and the abolished Nrg1 cleavage in BACE1-null Schwann cells contributed to decreased remyelination of regenerated axons. Hence, this study is the first to demonstrate the equal importance of axonal and Schwann cell BACE1 for remyelination of injured nerves.


Assuntos
Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/fisiologia , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/fisiologia , Axônios/fisiologia , Bainha de Mielina/fisiologia , Regeneração Nervosa/fisiologia , Nervos Periféricos/fisiologia , Células de Schwann/fisiologia , Animais , Axônios/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Bainha de Mielina/genética , Regeneração Nervosa/genética , Tecido Nervoso/transplante , Neuregulina-1/biossíntese , Neuregulina-1/genética , Células de Schwann/enzimologia , Nervo Isquiático/fisiologia , Nervo Isquiático/transplante
16.
Neurobiol Aging ; 36(3): 1310-5, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25557959

RESUMO

One major pathologic hallmark and trigger of Alzheimer's disease (AD) is overproduction and accumulation of ß-amyloid (Aß) species in the brain. Aß is derived from ß-amyloid precursor protein (APP) through sequential cleavages by ß- and γ-secretases. Abnormal copper homeostasis also contributes to AD pathogenesis. Recently, we find that a copper-related protein, CutA divalent cation tolerance homolog of Escherichia coli (CUTA), interacts with the ß-secretase ß-site APP cleaving enzyme 1 (BACE1) and inhibits APP ß-processing and Aß generation. Herein, we further found that overexpression of CUTA increases intracellular copper level, whereas copper treatments promote CUTA expression. We also confirmed that copper treatments promote APP expression and Aß secretion. In addition, copper treatments promoted the increase of Aß secretion induced by CUTA downregulation but had no effect on CUTA-ß-site APP cleaving enzyme 1 interaction. On the other hand, CUTA overexpression ameliorated copper-induced Aß secretion but had no effect on APP expression. Moreover, we found that Aß treatments can reduce both CUTA and copper levels in mouse primary neurons. Consistently, both CUTA and copper levels were decreased in the hippocampus of APP/PS1 AD mouse brain. Together, our results reveal a reciprocal modulation of copper and CUTA and suggest that both regulate Aß generation through different mechanisms, although Aß mutually affects copper and CUTA levels.


Assuntos
Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Cobre/fisiologia , Proteínas de Membrana/fisiologia , Secretases da Proteína Precursora do Amiloide/fisiologia , Animais , Ácido Aspártico Endopeptidases/fisiologia , Células Cultivadas , Cobre/metabolismo , Cobre/farmacologia , Expressão Gênica , Homeostase , Humanos , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Coelhos
17.
J Invest Dermatol ; 135(2): 551-559, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25268585

RESUMO

Substance P (SP) is a well-known neuropeptide implicated in the wound-healing process. The wound occasionally causes a pigmented scar. In the present study, we examined whether increased levels of SP affected melanogenesis. When human melanocytes were treated with SP, the melanin content increased and the pigmentation process accelerated in a dose-dependent manner. In addition to melanogenesis-related genes, the expression of neurokinin 1 receptor, endothelin 1 (EDN1), and EDN receptor type B (EDNRB) also increased at both the messenger RNA and protein levels. Interestingly, secreted EDN1 was observed in the melanocyte culture medium, and this phenomenon was significantly enhanced by SP treatment. Through knockdown experiments using small interfering RNAs (siRNAs), we confirmed that endothelin-converting enzyme 1 (ECE1), EDN1, and EDNRB were involved in SP-induced pigmentation and found that EDN1 secretion was affected by ECE1 and EDN1 siRNAs, but not by EDNRB siRNA. These findings indicate that ECE1 is essential for EDN1 secretion in melanocytes and that EDNRB functions downstream of secreted EDN1 to increase the cAMP levels and activate the melanogenesis-related phosphorylation cascade. This study provides in vitro evidence for a melanogenic function of SP in the skin and suggests that the SP-related signal is a potent target for regulating stress- or wound-induced pigmentation.


Assuntos
Ácido Aspártico Endopeptidases/fisiologia , Transformação Celular Neoplásica/metabolismo , Endotelina-1/metabolismo , Melaninas/biossíntese , Melanócitos/metabolismo , Metaloendopeptidases/fisiologia , Substância P/farmacologia , Células Cultivadas , AMP Cíclico/biossíntese , Endotelina-1/genética , Enzimas Conversoras de Endotelina , Humanos , Fosforilação , Receptor de Endotelina B/genética , Receptores da Neurocinina-1/genética , Receptores da Neurocinina-1/fisiologia , Transdução de Sinais , Pigmentação da Pele/efeitos dos fármacos
18.
J Cell Biol ; 205(6): 847-62, 2014 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-24958774

RESUMO

The regulated turnover of endoplasmic reticulum (ER)-resident membrane proteins requires their extraction from the membrane lipid bilayer and subsequent proteasome-mediated degradation. Cleavage within the transmembrane domain provides an attractive mechanism to facilitate protein dislocation but has never been shown for endogenous substrates. To determine whether intramembrane proteolysis, specifically cleavage by the intramembrane-cleaving aspartyl protease signal peptide peptidase (SPP), is involved in this pathway, we generated an SPP-specific somatic cell knockout. In a stable isotope labeling by amino acids in cell culture-based proteomics screen, we identified HO-1 (heme oxygenase-1), the rate-limiting enzyme in the degradation of heme to biliverdin, as a novel SPP substrate. Intramembrane cleavage by catalytically active SPP provided the primary proteolytic step required for the extraction and subsequent proteasome-dependent degradation of HO-1, an ER-resident tail-anchored protein. SPP-mediated proteolysis was not limited to HO-1 but was required for the dislocation and degradation of additional tail-anchored ER-resident proteins. Our study identifies tail-anchored proteins as novel SPP substrates and a specific requirement for SPP-mediated intramembrane cleavage in protein turnover.


Assuntos
Ácido Aspártico Endopeptidases/fisiologia , Proteínas de Membrana/metabolismo , Células HeLa , Heme Oxigenase-1/metabolismo , Humanos , Estrutura Terciária de Proteína , Proteólise , Proteômica , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Ubiquitinação
19.
Neural Plast ; 2014: 128631, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24963413

RESUMO

Alzheimer's disease (AD) is the most common form of age-related dementia, which is thought to result from overproduction and/or reduced clearance of amyloid-beta (Aß) peptides. Studies over the past few decades suggest that Aß is produced in an activity-dependent manner and has physiological relevance to normal brain functions. Similarly, physiological functions for ß- and γ-secretases, the two key enzymes that produce Aß by sequentially processing the amyloid precursor protein (APP), have been discovered over recent years. In particular, activity-dependent production of Aß has been suggested to play a role in homeostatic regulation of excitatory synaptic function. There is accumulating evidence that activity-dependent immediate early gene Arc is an activity "sensor," which acts upstream of Aß production and triggers AMPA receptor endocytosis to homeostatically downregulate the strength of excitatory synaptic transmission. We previously reported that Arc is critical for sensory experience-dependent homeostatic reduction of excitatory synaptic transmission in the superficial layers of visual cortex. Here we demonstrate that mice lacking the major neuronal ß-secretase, BACE1, exhibit a similar phenotype: stronger basal excitatory synaptic transmission and failure to adapt to changes in visual experience. Our results indicate that BACE1 plays an essential role in sensory experience-dependent homeostatic synaptic plasticity in the neocortex.


Assuntos
Secretases da Proteína Precursora do Amiloide/fisiologia , Ácido Aspártico Endopeptidases/fisiologia , Plasticidade Neuronal/fisiologia , Córtex Visual/fisiologia , Secretases da Proteína Precursora do Amiloide/genética , Animais , Ácido Aspártico Endopeptidases/genética , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/fisiologia , Interpretação Estatística de Dados , Potenciais Pós-Sinápticos Excitadores/genética , Potenciais Pós-Sinápticos Excitadores/fisiologia , Homeostase , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Células Piramidais/fisiologia , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia , Córtex Visual/química
20.
J Neurochem ; 130(1): 4-28, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24646365

RESUMO

The ß-site APP cleaving enzymes 1 and 2 (BACE1 and BACE2) were initially identified as transmembrane aspartyl proteases cleaving the amyloid precursor protein (APP). BACE1 is a major drug target for Alzheimer's disease because BACE1-mediated cleavage of APP is the first step in the generation of the pathogenic amyloid-ß peptides. BACE1, which is highly expressed in the nervous system, is also required for myelination by cleaving neuregulin 1. Several recent proteomic and in vivo studies using BACE1- and BACE2-deficient mice demonstrate a much wider range of physiological substrates and functions for both proteases within and outside of the nervous system. For BACE1 this includes axon guidance, neurogenesis, muscle spindle formation, and neuronal network functions, whereas BACE2 was shown to be involved in pigmentation and pancreatic ß-cell function. This review highlights the recent progress in understanding cell biology, substrates, and functions of BACE proteases and discusses the therapeutic options and potential mechanism-based liabilities, in particular for BACE inhibitors in Alzheimer's disease. The protease BACE1 is a major drug target in Alzheimer disease. Together with its homolog BACE2, both proteases have an increasing number of functions within and outside of the nervous system. This review highlights recent progress in understanding cell biology, substrates, and functions of BACE proteases and discusses the therapeutic options and potential mechanism-based liabilities, in particular for BACE inhibitors in Alzheimer disease.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/fisiologia , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ácido Aspártico Endopeptidases/fisiologia , Líquido Intracelular/enzimologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Precursor de Proteína beta-Amiloide/fisiologia , Animais , Previsões , Humanos , Líquido Intracelular/efeitos dos fármacos , Transporte Proteico/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA