Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Anesthesiology ; 140(4): 786-802, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38147625

RESUMO

BACKGROUND: Analgesic tolerance due to long-term use of morphine remains a challenge for pain management. Morphine acts on µ-opioid receptors and downstream of the phosphatidylinositol 3-kinase signaling pathway to activate the mammalian target of rapamycin (mTOR) pathway. Rheb is an important regulator of growth and cell-cycle progression in the central nervous system owing to its critical role in the activation of mTOR. The hypothesis was that signaling via the GTP-binding protein Rheb in the dorsal horn of the spinal cord is involved in morphine-induced tolerance. METHODS: Male and female wild-type C57BL/6J mice or transgenic mice (6 to 8 weeks old) were injected intrathecally with saline or morphine twice daily at 12-h intervals for 5 consecutive days to establish a tolerance model. Analgesia was assessed 60 min later using the tail-flick assay. After 5 days, the spine was harvested for Western blot or immunofluorescence analysis. RESULTS: Chronic morphine administration resulted in the upregulation of spinal Rheb by 4.27 ± 0.195-fold (P = 0.0036, n = 6), in turn activating mTOR by targeting rapamycin complex 1 (mTORC1). Genetic overexpression of Rheb impaired morphine analgesia, resulting in a tail-flick latency of 4.65 ± 1.10 s (P < 0.0001, n = 7) in Rheb knock-in mice compared to 10 s in control mice (10 ± 0 s). Additionally, Rheb overexpression in spinal excitatory neurons led to mTORC1 signaling overactivation. Genetic knockout of Rheb or inhibition of mTORC1 signaling by rapamycin potentiated morphine-induced tolerance (maximum possible effect, 52.60 ± 9.56% in the morphine + rapamycin group vs. 16.60 ± 8.54% in the morphine group; P < 0.0001). Moreover, activation of endogenous adenosine 5'-monophosphate-activated protein kinase inhibited Rheb upregulation and retarded the development of morphine-dependent tolerance (maximum possible effect, 39.51 ± 7.40% in morphine + metformin group vs. 15.58 ± 5.79% in morphine group; P < 0.0001). CONCLUSIONS: This study suggests spinal Rheb as a key molecular factor for regulating mammalian target of rapamycin signaling.


Assuntos
Proteínas Monoméricas de Ligação ao GTP , Feminino , Masculino , Camundongos , Animais , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Morfina/farmacologia , Sirolimo/farmacologia , Camundongos Endogâmicos C57BL , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Dor , Mamíferos/metabolismo
2.
Cell Rep ; 37(7): 110014, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34788607

RESUMO

Mutations of SHANK3 cause Phelan-McDermid syndrome (PMS), and these individuals can exhibit sensitivity to stress, resulting in behavioral deterioration. Here, we examine the interaction of stress with genotype using a mouse model with face validity to PMS. In Shank3ΔC/+ mice, swim stress produces an altered transcriptomic response in pyramidal neurons that impacts genes and pathways involved in synaptic function, signaling, and protein turnover. Homer1a, which is part of the Shank3-mGluR-N-methyl-D-aspartate (NMDA) receptor complex, is super-induced and is implicated in the stress response because stress-induced social deficits in Shank3ΔC/+ mice are mitigated in Shank3ΔC/+;Homer1a-/- mice. Several lines of evidence demonstrate that Shank3 expression is regulated by Homer1a in competition with crosslinking forms of Homer, and consistent with this model, Shank3 expression and function that are reduced in Shank3ΔC/+ mice are rescued in Shank3ΔC/+;Homer1a-/- mice. Studies highlight the interaction between stress and genetics and focus attention on activity-dependent changes that may contribute to pathogenesis.


Assuntos
Proteínas de Arcabouço Homer/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Estresse Psicológico/metabolismo , Animais , Deleção Cromossômica , Transtornos Cromossômicos/metabolismo , Transtornos Cromossômicos/fisiopatologia , Cromossomos Humanos Par 22/metabolismo , Modelos Animais de Doenças , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Proteínas de Arcabouço Homer/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Proteínas do Tecido Nervoso/genética , Fenótipo , Células Piramidais/metabolismo , Estresse Psicológico/fisiopatologia
3.
Biol Psychiatry ; 89(11): 1058-1072, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33353667

RESUMO

BACKGROUND: The serine-threonine kinase mTORC1 (mechanistic target of rapamycin complex 1) is essential for normal cell function but is aberrantly activated in the brain in both genetic-developmental and sporadic diseases and is associated with a spectrum of neuropsychiatric symptoms. The underlying molecular mechanisms of cognitive and neuropsychiatric symptoms remain controversial. METHODS: The present study examines behaviors in transgenic models that express Rheb, the most proximal known activator of mTORC1, and profiles striatal phosphoproteomics in a model with persistently elevated mTORC1 signaling. Biochemistry, immunohistochemistry, electrophysiology, and behavior approaches are used to examine the impact of persistently elevated mTORC1 on D1 dopamine receptor (D1R) signaling. The effect of persistently elevated mTORC1 was confirmed using D1-Cre to elevate mTORC1 activity in D1R neurons. RESULTS: We report that persistently elevated mTORC1 signaling blocks canonical D1R signaling that is dependent on DARPP-32 (dopamine- and cAMP-regulated neuronal phosphoprotein). The immediate downstream effector of mTORC1, ribosomal S6 kinase 1 (S6K1), phosphorylates and activates DARPP-32. Persistent elevation of mTORC1-S6K1 occludes dynamic D1R signaling downstream of DARPP-32 and blocks multiple D1R responses, including dynamic gene expression, D1R-dependent corticostriatal plasticity, and D1R behavioral responses including sociability. Candidate biomarkers of mTORC1-DARPP-32 occlusion are increased in the brain of human disease subjects in association with elevated mTORC1-S6K1, supporting a role for this mechanism in cognitive disease. CONCLUSIONS: The mTORC1-S6K1 intersection with D1R signaling provides a molecular framework to understand the effects of pathological mTORC1 activation on behavioral symptoms in neuropsychiatric disease.


Assuntos
Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Receptores de Dopamina D1/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais , Humanos , Fosforilação , Serina-Treonina Quinases TOR/metabolismo
4.
Cell Death Dis ; 11(9): 747, 2020 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-32920594

RESUMO

The small GTPase Ras homolog enriched in the brain (Rheb) can activate mammalian target of rapamycin (mTOR) and regulate the growth and cell cycle progression. We investigated the role of Rheb-mediated mTORC1 signaling in neuropathic pain. A chronic constriction injury (CCI) model was dopted. CCI induced obvious spinal Rheb expression and phosphorylation of mTOR, S6, and 4-E-BP1. Blocking mTORC1 signal with rapamycin alleviated the neuropathic pain and restored morphine efficacy in CCI model. Immunofluoresence showed a neuronal co-localization of CCI-induced Rheb and pS6. Rheb knockin mouse showed a similar behavioral phenotype as CCI. In spinal slice recording, CCI increased the firing frequency of neurons expressing HCN channels; inhibition of mTORC1 with rapamycin could reverse the increased spinal neuronal activity in neuropathic pain. Spinal Rheb is induced in neuropathic pain, which in turn active the mTORC1 signaling in CCI. Spinal Rheb-mTOR signal plays an important role in regulation of spinal sensitization in neuropathic pain, and targeting mTOR may give a new strategy for pain management.


Assuntos
Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Neuralgia/metabolismo , Neurônios/metabolismo , Proteína Enriquecida em Homólogo de Ras do Encéfalo/metabolismo , Medula Espinal/metabolismo , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuralgia/patologia , Neurônios/patologia , Transdução de Sinais , Medula Espinal/patologia , Serina-Treonina Quinases TOR/metabolismo
5.
Mol Cell ; 75(1): 13-25.e5, 2019 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-31151856

RESUMO

Arc is a synaptic protein essential for memory consolidation. Recent studies indicate that Arc originates in evolution from a Ty3-Gypsy retrotransposon GAG domain. The N-lobe of Arc GAG domain acquired a hydrophobic binding pocket in higher vertebrates that is essential for Arc's canonical function to weaken excitatory synapses. Here, we report that Arc GAG also acquired phosphorylation sites that can acutely regulate its synaptic function. CaMKII phosphorylates the N-lobe of the Arc GAG domain and disrupts an interaction surface essential for high-order oligomerization. In Purkinje neurons, CaMKII phosphorylation acutely reverses Arc's synaptic action. Mutant Arc that cannot be phosphorylated by CaMKII enhances metabotropic receptor-dependent depression in the hippocampus but does not alter baseline synaptic transmission or long-term potentiation. Behavioral studies indicate that hippocampus- and amygdala-dependent learning requires Arc GAG domain phosphorylation. These studies provide an atomic model for dynamic and local control of Arc function underlying synaptic plasticity and memory.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteínas do Citoesqueleto/metabolismo , Potenciação de Longa Duração/fisiologia , Memória/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Células de Purkinje/metabolismo , Sequência de Aminoácidos , Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/metabolismo , Animais , Sítios de Ligação , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/química , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/genética , Técnicas de Introdução de Genes , Células HEK293 , Hipocampo/citologia , Hipocampo/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Moleculares , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Fosforilação , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Células de Purkinje/citologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Sinapses/fisiologia , Transmissão Sináptica
6.
iScience ; 13: 82-97, 2019 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-30826728

RESUMO

N-methyl-D-aspartate (NMDA) receptor (NMDAR) is highly compartmentalized in neurons, and its dysfunction has been implicated in various neuropsychiatric and neurodegenerative disorders. Recent failure to exploit NMDAR antagonization as a potential therapeutic target has driven the need to identify molecular mechanisms that regulate NMDAR compartmentalization. Here, we report that the reduction of Kif5b, the heavy chain of kinesin-1, protected neurons against NMDA-induced excitotoxicity and ischemia-provoked neurodegeneration. Direct binding of kinesin-1 to the GluN2B cytoplasmic tails regulated the levels of NMDAR at extrasynaptic sites and the subsequent influx of calcium mediated by extrasynaptic NMDAR by regulating the insertion of NMDARs into neuronal surface. Transient increase of Kif5b restored the surface levels of NMDAR and the decreased neuronal susceptibility to NMDA-induced excitotoxicity. The expression of Kif5b was repressed in cerebral ischemia preconditioning. Our findings reveal that kinesin-1 regulates extrasynaptic NMDAR targeting and signaling, and the reduction of kinesin-1 could be exploited to defer neurodegeneration.

7.
Cell Discov ; 4: 65, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30603101

RESUMO

Kif5b-driven anterograde transport and clathrin-mediated endocytosis (CME) are responsible for opposite intracellular trafficking, contributing to plasma membrane homeostasis. However, whether and how the two trafficking processes coordinate remain unclear. Here, we show that Kif5b directly interacts with clathrin heavy chain (CHC) at a region close to that for uncoating catalyst (Hsc70) and preferentially localizes on relatively large clathrin-coated vesicles (CCVs). Uncoating in vitro is decreased for CCVs from the cortex of kif5b conditional knockout (mutant) mouse and facilitated by adding Kif5b fragments containing CHC-binding site, while cell peripheral distribution of CHC or Hsc70 keeps unaffected by Kif5b depletion. Furthermore, cellular entry of vesicular stomatitis virus that internalizes into large CCV is inhibited by Kif5b depletion or introducing a dominant-negative Kif5b fragment. These findings showed a new role of Kif5b in regulating large CCV-mediated CME via affecting CCV uncoating, indicating Kif5b as a molecular knot connecting anterograde transport to CME.

8.
Biochem Biophys Res Commun ; 463(1-2): 123-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26002460

RESUMO

Polarization of epithelial cells requires vectorial sorting and transport of polarity proteins to apical or basolateral domains. Kif5b is the mouse homologue of the human ubiquitous Kinesin Heavy Chain (uKHC). To investigate the function of Kif5b in epithelial cells, we examined the phenotypes of Kif5b-deficient MDCK cells. Stable knockdown of Kif5b in MDCK cells resulted in reduced cell proliferation rate, profound changes in cell morphology, loss of epithelial cell marker, and gain of mesenchymal marker, as well as increased cell migration, invasion, and tumorigenesis abilities. E-cadherin and NMMIIA could interact with Kif5b in polarized MDCK cells, and their expression levels were decreased in Kif5b-deficient MDCK cells. Overexpression of E-cadherin and NMMIIA in Kif5b depleted MDCK cells could decrease mesenchymal marker expression and cell migration ability. These results indicate that stable knockdown of Kif5b in MDCK cells can lead to epithelial-mesenchymal transition, which is mediated by defective E-cadherin and NMMIIA expression.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Cinesinas/deficiência , Animais , Caderinas/metabolismo , Ciclo Celular , Linhagem Celular , Polaridade Celular/genética , Polaridade Celular/fisiologia , Proliferação de Células , Cães , Transição Epitelial-Mesenquimal/genética , Técnicas de Silenciamento de Genes , Humanos , Cinesinas/antagonistas & inibidores , Cinesinas/genética , Células Madin Darby de Rim Canino , Camundongos , Camundongos Nus , Cadeias Pesadas de Miosina/metabolismo , Metástase Neoplásica
9.
PLoS One ; 10(4): e0126002, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25885434

RESUMO

Recent studies showed that kidney-specific inactivation of Kif3a produces kidney cysts and renal failure, suggesting that kinesin-mediated intracellular transportation is important for the establishement and maintenance of renal epithelial cell polarity and normal nephron functions. Kif5b, one of the most conserved kinesin heavy chain, is the mouse homologue of the human ubiquitous Kinesin Heavy Chain (uKHC). In order to elucidate the role of Kif5b in kidney development and function, it is essential to establish its expression profile within the organ. Therefore, in this study, we examined the expression pattern of Kif5b in mouse kidney. Kidneys from embryonic (E) 12.5-, 16.5-dpc (days post coitus) mouse fetuses, from postnatal (P) day 0, 10, 20 pups and from adult mice were collected. The distribution of Kif5b was analyzed by immunostaining. The possible involvement of Kif5b in kidney development was investigated in conditional mutant mice by using a Cre-LoxP strategy. This study showed that the distribution of Kif5b displayed spatiotemporal changes during postnatal kidney development. In kidneys of new born mice, Kif5b was strongly expressed in all developing tubules and in the ureteric bud, but not in the glomerulus or in other early-developing structures, such as the cap mesenchyme, the comma-shaped body, and the S-shaped body. In kidneys of postnatal day 20 or of older mice, however, Kif5b was localized selectively in the basolateral domain of epithelial cells of the thick ascending loop of Henle, as well as of the distal convoluted tubule, with little expression being observed in the proximal tubule or in the collecting duct. Conditional knock-down of Kif5b in mouse kidney did not result in detectable morphological defects, but it did lead to a decrease in cell proliferation rate and also to a mislocalization of Na+/K+/-ATPase, indicating that although Kif5b is non-essential for kidney morphogenesis, it is important for nephron maturation.


Assuntos
Rim/crescimento & desenvolvimento , Cinesinas/metabolismo , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Rim/citologia , Rim/fisiologia , Cinesinas/genética , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Dados de Sequência Molecular , ATPase Trocadora de Sódio-Potássio/metabolismo
10.
Biochem Biophys Res Commun ; 432(2): 242-7, 2013 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-23402760

RESUMO

The microtubule motor kinesin-1 is responsible for the nuclear positioning during myogenesis. Here we show that the coiled-coil stalk/tail domain containing the kinesin light chain (KLC) binding sites targets to the perinuclear region like endogenous Kif5b, while the globular tail domain cannot. To investigate which fragments of kinesin heavy chain (Kif5b) is responsible for the myonuclear positioning, we transfect Kif5b expression constructs into Kif5b deficient myoblasts and test their ability to rescue the myonuclear phenotype. We find that the KLC binding domain and the autoinhibitory peptide in the globular tail region are both indispensable for the nuclear membrane localization of Kif5b and the kinesin-1-mediated myonuclear positioning. These results suggest that while the KLC binding domain may directly targets Kif5b to the myonuclear membrane, the autoinhibitory peptide may play an indirect role in regulating the kinesin-1-mediated myonuclear positioning.


Assuntos
Núcleo Celular/metabolismo , Cinesinas/metabolismo , Desenvolvimento Muscular , Mioblastos Esqueléticos/metabolismo , Animais , Células Cultivadas , Cinesinas/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Membrana Nuclear/metabolismo , Sinais de Exportação Nuclear , Proteínas Nucleares/metabolismo , Estrutura Terciária de Proteína
11.
Development ; 140(3): 617-26, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23293293

RESUMO

Controlled delivery of myofibril components to the appropriate sites of assembly is crucial for myofibrillogenesis. Here, we show that kinesin-1 heavy chain Kif5b plays important roles in anterograde transport of α-sarcomeric actin, non-muscle myosin IIB, together with intermediate filament proteins desmin and nestin to the growing tips of the elongating myotubes. Mice with Kif5b conditionally knocked out in myogenic cells showed aggregation of actin filaments and intermediate filament proteins in the differentiating skeletal muscle cells, which further affected myofibril assembly and their linkage to the myotendinous junctions. The expression of Kif5b in mutant myotubes rescued the localization of the affected proteins. Functional mapping of Kif5b revealed a 64-amino acid α-helix domain in the tail region, which directly interacted with desmin and might be responsible for the transportation of these proteins in a complex.


Assuntos
Junções Intercelulares/metabolismo , Cinesinas/metabolismo , Desenvolvimento Muscular , Miofibrilas/metabolismo , Tendões/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Diferenciação Celular , Desmina/genética , Desmina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Complexo de Golgi/metabolismo , Complexo de Golgi/patologia , Proteínas de Fluorescência Verde/metabolismo , Membro Posterior/metabolismo , Membro Posterior/patologia , Proteínas de Filamentos Intermediários/genética , Proteínas de Filamentos Intermediários/metabolismo , Cinesinas/genética , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patologia , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/patologia , Miofibrilas/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Nestina , Miosina não Muscular Tipo IIB/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Transporte Proteico
12.
Diabetes ; 60(1): 320-30, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20870970

RESUMO

OBJECTIVE: Suppression of Kinesin-1 by antisense oligonucleotides, or overexpression of dominant-negative acting kinesin heavy chain, has been reported to affect the sustained phase of glucose-stimulated insulin secretion in ß-cells in vitro. In this study, we examined the in vivo physiological role of Kinesin-1 in ß-cell development and function. RESEARCH DESIGN AND METHODS: A Cre-LoxP strategy was used to generate conditional knockout mice in which the Kif5b gene is specifically inactivated in pancreatic ß-cells. Physiological and histological analyses were carried out in Kif5b knockout mice as well as littermate controls. RESULTS: Mice with ß-cell specific deletion of Kif5b (Kif5b(fl/)⁻:RIP2-Cre) displayed significantly retarded growth as well as slight hyperglycemia in both nonfasting and 16-h fasting conditions compared with control littermates. In addition, Kif5b(fl/)⁻:RIP2-Cre mice displayed significant glucose intolerance, which was not due to insulin resistance but was related to an insulin secretory defect in response to glucose challenge. These defects of ß-cell function in mutant mice were not coupled with observable changes in islet morphology, islet cell composition, or ß-cell size. However, compared with controls, pancreas of Kif5b(fl/)⁻:RIP2-Cre mice exhibited both reduced islet size and increased islet number, concomitant with an increased insulin vesicle density in ß-cells. CONCLUSIONS: In addition to being essential for maintaining glucose homeostasis and regulating ß-cell function, Kif5b may be involved in ß-cell development by regulating ß-cell proliferation and insulin vesicle synthesis.


Assuntos
Células Secretoras de Insulina/fisiologia , Insulina/metabolismo , Cinesinas/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Sequência de Bases , Western Blotting , Primers do DNA , Ensaio de Imunoadsorção Enzimática , Genótipo , Humanos , Insulina/deficiência , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Cinesinas/química , Cinesinas/deficiência , Cinesinas/efeitos dos fármacos , Cinesinas/genética , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Oligonucleotídeos Antissenso/farmacologia , Fragmentos de Peptídeos/química
13.
Neurosci Lett ; 440(3): 344-7, 2008 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-18583042

RESUMO

Amyloid beta peptide (Abeta), generated by proteolytic cleavage of the amyloid precursor protein (APP), play a pivotal role in the pathogenesis of Alzheimer's disease (AD). The key step in the generation of Abeta is cleavage of APP by beta-site APP-cleaving enzyme 1 (BACE1). There is increasing evidence supporting an interaction between APP, Abeta and metal ions. Both APP and Abeta affect ion homeostasis. Conversely, metal ions may interact with several AD-associated pathways involved in neurofibrillary tangle formation, secretase cleavage of APP, proteolytic degradation of Abeta and the generation of reactive oxygen species. However, the underlying mechanisms remain elusive. Here we first reported the differential effects of AD-related metal ions at subtoxic concentrations on the transcription levels of APP and BACE1 in PC12 cells. Copper (Cu(2+), 50-100 microM) and manganese (Mn(2+), 50-100 microM) potently increased the expression of both APP and BACE1 in a time- and concentration-dependent pattern, while zinc (Zn(2+)), iron (Fe(2+)) and aluminum (Al(3+)) did not. To uncover the mechanism(s) of the increasing expression by these ions, we observed the effects of several antioxidants and some specific inhibitors on the up-expression of APP and BACE1 by metal ions. Curcumin almost completely blocked the effects of these irons, while minocycline and sodium ferulate slightly suppressed the increased BACE1 mRNA level. Signaling pathway specific inhibitors PD98059, SB203580 and CEP11004 modestly blocked the up-transcription of APP induced by copper. These results suggest that these irons cause differential effects on the expression of APP and BACE1 in PC12 cells, and curcumin can significantly reverse their effects.


Assuntos
Secretases da Proteína Precursora do Amiloide/genética , Precursor de Proteína beta-Amiloide/genética , Curcumina/farmacologia , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Metais/farmacologia , RNA Mensageiro/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Análise de Variância , Animais , Relação Dose-Resposta a Droga , Células PC12/efeitos dos fármacos , Ratos , Sais de Tetrazólio , Tiazóis , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...