Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
EMBO Rep ; 23(8): e53659, 2022 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-35766170

RESUMO

Neuronal presynaptic terminals contain hundreds of neurotransmitter-filled synaptic vesicles (SVs). The morphologically uniform SVs differ in their release competence segregating into functional pools that differentially contribute to neurotransmission. The presynaptic scaffold bassoon is required for neurotransmission, but the underlying molecular mechanisms are unknown. We report that glutamatergic synapses lacking bassoon feature decreased SV release competence and increased resting pool of SVs as assessed by imaging of SV release in cultured neurons. CDK5/calcineurin and cAMP/PKA presynaptic signalling are dysregulated, resulting in an aberrant phosphorylation of their downstream effectors synapsin1 and SNAP25, well-known regulators of SV release competence. An acute pharmacological restoration of physiological CDK5 and cAMP/PKA activity fully normalises the SV pools in neurons lacking bassoon. Finally, we demonstrate that CDK5-dependent regulation of PDE4 activity interacts with cAMP/PKA signalling and thereby controls SV release competence. These data reveal that bassoon organises SV pools in glutamatergic synapses via regulation of presynaptic phosphorylation and cAMP homeostasis and indicate a role of CDK5/PDE4/cAMP axis in the control of neurotransmitter release.


Assuntos
Proteínas do Tecido Nervoso , Vesículas Sinápticas , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fosforilação , Terminações Pré-Sinápticas/metabolismo , Sinapses/fisiologia , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/fisiologia
2.
J Biol Chem ; 298(8): 102245, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35835216

RESUMO

Cortical glutamate and midbrain dopamine neurotransmission converge to mediate striatum-dependent behaviors, while maladaptations in striatal circuitry contribute to mental disorders. However, the crosstalk between glutamate and dopamine signaling has not been entirely elucidated. Here we uncover a molecular mechanism by which glutamatergic and dopaminergic signaling integrate to regulate cAMP-dependent protein kinase (PKA) via phosphorylation of the PKA regulatory subunit, RIIß. Using a combination of biochemical, pharmacological, neurophysiological, and behavioral approaches, we find that glutamate-dependent reduction in cyclin-dependent kinase 5 (Cdk5)-dependent RIIß phosphorylation alters the PKA holoenzyme autoinhibitory state to increase PKA signaling in response to dopamine. Furthermore, we show that disruption of RIIß phosphorylation by Cdk5 enhances cortico-ventral striatal synaptic plasticity. In addition, we demonstrate that acute and chronic stress in rats inversely modulate RIIß phosphorylation and ventral striatal infusion of a small interfering peptide that selectively targets RIIß regulation by Cdk5 improves behavioral response to stress. We propose this new signaling mechanism integrating ventral striatal glutamate and dopamine neurotransmission is important to brain function, may contribute to neuropsychiatric conditions, and serves as a possible target for the development of novel therapeutics for stress-related disorders.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico , Núcleo Accumbens , Estresse Fisiológico , Transmissão Sináptica , Animais , Corpo Estriado/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dopamina/metabolismo , Glutamatos/metabolismo , Núcleo Accumbens/fisiologia , Ratos , Transdução de Sinais , Estresse Fisiológico/fisiologia
3.
J Biol Chem ; 290(26): 16319-29, 2015 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-25971971

RESUMO

Metabotropic (slow) and ionotropic (fast) neurotransmission are integrated by intracellular signal transduction mechanisms involving protein phosphorylation/dephosphorylation to achieve experience-dependent alterations in brain circuitry. ERK is an important effector of both slow and fast forms of neurotransmission and has been implicated in normal brain function and CNS diseases. Here we characterize phosphorylation of the ERK-activating protein kinase MEK1 by Cdk5, ERK, and Cdk1 in vitro in intact mouse brain tissue and in the context of an animal behavioral paradigm of stress. Cdk5 only phosphorylates Thr-292, whereas ERK and Cdk1 phosphorylate both Thr-292 and Thr-286 MEK1. These sites interact in a kinase-specific manner and inhibit the ability of MEK1 to activate ERK. Thr-292 and Thr-286 MEK1 are phosphorylated in most mouse brain regions to stoichiometries of ~5% or less. Phosphorylation of Thr-292 MEK1 is regulated by cAMP-dependent signaling in mouse striatum in a manner consistent with negative feedback inhibition in response to ERK activation. Protein phosphatase 1 and 2A contribute to the maintenance of the basal phosphorylation state of both Thr-292 and Thr-286 MEK1 and that of ERK. Activation of the NMDA class of ionotropic glutamate receptors reduces inhibitory MEK1 phosphorylation, whereas forced swim, a paradigm of acute stress, attenuates Thr-292 MEK1 phosphorylation. Together, the data indicate that these inhibitory MEK1 sites phosphorylated by Cdk5 and ERK1 serve as mechanistic points of convergence for the regulation of ERK signaling by both slow and fast neurotransmission.


Assuntos
Encéfalo/enzimologia , MAP Quinase Quinase 1/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Motivos de Aminoácidos , Animais , Encéfalo/metabolismo , Proteína Quinase CDC2/genética , Proteína Quinase CDC2/metabolismo , Quinase 5 Dependente de Ciclina/genética , Quinase 5 Dependente de Ciclina/metabolismo , MAP Quinase Quinase 1/química , MAP Quinase Quinase 1/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 3 Ativada por Mitógeno/genética , Fosforilação
4.
J Neurochem ; 138(2): 317-27, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26998748

RESUMO

Traumatic brain injury (TBI) is associated with adverse effects on brain functions, including sensation, language, emotions and/or cognition. Therapies for improving outcomes following TBI are limited. A better understanding of the pathophysiological mechanisms of TBI may suggest novel treatment strategies to facilitate recovery and improve treatment outcome. Aberrant activation of cyclin-dependent kinase 5 (Cdk5) has been implicated in neuronal injury and neurodegeneration. Cdk5 is a neuronal protein kinase activated via interaction with its cofactor p35 that regulates numerous neuronal functions, including synaptic remodeling and cognition. However, conversion of p35 to p25 via Ca(2+) -dependent activation of calpain results in an aberrantly active Cdk5/p25 complex that is associated with neuronal damage and cell death. Here, we show that mice subjected to controlled cortical impact (CCI), a well-established experimental TBI model, exhibit increased p25 levels and consistently elevated Cdk5-dependent phosphorylation of microtubule-associated protein tau and retinoblastoma (Rb) protein in hippocampal lysates. Moreover, CCI-induced neuroinflammation as indicated by increased astrocytic activation and number of reactive microglia. Brain-wide conditional Cdk5 knockout mice (Cdk5 cKO) subjected to CCI exhibited significantly reduced edema, ventricular dilation, and injury area. Finally, neurophysiological recordings revealed that CCI attenuated excitatory post-synaptic potential field responses in the hippocampal CA3-CA1 pathway 24 h after injury. This neurophysiological deficit was attenuated in Cdk5 cKO mice. Thus, TBI induces increased levels of p25 generation and aberrant Cdk5 activity, which contributes to pathophysiological processes underlying TBI progression. Hence, selectively preventing aberrant Cdk5 activity may be an effective acute strategy to improve recovery from TBI. Traumatic brain injury (TBI) increases astrogliosis and microglial activation. Moreover, TBI deregulates Ca(2+) -homeostasis triggering p25 production. The protein kinase Cdk5 is aberrantly activated by p25 leading to phosphorylation of substrates including tau and Rb protein. Loss of Cdk5 attenuates TBI lesion size, indicating that Cdk5 is a critical player in TBI pathogenesis and thus may be a suitable therapeutic target for TBI.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Encéfalo/metabolismo , Quinase 5 Dependente de Ciclina/metabolismo , Neurônios/metabolismo , Animais , Calpaína/metabolismo , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Proteínas tau/metabolismo
5.
J Neurosci ; 34(24): 8259-67, 2014 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-24920629

RESUMO

Ischemic stroke is one of the leading causes of morbidity and mortality. Treatment options are limited and only a minority of patients receive acute interventions. Understanding the mechanisms that mediate neuronal injury and death may identify targets for neuroprotective treatments. Here we show that the aberrant activity of the protein kinase Cdk5 is a principal cause of neuronal death in rodents during stroke. Ischemia induced either by embolic middle cerebral artery occlusion (MCAO) in vivo or by oxygen and glucose deprivation in brain slices caused calpain-dependent conversion of the Cdk5-activating cofactor p35 to p25. Inhibition of aberrant Cdk5 during ischemia protected dopamine neurotransmission, maintained field potentials, and blocked excitotoxicity. Furthermore, pharmacological inhibition or conditional knock-out (CKO) of Cdk5 prevented neuronal death in response to ischemia. Moreover, Cdk5 CKO dramatically reduced infarctions following MCAO. Thus, targeting aberrant Cdk5 activity may serve as an effective treatment for stroke.


Assuntos
Quinase 5 Dependente de Ciclina/metabolismo , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/metabolismo , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/prevenção & controle , Animais , Calpaína/farmacologia , Morte Celular/genética , Morte Celular/fisiologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Quinase 5 Dependente de Ciclina/genética , Modelos Animais de Doenças , Estrogênios/metabolismo , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Hipóxia/fisiopatologia , Técnicas In Vitro , Infarto da Artéria Cerebral Média/terapia , Masculino , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Fosfotransferases , Ratos , Ratos Sprague-Dawley , Sais de Tetrazólio , Fatores de Tempo , Ativador de Plasminogênio Tecidual/uso terapêutico
6.
J Biol Chem ; 288(11): 7968-7977, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23362255

RESUMO

Neurodegenerative diseases associated with the pathological aggregation of microtubule-associated protein Tau are classified as tauopathies. Alzheimer disease, the most common tauopathy, is characterized by neurofibrillary tangles that are mainly composed of abnormally phosphorylated Tau. Similar hyperphosphorylated Tau lesions are found in patients with frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17) that is induced by mutations within the tau gene. To further understand the etiology of tauopathies, it will be important to elucidate the mechanism underlying Tau hyperphosphorylation. Tau phosphorylation occurs mainly at proline-directed Ser/Thr sites, which are targeted by protein kinases such as GSK3ß and Cdk5. We reported previously that dephosphorylation of Tau at Cdk5-mediated sites was enhanced by Pin1, a peptidyl-prolyl isomerase that stimulates dephosphorylation at proline-directed sites by protein phosphatase 2A. Pin1 deficiency is suggested to cause Tau hyperphosphorylation in Alzheimer disease. Up to the present, Pin1 binding was only shown for two Tau phosphorylation sites (Thr-212 and Thr-231) despite the presence of many more hyperphosphorylated sites. Here, we analyzed the interaction of Pin1 with Tau phosphorylated by Cdk5-p25 using a GST pulldown assay and Biacore approach. We found that Pin1 binds and stimulates dephosphorylation of Tau at all Cdk5-mediated sites (Ser-202, Thr-205, Ser-235, and Ser-404). Furthermore, FTDP-17 mutant Tau (P301L or R406W) showed slightly weaker Pin1 binding than non-mutated Tau, suggesting that FTDP-17 mutations induce hyperphosphorylation by reducing the interaction between Pin1 and Tau. Together, these results indicate that Pin1 is generally involved in the regulation of Tau hyperphosphorylation and hence the etiology of tauopathies.


Assuntos
Doença de Alzheimer/metabolismo , Quinase 5 Dependente de Ciclina/fisiologia , Mutação , Peptidilprolil Isomerase/metabolismo , Proteínas tau/metabolismo , Animais , Sequência de Bases , Encéfalo/metabolismo , Células COS , Chlorocebus aethiops , Humanos , Cinética , Camundongos , Dados de Sequência Molecular , Peptidilprolil Isomerase de Interação com NIMA , Fosforilação , Plasmídeos/metabolismo , Ligação Proteica , Isoformas de Proteínas , Tauopatias/metabolismo
7.
Hippocampus ; 24(12): 1413-6, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25208523

RESUMO

Glycogen synthase kinase-3 (GSK3), particularly the isoform GSK3ß, has been implicated in a wide range of physiological systems and neurological disorders including Alzheimer's Disease. However, the functional importance of GSK3α has been largely untested. The multifunctionality of GSK3 limits its potential as a drug target because of inevitable side effects. Due to its greater expression in the CNS, GSK3ß rather than GSK3α has also been assumed to be of primary importance in synaptic plasticity. Here, we investigate bidirectional long-term synaptic plasticity in knockin mice with a point mutation in GSK3α or GSK3ß that prevents their inhibitory regulation. We report that only the mutation in GSK3α affects long-term potentiation (LTP) and depression (LTD). This stresses the importance of investigating isoform specificity for GSK3 in all systems and suggests that GSK3α should be investigated as a drug target in cognitive disorders including Alzheimer's Disease.


Assuntos
Região CA1 Hipocampal/enzimologia , Região CA3 Hipocampal/enzimologia , Quinase 3 da Glicogênio Sintase/metabolismo , Plasticidade Neuronal/fisiologia , Sinapses/enzimologia , Envelhecimento/fisiologia , Animais , Região CA1 Hipocampal/crescimento & desenvolvimento , Região CA3 Hipocampal/crescimento & desenvolvimento , Potenciais Pós-Sinápticos Excitadores/fisiologia , Técnicas de Introdução de Genes , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Isoenzimas , Masculino , Camundongos Transgênicos , Mutação , Técnicas de Cultura de Tecidos
8.
Front Pharmacol ; 13: 863762, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35645825

RESUMO

Cyclin-dependent kinase 5 (Cdk5) is a crucial regulator of neuronal signal transduction. Cdk5 activity is implicated in various neuropsychiatric and neurodegenerative conditions such as stress, anxiety, depression, addiction, Alzheimer's disease, and Parkinson's disease. While constitutive Cdk5 knockout is perinatally lethal, conditional knockout mice display resilience to stress-induction, enhanced cognition, neuroprotection from stroke and head trauma, and ameliorated neurodegeneration. Thus, Cdk5 represents a prime target for treatment in a spectrum of neurological and neuropsychiatric conditions. While intracranial infusions or treatment of acutely dissected brain tissue with compounds that inhibit Cdk5 have allowed the study of kinase function and corroborated conditional knockout findings, potent brain-penetrant systemically deliverable Cdk5 inhibitors are extremely limited, and no Cdk5 inhibitor has been approved to treat any neuropsychiatric or degenerative diseases to date. Here, we screened aminopyrazole-based analogs as potential Cdk5 inhibitors and identified a novel analog, 25-106, as a uniquely brain-penetrant anti-Cdk5 drug. We characterize the pharmacokinetic and dynamic responses of 25-106 in mice and functionally validate the effects of Cdk5 inhibition on open field and tail-suspension behaviors. Altogether, 25-106 represents a promising preclinical Cdk5 inhibitor that can be systemically administered with significant potential as a neurological/neuropsychiatric therapeutic.

9.
J Biol Chem ; 285(49): 38534-42, 2010 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-20889973

RESUMO

Progressive accumulation of DNA damage is causally involved in cellular senescence and organismal aging. The DNA damage kinase ATM plays a central role in maintaining genomic stability. ATM mutations cause the genetic disorder ataxia telangiectasia, which is primarily characterized by progressive neurodegeneration and cancer susceptibility. Although the importance of ATM function to protect against oxidative DNA damage and during aging is well described, the mechanism of ATM activation by these stimuli is not known. Here we identify ATM interactor (ATMIN) as an essential component of the ATM signaling pathway in response to oxidative stress and aging. Embryos lacking ATMIN (atmin(Δ/Δ)) died in utero and showed increased numbers of cells positive for phosphorylated histone H2aX, indicative of increased DNA damage. atmin(Δ/Δ) mouse embryonic fibroblasts accumulated DNA damage and prematurely entered senescence when cultured at atmospheric oxygen levels (20%), but this defect was rescued by addition of an antioxidant and also by culturing cells at physiological oxygen levels (3%). In response to acute oxidative stress, atmin(Δ/Δ) mouse embryonic fibroblasts showed slightly lower levels of ATM phosphorylation and reduced ATM substrate phosphorylation. Conditional deletion of ATMIN in the murine nervous system (atmin(ΔN)) resulted in reduced numbers of dopaminergic neurons, as does ATM deficiency. ATM activity was observed in old, but not in young, control mice, but aging-induced ATM signaling was impaired by ATMIN deficiency. Consequently, old atmin(ΔN) mice showed accumulation of DNA damage in the cortex accompanied by gliosis, resulting in increased mortality of aging mutant mice. These results suggest that ATMIN mediates ATM activation by oxidative stress, and thereby ATMIN protects the aging brain by preventing accumulation of DNA damage.


Assuntos
Envelhecimento/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Córtex Cerebral/metabolismo , Dano ao DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Estresse Oxidativo/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Envelhecimento/genética , Envelhecimento/patologia , Animais , Antioxidantes/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/genética , Células Cultivadas , Senescência Celular/genética , Córtex Cerebral/embriologia , Córtex Cerebral/patologia , Proteínas de Ligação a DNA/genética , Perda do Embrião/genética , Perda do Embrião/metabolismo , Perda do Embrião/patologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Histonas/genética , Histonas/metabolismo , Camundongos , Camundongos Mutantes , Neurônios/metabolismo , Neurônios/patologia , Proteínas Nucleares/genética , Oxigênio/metabolismo , Fosforilação/genética , Proteínas Serina-Treonina Quinases/genética , Fatores de Transcrição , Proteínas Supressoras de Tumor/genética
10.
J Neurochem ; 117(3): 504-15, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21338355

RESUMO

Cyclin-dependent kinase 5 (Cdk5) is a Ser/Thr kinase that is activated by binding to its regulatory subunit, p35. The calpain-mediated cleavage of p35 to p25 and the resulting aberrant activity and neurotoxicity of Cdk5 have been implicated in neurological disorders, such as Alzheimer's disease. To gain further insight into the molecular mechanisms underlying the pathological function of Cdk5, we investigated the role of the calpain inhibitor protein calpastatin (CAST), in controlling the aberrant production of p25. For this purpose, brain tissue from wild-type, CAST-over-expressing (transgenic), and CAST knockout mice were analyzed. Cleavage of p35 to p25 was increased in extracts from CAST knockout mice, compared with wild-type. Conversely, generation of p25 was not detected in brain lysates from CAST-over-expressing mice. CAST expression was 5-fold higher in mouse cerebellum than cerebral cortex. Accordingly, p25 production was lower in the cerebellum than the cerebral cortex. Furthermore, the Ca(2+) -dependent degradation of p35 by proteasome was evident when calpain was inhibited. Taken together, these results suggest that CAST is a crucial regulator of calpain activity, the production of p25, and, hence, the deregulation of Cdk5. Therefore, impairment of CAST expression and its associated mechanisms may contribute to the pathogenesis of neurodegenerative disorders.


Assuntos
Encéfalo/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Quinase 5 Dependente de Ciclina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/citologia , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/genética , Células Cultivadas , Quinase 5 Dependente de Ciclina/genética , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Neurônios/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia
11.
ACS Chem Neurosci ; 12(16): 3038-3048, 2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34370453

RESUMO

Novel treatments, screening, and detection methods have prolonged the lives of numerous cancer patients worldwide. Unfortunately, existing and many promising new chemotherapeutics can cause deleterious, off-target side effects in normal tissue and organ systems. The central and peripheral nervous systems are widely recognized as frequent off-target effectors of anticancer drugs which can produce persistent neurological and neuropsychiatric symptoms collectively termed "chemobrain". Following chemotherapy, patients report several forms of cognitive impairment occurring acutely and sometimes persisting years after treatment. There are no effective treatments for cognitive decline induced by chemotherapeutics, and the underlying molecular mechanisms are poorly characterized and understood. In this study, we find that chronic treatment with two common chemotherapeutic agents, cisplatin and gemcitabine, impairs brain region-specific metabolism, hippocampus-dependent memory formation, and stress response behavior. This corresponds to reduced hippocampal synaptic excitability, altered neuronal signal transduction, and neuroinflammation. These findings underline that a better understanding of the basic pathological consequences of chemotherapy-induced cognitive impairment is the first step toward improving cancer treatment survivorship.


Assuntos
Antineoplásicos , Doenças do Sistema Nervoso , Preparações Farmacêuticas , Antineoplásicos/efeitos adversos , Cisplatino , Hipocampo , Humanos
12.
J Vis Exp ; (165)2020 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-33226021

RESUMO

Traumatic brain injury (TBI) is a large-scale public health problem. Mild TBI is the most prevalent form of neurotrauma and accounts for a large number of medical visits in the United States. There are currently no FDA-approved treatments available for TBI. The increased incidence of military-related, blast-induced TBI further accentuates the urgent need for effective TBI treatments. Therefore, new preclinical TBI animal models that recapitulate aspects of human blast-related TBI will greatly advance the research efforts into the neurobiological and pathophysiological processes underlying mild to moderate TBI as well as the development of novel therapeutic strategies for TBI. Here we present a reliable, reproducible model for the investigation of the molecular, cellular, and behavioral effects of mild to moderate blast-induced TBI. We describe a step-by-step protocol for closed-head, blast-induced mild TBI in rodents using a bench-top setup consisting of a gas-driven shock tube equipped with piezoelectric pressure sensors to ensure consistent test conditions. The benefits of the setup that we have established are its relative low-cost, ease of installation, ease of use and high-throughput capacity. Further advantages of this non-invasive TBI model include the scalability of the blast peak overpressure and the generation of controlled reproducible outcomes. The reproducibility and relevance of this TBI model has been evaluated in a number of downstream applications, including neurobiological, neuropathological, neurophysiological and behavioral analyses, supporting the use of this model for the characterization of processes underlying the etiology of mild to moderate TBI.


Assuntos
Traumatismos por Explosões/patologia , Lesões Encefálicas Traumáticas/patologia , Animais , Comportamento Animal , Modelos Animais de Doenças , Explosões , Humanos , Masculino , Camundongos Endogâmicos C57BL , Pressão , Reprodutibilidade dos Testes
13.
Neuron ; 46(6): 917-31, 2005 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-15953420

RESUMO

NCAM 180 isoform null neuromuscular junctions are unable to effectively mobilize and exocytose synaptic vesicles and thus exhibit periods of total transmission failure during high-frequency repetitive stimulation. We have identified a highly conserved C-terminal (KENESKA) domain on NCAM that is required to maintain effective transmission and demonstrate that it acts via a pathway involving MLCK and probably myosin light chain (MLC) and myosin II. By perfecting a method of introducing peptides into adult NMJs, we tested the hypothesized role of proteins in this pathway by competitive disruption of protein-protein interactions. The effects of KENESKA and other peptides on MLCK and MLC activation and on failures in both wild-type and NCAM 180 null junctions supported this pathway, and serine phosphorylation of KENESKA was critical. We propose that this pathway is required to replenish synaptic vesicles utilized during high levels of exocytosis by facilitating myosin-driven delivery of synaptic vesicles to active zones or their subsequent exocytosis.


Assuntos
4-Aminopiridina/análogos & derivados , Estimulação Elétrica , Moléculas de Adesão de Célula Nervosa/metabolismo , Junção Neuromuscular/efeitos da radiação , Peptídeos/metabolismo , Estrutura Terciária de Proteína/fisiologia , Transmissão Sináptica/efeitos da radiação , 4-Aminopiridina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Potenciais de Ação/efeitos da radiação , Amifampridina , Animais , Animais Recém-Nascidos , Western Blotting/métodos , Bungarotoxinas/metabolismo , Biologia Computacional/métodos , Cisteamina/análogos & derivados , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Imuno-Histoquímica/métodos , Técnicas In Vitro , Camundongos , Camundongos Knockout , Modelos Neurológicos , Miosinas/metabolismo , Moléculas de Adesão de Célula Nervosa/química , Moléculas de Adesão de Célula Nervosa/deficiência , Junção Neuromuscular/efeitos dos fármacos , Junção Neuromuscular/metabolismo , Junção Neuromuscular/fisiologia , Oligopeptídeos/farmacologia , Células PC12 , Fragmentos de Peptídeos/farmacologia , Peptídeos/agonistas , Peptídeos/antagonistas & inibidores , Peptídeos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Transdução de Sinais/efeitos da radiação , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Xenopus
14.
Sci Rep ; 9(1): 16133, 2019 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-31695068

RESUMO

Huntington's disease (HD) is a fatal inherited autosomal dominant neurodegenerative disorder caused by an expansion in the number of CAG trinucleotide repeats in the huntingtin gene. The disease is characterized by motor, behavioural and cognitive symptoms for which at present there are no disease altering treatments. It has been shown that manipulating the mTOR (mammalian target of rapamycin) pathway using rapamycin or its analogue CCI-779 can improve the cellular and behavioural phenotypes of HD models. Ribosomal protein S6 kinase 1 (S6K1) is a major downstream signalling molecule of mTOR, and its activity is reduced by rapamycin suggesting that deregulation of S6K1 activity may be beneficial in HD. Furthermore, S6k1 knockout mice have increased lifespan and improvement in age-related phenotypes. To evalute the potential benefit of S6k1 loss on HD-related phenotypes, we crossed the R6/2 HD model with the long-lived S6k1 knockout mouse line. We found that S6k1 knockout does not ameliorate behavioural or physiological phenotypes in the R6/2 mouse model. Additionally, no improvements were seen in brain mass reduction or mutant huntingtin protein aggregate levels. Therefore, these results suggest that while a reduction in S6K1 signalling has beneficial effects on ageing it is unlikely to be a therapeutic strategy for HD patients.


Assuntos
Deleção de Genes , Doença de Huntington/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Animais , Modelos Animais de Doenças , Feminino , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fenótipo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo
15.
Trends Neurosci ; 29(8): 459-65, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16806507

RESUMO

Alpha Ca(2+)/calmodulin-dependent kinase II (alphaCaMKII), the major synaptic protein in the forebrain, can switch into a state of autonomous activity upon autophosphorylation. It has been proposed that alphaCaMKII autophosphorylation mediates long-term memory (LTM) storage. However, recent evidence shows that synaptic stimulation and behavioural training only transiently increase the autonomous alphaCaMKII activity, implicating alphaCaMKII autophosphorylation in LTM formation rather than storage. Consistent with this, mutant mice deficient in alphaCaMKII autophosphorylation can store LTM after a massed training protocol, but cannot form LTM after a single trial. Here, we review evidence that the role of alphaCaMKII autophosphorylation is in fact to enable LTM formation after a single training trial, possibly by regulating LTM consolidation-specific transcription.


Assuntos
Memória/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina , Humanos , Modelos Biológicos , Fosforilação , Proteínas Serina-Treonina Quinases/genética
17.
Mol Neurobiol ; 55(8): 6250-6268, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29288339

RESUMO

Cognitive deficits are a major hallmark of Huntington's disease (HD) with a great impact on the quality of patient's life. Gaining a better understanding of the molecular mechanisms underlying learning and memory impairments in HD is, therefore, of critical importance. Cdk5 is a proline-directed Ser/Thr kinase involved in the regulation of synaptic plasticity and memory processes that has been associated with several neurodegenerative disorders. In this study, we aim to investigate the role of Cdk5 in learning and memory impairments in HD using a novel animal model that expresses mutant huntingtin (mHtt) and has genetically reduced Cdk5 levels. Genetic reduction of Cdk5 in mHtt knock-in mice attenuated both corticostriatal learning deficits as well as hippocampal-dependent memory decline. Moreover, the molecular mechanisms by which Cdk5 counteracts the mHtt-induced learning and memory impairments appeared to be differentially regulated in a brain region-specific manner. While the corticostriatal learning deficits are attenuated through compensatory regulation of NR2B surface levels, the rescue of hippocampal-dependent memory was likely due to restoration of hippocampal dendritic spine density along with an increase in Rac1 activity. This work identifies Cdk5 as a critical contributor to mHtt-induced learning and memory deficits. Furthermore, we show that the Cdk5 downstream targets involved in memory and learning decline differ depending on the brain region analyzed suggesting that distinct Cdk5 effectors could be involved in cognitive impairments in HD.


Assuntos
Encéfalo/enzimologia , Encéfalo/patologia , Quinase 5 Dependente de Ciclina/metabolismo , Doença de Huntington/enzimologia , Doença de Huntington/fisiopatologia , Aprendizagem , Transtornos da Memória/enzimologia , Animais , Quinase 5 Dependente de Ciclina/genética , Espinhas Dendríticas/metabolismo , Técnicas de Introdução de Genes , Proteína Huntingtina/metabolismo , Doença de Huntington/patologia , Transtornos da Memória/patologia , Transtornos da Memória/fisiopatologia , Camundongos Endogâmicos C57BL , Atividade Motora , Neostriado/metabolismo , Neostriado/patologia , Especificidade de Órgãos , Fosforilação , Fosfotirosina/metabolismo , Subunidades Proteicas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Especificidade por Substrato , Proteínas rac1 de Ligação ao GTP/metabolismo , Quinases da Família src/metabolismo
18.
Elife ; 72018 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-30375977

RESUMO

ApoE4 genotype is the most prevalent and also clinically most important risk factor for late-onset Alzheimer's disease (AD). Available evidence suggests that the root cause for this increased risk is a trafficking defect at the level of the early endosome. ApoE4 differs from the most common ApoE3 isoform by a single amino acid that increases its isoelectric point and promotes unfolding of ApoE4 upon endosomal vesicle acidification. We found that pharmacological and genetic inhibition of NHE6, the primary proton leak channel in the early endosome, in rodents completely reverses the ApoE4-induced recycling block of the ApoE receptor Apoer2/Lrp8 and the AMPA- and NMDA-type glutamate receptors that are regulated by, and co-endocytosed in a complex with, Apoer2. Moreover, NHE6 inhibition restores the Reelin-mediated modulation of excitatory synapses that is impaired by ApoE4. Our findings suggest a novel potential approach for the prevention of late-onset AD.


Assuntos
Doença de Alzheimer/prevenção & controle , Apolipoproteína E4/metabolismo , Endossomos/metabolismo , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Animais , Apolipoproteína E4/genética , Modelos Animais de Doenças , Genótipo , Células HEK293 , Humanos , Proteínas Relacionadas a Receptor de LDL/metabolismo , Camundongos Endogâmicos C57BL , Ratos Sprague-Dawley , Receptores de Glutamato/metabolismo , Proteína Reelina
19.
Mol Brain ; 11(1): 64, 2018 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-30409147

RESUMO

Direct or indirect exposure to an explosion can induce traumatic brain injury (TBI) of various severity levels. Primary TBI from blast exposure is commonly characterized by internal injuries, such as vascular damage, neuronal injury, and contusion, without external injuries. Current animal models of blast-induced TBI (bTBI) have helped to understand the deleterious effects of moderate to severe blast forces. However, the neurological effects of mild blast forces remain poorly characterized. Here, we investigated the effects caused by mild blast forces combining neuropathological, histological, biochemical and neurophysiological analysis. For this purpose, we employed a rodent blast TBI model with blast forces below the level that causes macroscopic neuropathological changes. We found that mild blast forces induced neuroinflammation in cerebral cortex, striatum and hippocampus. Moreover, mild blast triggered microvascular damage and axonal injury. Furthermore, mild blast caused deficits in hippocampal short-term plasticity and synaptic excitability, but no impairments in long-term potentiation. Finally, mild blast exposure induced proteolytic cleavage of spectrin and the cyclin-dependent kinase 5 activator, p35 in hippocampus. Together, these findings show that mild blast forces can cause aberrant neurological changes that critically impact neuronal functions. These results are consistent with the idea that mild blast forces may induce subclinical pathophysiological changes that may contribute to neurological and psychiatric disorders.


Assuntos
Traumatismos por Explosões/patologia , Traumatismos por Explosões/fisiopatologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Animais , Traumatismos por Explosões/complicações , Encéfalo/irrigação sanguínea , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/fisiopatologia , Gliose/complicações , Gliose/patologia , Gliose/fisiopatologia , Hipocampo/patologia , Hipocampo/fisiopatologia , Inflamação/complicações , Inflamação/patologia , Inflamação/fisiopatologia , Masculino , Microglia/patologia , Microvasos/patologia , Plasticidade Neuronal , Neurônios/patologia , Proteólise , Ratos Sprague-Dawley
20.
Elife ; 62017 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-29144234

RESUMO

Low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional cell surface receptor with diverse physiological roles, ranging from cellular uptake of lipoproteins and other cargo by endocytosis to sensor of the extracellular environment and integrator of a wide range of signaling mechanisms. As a chylomicron remnant receptor, LRP1 controls systemic lipid metabolism in concert with the LDL receptor in the liver, whereas in smooth muscle cells (SMC) LRP1 functions as a co-receptor for TGFß and PDGFRß in reverse cholesterol transport and the maintenance of vascular wall integrity. Here we used a knockin mouse model to uncover a novel atheroprotective role for LRP1 in macrophages where tyrosine phosphorylation of an NPxY motif in its intracellular domain initiates a signaling cascade along an LRP1/SHC1/PI3K/AKT/PPARγ/LXR axis to regulate and integrate cellular cholesterol homeostasis through the expression of the major cholesterol exporter ABCA1 with apoptotic cell removal and inflammatory responses.


Assuntos
Aterosclerose/patologia , Colesterol/metabolismo , Homeostase , Inflamação/patologia , Macrófagos/imunologia , Receptores de LDL/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Animais , Apoptose , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Camundongos , Receptores de LDL/genética , Transdução de Sinais , Proteínas Supressoras de Tumor/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa