Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 199
Filtrar
1.
Toxicology ; 465: 153052, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34838597

RESUMO

Bisphenol-A (BPA), an environmental endocrine disruptor, is toxic to the central nervous system. Although recent studies have shown BPA-induced neurotoxicity, it is far from clear what precisely epigenetic mechanisms are involved in BPA-induced cognitive deficits. In this study, pheochromocytoma (PC12) cells were treated with BPA at 1 µM for 36 h in vitro. In vivo, C57BL/6 mice were administered to BPA at a dose of 1 mg/kg/day for 10 weeks. The results showed that 1 µM BPA exposure for 36 h impaired neurite outgrowth of PC12 cells through decreasing the primary and secondary branches. Besides, BPA exposure decreased the level of Ac-H3K9 (histone H3 Lys9 acetylation) by upregulating the expression of HDAC2 (histone deacetylases 2) in PC12 cells. Furthermore, treatment of both TSA (Trichostatin A, inhibitor of the histone deacetylase) and shHDAC2 plasmid (HDAC2 knockdown construct) resulted in amelioration neurite outgrowth deficits induced by BPA. In addition, it was shown that repression of HDAC2 could markedly rescue the spine density impairment in the hippocampus and prevent the cognitive impairment caused by BPA exposure in mice. Collectively, HDAC2 plays an essential role in BPA-induced neurotoxicity, which provides a potential molecular target for medical intervention.


Assuntos
Compostos Benzidrílicos/toxicidade , Espinhas Dendríticas/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Hipocampo/efeitos dos fármacos , Histona Desacetilase 2/metabolismo , Neuritos/efeitos dos fármacos , Síndromes Neurotóxicas/etiologia , Fenóis/toxicidade , Animais , Comportamento Animal/efeitos dos fármacos , Cognição/efeitos dos fármacos , Espinhas Dendríticas/enzimologia , Espinhas Dendríticas/patologia , Feminino , Hipocampo/enzimologia , Hipocampo/patologia , Hipocampo/fisiopatologia , Histona Desacetilase 2/genética , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Neuritos/enzimologia , Neuritos/patologia , Crescimento Neuronal/efeitos dos fármacos , Síndromes Neurotóxicas/enzimologia , Síndromes Neurotóxicas/patologia , Síndromes Neurotóxicas/fisiopatologia , Células PC12 , Ratos , Regulação para Cima
2.
Neurochem Int ; 139: 104795, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32652266

RESUMO

Collapsin response mediator proteins (CRMPs) are ubiquitously expressed in neurons from worms to humans. A cardinal feature of CRMPs is to mediate growth cone collapse in response to Semaphorin-3A signaling through interactions with cytoskeletal proteins. These are critical regulatory roles that CRMPs play during neuritogenesis and neural network formation. Through post-translational modifications, such as phosphorylation, O-GlcNAcylation, SUMOylation, and proteolytic cleavage, CRMPs participate in synaptic plasticity by modulating NMDA receptors, L- and N-type voltage-gated calcium channels (VGCCs), thus affecting neurotransmitter release. CRMPs also possess histone deacetylase (HDAC) activity, which deacetylates histone H4 during neuronal death. Calcium-dependent proteolytic cleavage of CRMPs results in the truncation of CRMPs, producing a large 54 kD fragment (p54). Translocation of the p54 fragment into the nucleus leads to deacetylation of nuclear histone H4 and de-repression of transcription factor E2F1 expression. Increased expression of E2F1 elevates the expression of genes in cell cycle and death. These new and exciting studies lead to the realization that CRMPs are multifunctional proteins with both regulatory and enzymatic functions. Increasing numbers of studies associate these functions of CRMPs with the development of mental and neurological disorders, such as schizophrenia, Alzheimer's diseases, brain trauma, and stroke. This review focuses on new evidence showing the regulatory and enzymatic functions of CRMPs and highlights recent understandings of CRMPs' roles in neurological diseases.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuritos/enzimologia , Plasticidade Neuronal/fisiologia , Transcrição Gênica/fisiologia , Animais , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas do Tecido Nervoso/genética , Neuritos/patologia
3.
FASEB J ; 32(5): 2891-2898, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29401590

RESUMO

Cells have developed lineage-specific mechanisms to control proliferation and drive morphologic changes upon differentiation. A hallmark of differentiation is the assembly of signaling molecules that transduce extracellular signals, such as the production of the G protein-regulated enzyme phospholipase Cß (PLCß), which generates calcium signals from sensory stimuli. We found that in most cancerous cell lines there is positive correlation between PLCß1 levels and cell proliferation. In cells of neuronal lineage, however, reducing PLCß1 levels increases the rate of proliferation. Using a combination of biochemical and biophysical methods, we find that, in the G1 phase, a cytosolic population of PLCß1 associates with cyclin-dependent kinase 16 (CDK16), a neuron-specific enzyme that is activated by cyclin Y to inactivate the antioncogenic protein p27Kip1. Binding of PLCß1 directly inhibits CDK16 activity and in turn reduces the ability of cells to enter the S phase. Activation of Gαq by carbachol causes movement of PLCß from the cytosol to the plasma membrane, reducing its association with CDK16. Similarly, the overexpression of activated Gαq moves PLCß1 to the membrane, reverses G1 arrest, and promotes proliferation, thereby connecting external stimuli with cell proliferation. Our results present a model in which the transient high expression of PLCß1 that occurs at the onset of differentiation arrests cells in the G1 phase through its association with CDK16 and allows CDK16 to transition to its postmitotic function of neurite outgrowth and trafficking of synaptic vesicles. The novel role of PLCß1 in neuronal cell proliferation offers a unique interaction that can be manipulated to guide cells into a neuronal phenotype or to develop therapies for neuroblastomas.-Garwain, O., Valla, K., Scarlata, S. Phospholipase Cß1 regulates proliferation of neuronal cells.


Assuntos
Fase G1 , Regulação Enzimológica da Expressão Gênica , Neuritos/enzimologia , Fosfolipase C beta/biossíntese , Fase S , Animais , Membrana Celular/enzimologia , Membrana Celular/genética , Membrana Celular/patologia , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Citosol/enzimologia , Citosol/patologia , Neuritos/patologia , Neuroblastoma/enzimologia , Neuroblastoma/genética , Neuroblastoma/patologia , Neuroblastoma/terapia , Células PC12 , Fosfolipase C beta/genética , Ratos
4.
Mol Cell Biochem ; 444(1-2): 1-13, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29159770

RESUMO

The JNK-interacting protein 3 (JIP3) is a molecular scaffold, expressed predominantly in neurons, that serves to coordinate the activation of the c-Jun N-terminal kinase (JNK) by binding to JNK and the upstream kinases involved in its activation. The JNK pathway is involved in the regulation of many cellular processes including the control of cell survival, cell death and differentiation. JIP3 also associates with microtubule motor proteins such as kinesin and dynein and is likely an adapter protein involved in the tethering of vesicular cargoes to the motors involved in axonal transport in neurons. We have used immunofluorescence microscopy and biochemical fractionation to investigate the subcellular distribution of JIP3 in relation to JNK and to vesicular and organelle markers in rat pheochromocytoma cells (PC12) differentiating in response to nerve growth factor. In differentiated PC12 cells, JIP3 was seen to accumulate in growth cones at the tips of developing neurites where it co-localised with both JNK and the JNK substrate paxillin. Cellular fractionation of PC12 cells showed that JIP3 was associated with a subpopulation of vesicles in the microsomal fraction, distinct from synaptic vesicles, likely to be an anterograde-directed exocytic vesicle pool. In differentiated PC12 cells, JIP3 did not appear to associate with retrograde endosomal vesicles thought to be involved in signalling axonal injury. Together, these observations indicate that JIP3 may be involved in transporting vesicular cargoes to the growth cones of PC12 cells, possibly targeting JNK to its substrate paxillin, and thus facilitating neurite outgrowth.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Diferenciação Celular , Cones de Crescimento/enzimologia , Proteínas do Tecido Nervoso/metabolismo , Neuritos/enzimologia , Vesículas Secretórias/enzimologia , Vesículas Sinápticas/enzimologia , Animais , Células PC12 , Ratos
5.
J Biol Chem ; 292(24): 9906-9918, 2017 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-28432124

RESUMO

The R7 regulator of G protein signaling family (R7-RGS) critically regulates nervous system development and function. Mice lacking all R7-RGS subtypes exhibit diverse neurological phenotypes, and humans bearing mutations in the retinal R7-RGS isoform RGS9-1 have vision deficits. Although each R7-RGS subtype forms heterotrimeric complexes with Gß5 and R7-RGS-binding protein (R7BP) that regulate G protein-coupled receptor signaling by accelerating deactivation of Gi/o α-subunits, several neurological phenotypes of R7-RGS knock-out mice are not readily explained by dysregulated Gi/o signaling. Accordingly, we used tandem affinity purification and LC-MS/MS to search for novel proteins that interact with R7-RGS heterotrimers in the mouse brain. Among several proteins detected, we focused on Gα13 because it had not been linked to R7-RGS complexes before. Split-luciferase complementation assays indicated that Gα13 in its active or inactive state interacts with R7-RGS heterotrimers containing any R7-RGS isoform. LARG (leukemia-associated Rho guanine nucleotide exchange factor (GEF)), PDZ-RhoGEF, and p115RhoGEF augmented interaction between activated Gα13 and R7-RGS heterotrimers, indicating that these effector RhoGEFs can engage Gα13·R7-RGS complexes. Because Gα13/R7-RGS interaction required R7BP, we analyzed phenotypes of neuronal cell lines expressing RGS7 and Gß5 with or without R7BP. We found that neurite retraction evoked by Gα12/13-dependent lysophosphatidic acid receptors was augmented in R7BP-expressing cells. R7BP expression blunted neurite formation evoked by serum starvation by signaling mechanisms involving Gα12/13 but not Gαi/o These findings provide the first evidence that R7-RGS heterotrimers interact with Gα13 to augment signaling pathways that regulate neurite morphogenesis. This mechanism expands the diversity of functions whereby R7-RGS complexes regulate critical aspects of nervous system development and function.


Assuntos
Encéfalo/metabolismo , Proteínas de Transporte/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuritos/metabolismo , Neurônios/metabolismo , Proteínas RGS/metabolismo , Substituição de Aminoácidos , Animais , Encéfalo/citologia , Encéfalo/enzimologia , Proteínas de Transporte/química , Proteínas de Transporte/genética , Linhagem Celular , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/química , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Neuritos/enzimologia , Neurônios/citologia , Neurônios/enzimologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Proteínas RGS/química , Proteínas RGS/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transdução de Sinais
6.
J Cell Biochem ; 118(11): 3960-3967, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28398601

RESUMO

In our work, we used an in vitro culture model to investigate whether antidepressant imipramine (Ip) may protect bupivacaine (Bv)-induced neurotoxicity in mouse dorsal root ganglion (DRG). Adult mouse DRG was treated with 5 mM Bv in vitro to induce neurotoxicity. DRG was then pre-treated with Ip, prior to Bv, to examine its effects on protecting Bv-induced DRG apoptosis and neurite degeneration. Ip-induced dynamic changes in Trk receptors, including TrkA/B/C and phosphor (p-)TrkA/B/C, were examined by qPCR and Western blot. TrkA and TrkB were inhibited by siRNAs to further investigate their functional role in Ip- and Bv-treated DRG. Ip protected Bv-induced apoptosis and neurite loss in DRG. Ip did not alter TrkA/B/C expressions, whereas significantly augmented protein productions of p-TrkA and p-TrkB, but not p-TrkC. SiRNA-mediated TrkA or TrkB downregulation inhibited Trk receptors, and reduced p-TrkA and p-TrkB in DRG. TrkA or TrkB downregulation alone had no effect on Ip-induced protection in Bv-injured DRG. However, co-inhibition of TrkA and TrkB significantly ameliorated the protective effect of Ip on Bv-induced apoptosis and neurite loss in DRG. Imipramine protected bupivacaine-induced neurotoxicity in DRG, likely via the co-activation of TrkA and TrkB signaling pathways. J. Cell. Biochem. 118: 3960-3967, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Antidepressivos/farmacologia , Bupivacaína/efeitos adversos , Gânglios Espinais/metabolismo , Imipramina/farmacologia , Neuritos/enzimologia , Síndromes Neurotóxicas/prevenção & controle , Receptor trkA/metabolismo , Receptor trkB/metabolismo , Animais , Bupivacaína/farmacologia , Ativação Enzimática/efeitos dos fármacos , Gânglios Espinais/patologia , Camundongos , Neuritos/patologia , Síndromes Neurotóxicas/enzimologia , Síndromes Neurotóxicas/patologia , Transdução de Sinais/efeitos dos fármacos
7.
Cell Mol Neurobiol ; 37(3): 417-426, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27098315

RESUMO

Minocycline, a semi-synthetic second-generation derivative of tetracycline, has been reported to exert neuroprotective effects both in animal models and in clinic trials of neurological diseases. In the present study, we first investigated the protective effects of minocycline on oxygen-glucose deprivation and reoxygenation-induced impairment of neurite outgrowth and its potential mechanism in the neuronal cell line, PC12 cells. We found that minocycline significantly increased cell viability, promoted neurite outgrowth and enhanced the expression of growth-associated protein-43 (GAP-43) in PC12 cells exposed to oxygen-glucose deprivation/reoxygenation injury. In addition, immunoblots revealed that minocycline reversed the overexpression of phosphorylated myosin light chain (MLC) and the suppression of activated extracellular signal-regulated kinase 1/2 (ERK1/2) caused by oxygen-glucose deprivation/reoxygenation injury. Moreover, the minocycline-induced neurite outgrowth was significantly blocked by Calyculin A (1 nM), an inhibitor of myosin light chain phosphatase (MLCP), but not by an ERK1/2 inhibitor (U0126; 10 µM). These findings suggested that minocycline activated the MLCP/MLC signaling pathway in PC12 cells after oxygen-glucose deprivation/reoxygenation injury, which resulted in the promotion of neurite outgrowth.


Assuntos
Glucose/deficiência , Minociclina/farmacologia , Cadeias Leves de Miosina/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Neuritos/patologia , Oxigênio/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína GAP-43/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/enzimologia , Células PC12 , Fosforilação/efeitos dos fármacos , Ratos
8.
J Biol Chem ; 291(46): 23895-23905, 2016 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-27655914

RESUMO

Protein tyrosine phosphatase MEG2 (PTP-MEG2) is a unique nonreceptor tyrosine phosphatase associated with transport vesicles, where it facilitates membrane trafficking by dephosphorylation of the N-ethylmaleimide-sensitive fusion factor. In this study, we identify the neurotrophin receptor TrkA as a novel cargo whose transport to the cell surface requires PTP-MEG2 activity. In addition, TrkA is also a novel substrate of PTP-MEG2, which dephosphorylates both Tyr-490 and Tyr-674/Tyr-675 of TrkA. As a result, overexpression of PTP-MEG2 down-regulates NGF/TrkA signaling and blocks neurite outgrowth and differentiation in PC12 cells and cortical neurons.


Assuntos
Neuritos/enzimologia , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Receptor trkA/metabolismo , Transdução de Sinais/fisiologia , Animais , Camundongos , Células PC12 , Transporte Proteico/fisiologia , Proteínas Tirosina Fosfatases não Receptoras/genética , Ratos
9.
Brain Res ; 1648(Pt A): 193-201, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27423518

RESUMO

Diacylglycerol kinase (DGK) is a lipid-metabolizing enzyme that phosphorylates diacylglycerol to produce phosphatidic acid. Previously, we reported that the δ isozyme of DGK was abundantly expressed in the mouse brain. However, the functions of DGKδ in the brain are still unclear. Because conventional DGKδ-knockout (KO) mice die within 24h after birth, we have generated brain-specific conditional DGKδ-KO mice to circumvent the lethality. In the novel object recognition test, the number of contacts in the DGKδ-KO mice to novel and familiar objects was greatly increased compared to the control mice, indicating that the DGKδ-KO mice showed irrational contacts with objects such as compulsive checking. In the marble burying test, which is used for analyzing obsessive-compulsive disorder (OCD)-like phenotypes, the DGKδ-KO mice buried more marbles than the control mice. Additionally, these phenotypes were significantly alleviated by the administration of an OCD remedy, fluoxetine. These results indicate that the DGKδ-KO mice showed OCD-like behaviors. Moreover, the number of long axon/neurites increased in both DGKδ-KO primary cortical neurons and DGKδ-knockdown neuroblastoma Neuro-2a cells compared to control cells. Conversely, overexpression of DGKδ decreased the number of long axon/neurites of Neuro-2a cells. Taken together, these results strongly suggest that a deficiency of DGKδ induces OCD-like behavior through enhancing axon/neurite outgrowth.


Assuntos
Comportamento Animal/fisiologia , Encéfalo/enzimologia , Diacilglicerol Quinase/fisiologia , Transtorno Obsessivo-Compulsivo/enzimologia , Animais , Comportamento Animal/efeitos dos fármacos , Linhagem Celular Tumoral , Diacilglicerol Quinase/genética , Feminino , Fluoxetina/administração & dosagem , Isoenzimas/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuritos/enzimologia , Fenótipo , Reconhecimento Psicológico/fisiologia , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem
10.
J Cell Biol ; 212(1): 91-111, 2016 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-26728857

RESUMO

Rho guanosine triphosphatases (GTPases) control the cytoskeletal dynamics that power neurite outgrowth. This process consists of dynamic neurite initiation, elongation, retraction, and branching cycles that are likely to be regulated by specific spatiotemporal signaling networks, which cannot be resolved with static, steady-state assays. We present NeuriteTracker, a computer-vision approach to automatically segment and track neuronal morphodynamics in time-lapse datasets. Feature extraction then quantifies dynamic neurite outgrowth phenotypes. We identify a set of stereotypic neurite outgrowth morphodynamic behaviors in a cultured neuronal cell system. Systematic RNA interference perturbation of a Rho GTPase interactome consisting of 219 proteins reveals a limited set of morphodynamic phenotypes. As proof of concept, we show that loss of function of two distinct RhoA-specific GTPase-activating proteins (GAPs) leads to opposite neurite outgrowth phenotypes. Imaging of RhoA activation dynamics indicates that both GAPs regulate different spatiotemporal Rho GTPase pools, with distinct functions. Our results provide a starting point to dissect spatiotemporal Rho GTPase signaling networks that regulate neurite outgrowth.


Assuntos
Neuritos/enzimologia , Transdução de Sinais , Análise Espaço-Temporal , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Camundongos , Neuritos/metabolismo , Fenótipo , Células Tumorais Cultivadas
11.
Food Funct ; 5(12): 3160-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25288148

RESUMO

Hericium erinaceus (Bull.: Fr.) Pers. is an edible and medicinal mushroom used traditionally to improve memory. In this study, we investigated the neuritogenic effects of hericenones isolated from H. erinaceus and the mechanisms of action involved. H. erinaceus was cultivated and the secondary metabolites were elucidated by high performance liquid chromatography (HPLC), liquid chromatography-mass spectrometry (LC-MS), and nuclear magnetic resonance (NMR). The secondary metabolites were tested for neurite outgrowth activity (if any). Rat pheochromocytoma (PC12) cells were employed and the nerve growth factor (NGF) level was also determined. The signaling pathways involved in the mushroom-induced neuritogenesis were investigated using several pharmacological inhibitors. Hericenones B-E (1-4), erinacerin A (5) and isohericerin (6) were isolated from the basidiocarps of H. erinaceus. The hericenones did not promote neurite outgrowth but when induced with a low concentration of NGF (5 ng mL(-1)), the neuritogenic activity was comparable to that of the positive control (50 ng mL(-1) of NGF). Hericenone E was able to stimulate NGF secretion which was two-fold higher than that of the positive control. The neuritogenesis process was partially blocked by the tyrosine kinase receptor (Trk) inhibitor, K252a, suggesting that the neuritogenic effect was not solely due to NGF. Hericenone E also increased the phosphorylation of extracellular-signal regulated kinases (ERKs) and protein kinase B (Akt). Taken together, this study suggests that hericenone E potentiated NGF-induced neuritogenesis in PC12 cells via the MEK/ERK and PI3K/Akt pathways.


Assuntos
Basidiomycota/química , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fator de Crescimento Neural/metabolismo , Neuritos/efeitos dos fármacos , Fenóis/farmacologia , Extratos Vegetais/farmacologia , Verduras/química , Animais , Basidiomycota/crescimento & desenvolvimento , Neuritos/enzimologia , Neuritos/metabolismo , Células PC12 , Fenóis/isolamento & purificação , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Extratos Vegetais/isolamento & purificação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Verduras/crescimento & desenvolvimento
12.
Cell Death Dis ; 5: e1225, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24832597

RESUMO

The Rho/ROCK/LIMK pathway is central for the mediation of repulsive environmental signals in the central nervous system. Several studies using pharmacological Rho-associated protein kinase (ROCK) inhibitors have shown positive effects on neurite regeneration and suggest additional pro-survival effects in neurons. However, as none of these drugs is completely target specific, it remains unclear how these effects are mediated and whether ROCK is really the most relevant target of the pathway. To answer these questions, we generated adeno-associated viral vectors to specifically downregulate ROCK2 and LIM domain kinase (LIMK)-1 in rat retinal ganglion cells (RGCs) in vitro and in vivo. We show here that specific knockdown of ROCK2 and LIMK1 equally enhanced neurite outgrowth of RGCs on inhibitory substrates and both induced substantial neuronal regeneration over distances of more than 5 mm after rat optic nerve crush (ONC) in vivo. However, only knockdown of ROCK2 but not LIMK1 increased survival of RGCs after optic nerve axotomy. Moreover, knockdown of ROCK2 attenuated axonal degeneration of the proximal axon after ONC assessed by in vivo live imaging. Mechanistically, we demonstrate here that knockdown of ROCK2 resulted in decreased intraneuronal activity of calpain and caspase 3, whereas levels of pAkt and collapsin response mediator protein 2 and autophagic flux were increased. Taken together, our data characterize ROCK2 as a specific therapeutic target in neurodegenerative diseases and demonstrate new downstream effects of ROCK2 including axonal degeneration, apoptosis and autophagy.


Assuntos
Degeneração Neural , Regeneração Nervosa , Traumatismos do Nervo Óptico/enzimologia , Nervo Óptico/enzimologia , Células Ganglionares da Retina/enzimologia , Quinases Associadas a rho/metabolismo , Animais , Apoptose , Autofagia , Axônios/enzimologia , Axônios/patologia , Calpaína/metabolismo , Caspase 3/metabolismo , Morte Celular , Células Cultivadas , Dependovirus/genética , Modelos Animais de Doenças , Feminino , Técnicas de Silenciamento de Genes , Técnicas de Transferência de Genes , Vetores Genéticos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Quinases Lim/genética , Quinases Lim/metabolismo , Compressão Nervosa , Proteínas do Tecido Nervoso/metabolismo , Neuritos/enzimologia , Neuritos/patologia , Nervo Óptico/patologia , Nervo Óptico/fisiopatologia , Traumatismos do Nervo Óptico/genética , Traumatismos do Nervo Óptico/patologia , Traumatismos do Nervo Óptico/fisiopatologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Ratos , Ratos Wistar , Células Ganglionares da Retina/patologia , Transdução de Sinais , Fatores de Tempo , Transfecção , Quinases Associadas a rho/genética
13.
Cell Death Dis ; 5: e1237, 2014 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-24853415

RESUMO

The presence of tangles composed of phosphorylated tau is one of the neuropathological hallmarks of Alzheimer's disease (AD). Tau, a microtubule (MT)-associated protein, accumulates in AD potentially as a result of posttranslational modifications, such as hyperphosphorylation and conformational changes. However, it has not been fully understood how tau accumulation and phosphorylation are deregulated. In the present study, we identified a novel role of death-associated protein kinase 1 (DAPK1) in the regulation of the tau protein. We found that hippocampal DAPK1 expression is markedly increased in the brains of AD patients compared with age-matched normal subjects. DAPK1 overexpression increased tau protein stability and phosphorylation at multiple AD-related sites. In contrast, inhibition of DAPK1 by overexpression of a DAPK1 kinase-deficient mutant or by genetic knockout significantly decreased tau protein stability and abolished its phosphorylation in cell cultures and in mice. Mechanistically, DAPK1-enhanced tau protein stability was mediated by Ser71 phosphorylation of Pin1, a prolyl isomerase known to regulate tau protein stability, phosphorylation, and tau-related pathologies. In addition, inhibition of DAPK1 kinase activity significantly increased the assembly of MTs and accelerated nerve growth factor-mediated neurite outgrowth. Given that DAPK1 has been genetically linked to late onset AD, these results suggest that DAPK1 is a novel regulator of tau protein abundance, and that DAPK1 upregulation might contribute to tau-related pathologies in AD. Therefore, we offer that DAPK1 might be a novel therapeutic target for treating human AD and other tau-related pathologies.


Assuntos
Doença de Alzheimer/enzimologia , Encéfalo/enzimologia , Proteínas Quinases Associadas com Morte Celular/metabolismo , Neurônios/enzimologia , Proteínas tau/metabolismo , Fatores Etários , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Encéfalo/patologia , Estudos de Casos e Controles , Proteínas Quinases Associadas com Morte Celular/deficiência , Proteínas Quinases Associadas com Morte Celular/genética , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Knockout , Mutação , Células NIH 3T3 , Peptidilprolil Isomerase de Interação com NIMA , Neuritos/enzimologia , Neuritos/patologia , Neurônios/patologia , Células PC12 , Peptidilprolil Isomerase/deficiência , Peptidilprolil Isomerase/genética , Fosforilação , Estabilidade Proteica , Interferência de RNA , Ratos , Fatores de Tempo , Transfecção , Proteínas tau/genética
14.
PLoS One ; 9(2): e90070, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24587210

RESUMO

Abnormal biometal homeostasis is a central feature of many neurodegenerative disorders including Alzheimer's disease (AD), Parkinson's disease (PD), and motor neuron disease. Recent studies have shown that metal complexing compounds behaving as ionophores such as clioquinol and PBT2 have robust therapeutic activity in animal models of neurodegenerative disease; however, the mechanism of neuroprotective action remains unclear. These neuroprotective or neurogenerative processes may be related to the delivery or redistribution of biometals, such as copper and zinc, by metal ionophores. To investigate this further, we examined the effect of the bis(thiosemicarbazonato)-copper complex, Cu(II)(gtsm) on neuritogenesis and neurite elongation (neurogenerative outcomes) in PC12 neuronal-related cultures. We found that Cu(II)(gtsm) induced robust neurite elongation in PC12 cells when delivered at concentrations of 25 or 50 nM overnight. Analogous effects were observed with an alternative copper bis(thiosemicarbazonato) complex, Cu(II)(atsm), but at a higher concentration. Induction of neurite elongation by Cu(II)(gtsm) was restricted to neurites within the length range of 75-99 µm with a 2.3-fold increase in numbers of neurites in this length range with 50 nM Cu(II)(gtsm) treatment. The mechanism of neurogenerative action was investigated and revealed that Cu(II)(gtsm) inhibited cellular phosphatase activity. Treatment of cultures with 5 nM FK506 (calcineurin phosphatase inhibitor) resulted in analogous elongation of neurites compared to 50 nM Cu(II)(gtsm), suggesting a potential link between Cu(II)(gtsm)-mediated phosphatase inhibition and neurogenerative outcomes.


Assuntos
Complexos de Coordenação/farmacologia , Cobre/química , Neuritos/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Tiossemicarbazonas/química , Animais , Calcineurina/metabolismo , Inibidores de Calcineurina , Complexos de Coordenação/síntese química , Cobre/metabolismo , Inibidores Enzimáticos/farmacologia , Neuritos/enzimologia , Neuritos/ultraestrutura , Fármacos Neuroprotetores/síntese química , Células PC12 , Ratos , Tacrolimo/farmacologia , Zinco/metabolismo
15.
Mol Neurobiol ; 50(1): 15-25, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24390571

RESUMO

We proposed that group IIA secretory phospholipase A(2) (GIIA) participates in neuritogenesis based on our observations that the enzyme migrates to growth cones and neurite tips when PC12 cells are induced to differentiate by nerve growth factor (NGF) (Ferrini et al., Neurochem Res 35:2168-2174, 2010). The involvement of other secretory PLA(2) isoforms in neuronal development has been suggested by others but through different mechanisms. In the present study, we compared the subcellular distribution of GIIA and group X sPLA(2) (GX) after stimulation of PC12 cells with NGF. We found that GIIA, but not GX, localized at the neuritic tips after treatment with NGF, as demonstrated by immunofluorescence analysis. We also found that NGF stimulated the expression and the activity of GIIA. In addition, NGF induced the expressed myc-tagged GIIA protein to migrate to neurite tips in its active form. We propose that GIIA expression, activity, and subcellular localization is regulated by NGF and that the enzyme may participate in neuritogenesis through intracellular mechanisms, most likely by facilitating the remodelling of glycerophospholipid molecular species by deacylation-reacylation reactions necessary for the incorporation of polyunsaturated fatty acids.


Assuntos
Fosfolipases A2 do Grupo II/metabolismo , Fator de Crescimento Neural/farmacologia , Neuritos/enzimologia , Neurogênese/efeitos dos fármacos , Animais , Fosfolipases A2 do Grupo II/genética , Neuritos/efeitos dos fármacos , Células PC12 , Ratos
16.
Cell Signal ; 26(1): 9-18, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24056044

RESUMO

RACK1 binds proteins in a constitutive or transient manner and supports signal transmission by engaging in diverse and distinct signalling pathways. The emerging theme is that RACK1 functions as a signalling switch, recruiting proteins to form distinct molecular complexes. In focal adhesions, RACK1 is required for the regulation of FAK activity and for integrating a wide array of cellular signalling events including the integration of growth factor and adhesion signalling pathways. FAK is required for cell adhesion and migration and has a well-established role in neurite outgrowth and in the developing nervous system. However, the mechanism by which FAK activity is regulated in neurons remains unknown. Using neuronal cell lines, we determined that differentiation of these cells promotes an interaction between the scaffolding protein RACK1 and FAK. Disruption of the RACK1/FAK interaction leads to decreased neurite outgrowth suggesting a role for the interaction in neurite extension. We hypothesised that RACK1 recruits proteins to FAK, to regulate FAK activity in neuronal cells. To address this, we immunoprecipitated RACK1 from rat hippocampus and searched for interacting proteins by mass spectrometry. We identified AGAP2 as a novel RACK1-interacting protein. Having confirmed the RACK1-AGAP2 interaction biochemically, we show RACK1-AGAP2 to localise together in the growth cone of differentiated cells, and confirm that these proteins are in complex with FAK. This complex is disrupted when RACK1 expression is suppressed using siRNA or when mutants of RACK1 that do not interact with FAK are expressed in cells. Similarly, suppression of AGAP2 using siRNA leads to increased phosphorylation of FAK and increased cell adhesion resulting in decreased neurite outgrowth. Our results suggest that RACK1 scaffolds AGAP2 to FAK to regulate FAK activity and cell adhesion during the differentiation process.


Assuntos
Quinase 1 de Adesão Focal/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Neuritos/metabolismo , Sequência de Aminoácidos , Animais , Diferenciação Celular , Hipocampo/citologia , Masculino , Dados de Sequência Molecular , Proteínas Monoméricas de Ligação ao GTP/química , Mutação/genética , Neuritos/enzimologia , Células PC12 , Fosforilação , Ligação Proteica , Transporte Proteico , Ratos , Ratos Sprague-Dawley , Receptores de Quinase C Ativada , Reprodutibilidade dos Testes
17.
J Histochem Cytochem ; 61(12): 857-68, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24004859

RESUMO

Lipoprotein lipase (LPL) is involved in regulation of fatty acid metabolism, and facilitates cellular uptake of lipoproteins, lipids and lipid-soluble vitamins. We evaluated LPL distribution in healthy and Alzheimer's disease (AD) brain tissue and its relative levels in cerebrospinal fluid. LPL immunostaining is widely present in different neuronal subgroups, microglia, astrocytes and oligodendroglia throughout cerebrum, cerebellum and spinal cord. LPL immunoreactivity is also present in leptomeninges, small blood vessels, choroid plexus and ependymal cells, Schwann cells associated with cranial nerves, and in anterior and posterior pituitary. In vitro studies have shown presence of secreted LPL in conditioned media of human cortical neuronal cell line (HCN2) and neuroblastoma cells (SK-N-SH), but not in media of cultured primary human astrocytes. LPL was present in cytoplasmic and nuclear fractions of neuronal cells and astrocytes in vitro. LPL immunoreactivity strongly associates with AD-related pathology, staining diffuse plaques, dystrophic and swollen neurites, possible Hirano bodies and activated glial cells. We observed no staining associated with neurofibrillary tangles or granulovacuolar degeneration. Granule cells of the dentate gyrus and the associated synaptic network showed significantly reduced staining in AD compared to control tissue. LPL was also reduced in AD CSF samples relative to those in controls.


Assuntos
Doença de Alzheimer/enzimologia , Giro Denteado/enzimologia , Lipase Lipoproteica/metabolismo , Neuritos/enzimologia , Neuritos/patologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Giro Denteado/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
18.
BMC Complement Altern Med ; 13: 204, 2013 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-23915016

RESUMO

BACKGROUND: In order to gain insight into neuroprotective effects of ECa 233, a standardized extract of Centella asiatica, previously demonstrated in animal models of memory impairment induced by transient global ischemia or intracerebroventricular injection of ß-amyloid, the effect of ECa 233 on neurite outgrowth of human IMR-32 neuroblastoma cell line was investigated. METHODS: Cells were seeded and incubated with various concentrations of ECa 233. Morphometric analysis was carried out by a measurement of the longest neurite growth of cells at 24 and 48 h. Contributing signaling pathways possibly involved were subsequently elucidated by western blot analysis. RESULTS: While ECa 233 had only limited effects on cell viability, it significantly enhanced neurite outgrowth of IMR-32 cells at the concentrations of 1-100 µg/ml. Western blot analysis revealed that ECa 233 significantly upregulated the level of activated ERK1/2 and Akt of the treated cells suggesting their involvement in the neuritogenic effect observed, which was subsequently verified by the finding that an addition of their respective inhibitors could reverse the effect of ECa 233 on these cells. CONCLUSIONS: The present study clearly demonstrated neurite outgrowth promoting activity of ECa 233. ERK1/2 and Akt signaling pathways seemed to account for the neurotrophic effect observed. In conjunction with in vivo neuroprotective effect of ECa 233 previously reported, the results obtained support further development of ECa 233 for clinical use in neuronal injury or neurodegenerative diseases.


Assuntos
Centella/química , Neuritos/efeitos dos fármacos , Neuroblastoma/patologia , Fármacos Neuroprotetores/farmacologia , Extratos Vegetais/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neuritos/enzimologia , Neuritos/patologia , Neuroblastoma/enzimologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
19.
Schizophr Res ; 147(1): 14-23, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23583326

RESUMO

Recent studies have suggested that neurodegeneration is involved in the pathogenesis of schizophrenia, and some atypical antipsychotics appear to prevent or retard progressive morphological brain changes. However, the underlying molecular mechanisms are largely unknown. Whether changes in intracellular signaling pathways are related to their neuroprotective effects remains undefined. In the present study, we used mouse embryonic prefrontal cortical neurons to examine the neuroprotection of paliperidone against the neuronal damage induced by exposure to the NMDA receptor antagonist, MK-801. Paliperidone inhibited MK-801 induced neurotoxicity both in MTT metabolism assay (p<0.01) and in lactate dehydrogenase (LDH) activity assay (p<0.01). Time course studies revealed that paliperidone effectively attenuated the elevation of intracellular free calcium concentration ([Ca(2+)]i) induced by exposure to MK-801 (p<0.01). Moreover, paliperidone could significantly retard MK-801-mediated inhibition of neurite outgrowth (p<0.01) and reverse MK-801-induced decreases of gene expression and phosphorylation of Akt1 and GSK3ß (both p<0.01). Furthermore, these protective effects of paliperidone were blocked by pretreatment with a PI3K inhibitor LY294002. Taking together, our results demonstrated that paliperidone could protect prefrontal cortical neurons from MK-801-induced damages via Akt1/GSK3ß signaling pathway.


Assuntos
Antipsicóticos/farmacologia , Quinase 3 da Glicogênio Sintase/metabolismo , Isoxazóis/farmacologia , Neurônios/efeitos dos fármacos , Córtex Pré-Frontal/citologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Cálcio/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Maleato de Dizocilpina/toxicidade , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/toxicidade , Feminino , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , L-Lactato Desidrogenase/metabolismo , Camundongos , Neuritos/efeitos dos fármacos , Neuritos/enzimologia , Palmitato de Paliperidona , Gravidez , Proteínas Proto-Oncogênicas c-akt/genética , Fatores de Tempo , Transcriptoma/efeitos dos fármacos
20.
Exp Mol Med ; 45: e16, 2013 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-23538529

RESUMO

Neurite outgrowth, a cell differentiation process involving membrane morphological changes, is critical for neuronal network and development. The membrane lipid, phosphatidylinositol (PI) 4,5-bisphosphate (PIP2), is a key regulator of many important cell surface events of membrane signaling, trafficking and dynamics. This lipid is produced mainly by the type I PI 4-phosphate 5-kinase (PIP5K) family members. In this study, we addressed whether PIP5Kα, an isoform of PIP5K, could have a role in neurite outgrowth induced by nerve growth factor (NGF). For this purpose, we knocked down PIP5Kα in PC12 rat pheochromocytoma cells by stable expression of PIP5Kα microRNA that significantly reduced PIP5Kα expression and PIP2 level. Interestingly, NGF-induced neurite outgrowth was more prominent in PIP5Kα-knockdown (KD) cells than in control cells. Conversely, add-back of PIP5Kα into PIP5Kα KD cells abrogated the effect of NGF on neurite outgrowth. NGF treatment activated PI 3-kinase (PI3K)/Akt pathway, which seemed to be associated with reactive oxygen species generation. Similar to the changes in neurite outgrowth, the PI3K/Akt activation by NGF was potentiated by PIP5Kα KD, but was attenuated by the reintroduction of PIP5Kα. Moreover, exogenously applied PIP2 to PIP5Kα KD cells also suppressed Akt activation by NGF. Together, our results suggest that PIP5Kα acts as a negative regulator of NGF-induced neurite outgrowth by inhibiting PI3K/Akt signaling pathway in PC12 cells.


Assuntos
Fator de Crescimento Neural/farmacologia , Neuritos/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Ativação Enzimática/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Camundongos , Neuritos/efeitos dos fármacos , Células PC12 , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA