Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neuroscience ; 526: 204-222, 2023 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-37385335

RESUMO

Current data suggest a hypothesis of vascular pathogenesis for the development and progression of Alzheimer's disease (AD). To investigate this, we studied the association of apolipoprotein E4 (APOE4) gene on microvessels in human autopsy-confirmed AD with and without APOE4, compared with age/sex-matched control (AC) hippocampal CA1 stratum radiatum. AD arterioles (without APOE4 gene) had mild oxidative stress and loss of vascular endothelial growth factor (VEGF) and endothelial cell density, reflecting aging progression. In AD + APOE4, an increase in strong oxidative DNA damage marker 8-hydroxy-2'-deoxyguanosine (8-OHdG), VEGF, and endothelial cell density were associated with increased diameter of arterioles and perivascular space dilation. In cultured human brain microvascular cells (HBMECs), treatment of ApoE4 protein plus amyloid-ß (Aß) oligomers increased superoxide production and the apoptotic marker cleaved caspase 3, sustained hypoxia inducible factor-1α (HIF-1α) stability that was associated with an increase in MnSOD, VEGF, and cell density. This cell over-proliferation was inhibited with the antioxidants N-acetyl cysteine and MnTMPyP, the HIF-1α inhibitor echinomycin, the VEGFR-2 receptor blocker SU1498, the protein kinase C (PKC) ε knock-down (KD) and the extracellular signal-regulated kinase 1/2 (ERK) inhibitor FR180204. The PKCε KD and echinomycin decreased VEGF and/or ERK. In conclusion, AD capillaries and arterioles in hippocampal CA1 stratum radiatum of non-APOE4 carriers are related with aging, while those in APOE4 carriers with AD are related with pathogenesis of cerebrovascular disease.


Assuntos
Doença de Alzheimer , Equinomicina , Humanos , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteínas E , Região CA1 Hipocampal/patologia , Equinomicina/metabolismo , Hipocampo/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
Front Aging Neurosci ; 15: 1272361, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38187357

RESUMO

Background: Current evidence suggests that microvessel disease is involved in Alzheimer's disease (AD). Cerebrovascular disease correlates with cardiovascular disease and is complicated in ≈40% of AD patients. The protein kinase C (PKC) ε activator DCPLA can stimulate human antigen (Hu) R that prevents degradation and promotes the translation of mitochondrial Mn-superoxide dismutase (MnSOD) and vascular endothelial growth factor-A (VEGF) mRNAs. Methods: To induce brain microinfarcts, we injected triple transgenic (3×Tg) and wild-type (WT) control mice with microbeads (20 µm caliber) into common carotid arteries, with or without the DCPLA-ME (methyl-ester) for 2 weeks. After water maze training, mice at 16 months old were examined for confocal immunohistochemistry at a single cell or microvessel level in the hippocampal CA1 area, important for spatial memory storage, and in the dorsal hippocampus by western blots. Results: In 3×Tg mice without cerebral microinfarcts, an accelerating age-related increase in (mild) oxidative stress and hypoxia inducible factor (HIF)-1α, but a reduction in VEGF, mitochondrial transcription factor A (TFAM), and MnSOD were associated with capillary loss. The change was less pronounced in arterioles. However, in 3×Tg mice with cerebral microinfarcts, increasing arteriolar diameter and their wall cells were related with the strong oxidative DNA damage 8-hydroxy-2'-deoxyguanosine (8-OHdG), apoptosis (cleaved caspase 3), and sustained hypoxia (increased HIF-1α and VEGF/PKCε/extracellular signal regulated kinase or ERK pathway). Microocclusion enhanced the loss of the synaptic marker spinophilin, astrocytic number, and astrocyte-vascular coupling areas and demyelination of axons. DCPLA-ME prevented spatial memory defect; strong oxidative stress-related apoptosis; sustained hypoxia (by reducing HIF-1α and VEGF); and exaggerated cell repair in arteriolar walls, pericapillary space dilation, neuro-glial-vascular disruption, and demyelination. Conclusion: In conclusion, in 3×Tg mice with cerebral microinfarcts, sustained hypoxia (increased HIF-1α and VEGF signals) is dominant with arteriolar wall thickening, and DCPLA has a protective effect on sustained hypoxia.

3.
Mol Neurobiol ; 59(8): 4966-4986, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35665894

RESUMO

Current evidence suggests that mild cerebrovascular changes could induce neurodegeneration and contribute to HIV-associated neurocognitive disease (HAND) in HIV patients. We investigated both the quantitative and qualitative impact of HIV infection on brain microvessels, especially on hippocampal microvessels, which are crucial for optimal O2 supply, and thus for maintaining memory and cognitive abilities. The results obtained using cultured human brain microvascular endothelial cells (HBMEC) were reproduced using a suitable mouse model and autopsied human HIV hippocampus. In HBMEC, we found significantly higher oxidative stress-dependent apoptotic cell loss following 5 h of treatment of GST-Tat (1 µg/ml) compared to GST (1 µg/ml) control. We noticed complete recovery of HBMEC cells after 24 h of GST-Tat treatment, due to temporal degradation or inactivation of GST-Tat. Interestingly, we found a sustained increase in mitochondrial oxidative DNA damage marker 8-OHdG, as well as an increase in hypoxia-inducible factor hypoxia-inducible factor-1α (HIF-1α). In our mouse studies, upon short-term injection of GST-Tat, we found the loss of small microvessels (mostly capillaries) and vascular endothelial growth factor (VEGF), but not large microvessels (arterioles and venules) in the hippocampus. In addition to capillary loss, in the post-mortem HIV-infected human hippocampus, we observed large microvessels with increased wall cells and perivascular tissue degeneration. Together, our data show a crucial role of Tat in inducing HIF-1α-dependent inhibition of mitochondrial transcriptional factor A (TFAM) and dilated perivascular space. Thus, our results further define the underlying molecular mechanism promoting mild cerebrovascular disease, neuropathy, and HAND pathogenesis in HIV patients.


Assuntos
Infecções por HIV , Animais , Células Endoteliais/metabolismo , Infecções por HIV/complicações , Infecções por HIV/metabolismo , Hipocampo/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Microvasos , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Front Aging Neurosci ; 14: 836634, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35299945

RESUMO

Vascular endothelial dysfunction and capillary loss are currently considered to be a primary phenotype of normal human aging and Alzheimer's disease (AD). Activation of protein kinase C (PKCε) improves several molecular, cellular, physiological, and behavioral endpoints, yet it is not known whether a loss of PKCε activity occurs in the microvascular endothelium in aged and AD hippocampi, whether this loss contributes to microvascular change, or whether activation of PKCε protects against microvascular damage, an early change that induces age-associated memory defect and AD. We investigated the effect of the PKCε activation on microvascular loss in the hippocampus, important for memory storage. In cultured human brain microvascular endothelial cells, tert-butyl hydroperoxide induced oxidative stress and a decrease in manganese superoxide dismutase (MnSOD) mRNA and protein expression that were blocked by the antioxidant drugs. The PKCε activators bryostatin and DCPLA methyl ester increased PKCε, associated with an increase in MnSOD mRNA and its protein as well as vascular endothelial growth factor (VEGF), which was inhibited by the mRNA-stabilizing HuR inhibitors. In rats (>24 months old) and AD transgenic mice Tg2576 (5 months old), bryostatin or DCP-LA prevented a decrease in vascular PKCε, MnSOD, and VEGF and prevented microvascular loss and age-related memory impairment. An autopsy-confirmed AD hippocampus showed a decrease in PKCε and MnSOD mRNAs and their proteins and VEGF as well as in microvascular density compared to non-AD controls. In conclusion, the PKCε activation can rescue a decrease in PKCε, MnSOD, and VEGF via posttranscription regulation and alleviate oxidative stress, and in doing so, prevent microvascular loss during aging and AD.

5.
J Alzheimers Dis ; 63(3): 1173-1189, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29710707

RESUMO

Oxidative stress and amyloid-ß (Aß) oligomers have been implicated in Alzheimer's disease (AD). The growth and maintenance of neuronal networks are influenced by brain derived neurotrophic factor (BDNF) expression, which is promoted by protein kinase C epsilon (PKCɛ). We investigated the reciprocal interaction among oxidative stress, Aß, and PKCɛ levels and subsequent PKCɛ-dependent MnSOD and BDNF expression in hippocampal pyramidal neurons. Reduced levels of PKCɛ, MnSOD, and BDNF and an increased level of Aß were also found in hippocampal neurons from autopsy-confirmed AD patients. In cultured human primary hippocampal neurons, spherical aggregation of Aß (amylospheroids) decreased PKCɛ and MnSOD. Treatment with t-butyl hydroperoxide (TBHP) increased superoxide, the oxidative DNA/RNA damage marker, 8-OHG, and Aß levels, but reduced PKCɛ, MnSOD, BDNF, and cultured neuron density. These changes were reversed with the PKCɛ activators, bryostatin and DCPLA-ME. PKCɛ knockdown suppressed PKCɛ, MnSOD, and BDNF but increased Aß. In cultured neurons, the increase in reactive oxygen species (ROS) associated with reduced PKCɛ during neurodegeneration was inhibited by the SOD mimetic MnTMPyP and the ROS scavenger NAc, indicating that strong oxidative stress suppresses PKCɛ level. Reduction of PKCɛ and MnSOD was prevented with the PKCɛ activator bryostatin in 5-6-month-old Tg2576 AD transgenic mice. In conclusion, oxidative stress and Aß decrease PKCɛ expression. Reciprocally, a depression of PKCɛ reduces BDNF and MnSOD, resulting in oxidative stress. These changes can be prevented with the PKCɛ-specific activators.


Assuntos
Doença de Alzheimer/patologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Regulação para Baixo/fisiologia , Hipocampo/patologia , Neurônios/metabolismo , Proteína Quinase C-épsilon/deficiência , Adjuvantes Imunológicos/farmacologia , Idoso , Idoso de 80 Anos ou mais , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Animais , Briostatinas/metabolismo , Briostatinas/farmacologia , Células Cultivadas , Feminino , Feto/anatomia & histologia , Hipocampo/citologia , Hipocampo/metabolismo , Humanos , Masculino , Metaloporfirinas/farmacologia , Camundongos , Pessoa de Meia-Idade , Morfolinos/farmacologia , Proteína Quinase C-épsilon/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Transfecção , terc-Butil Hidroperóxido/farmacologia
6.
Free Radic Biol Med ; 120: 192-203, 2018 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-29572097

RESUMO

Vascular endothelial dysfunction is a primary phenotype of aging, and microvascular (MV) lesion is mainly associated with Alzheimer's disease (AD). Here we have studied the correlation of MV wall thickness and CA1 pyramidal neuronal pathology in autopsy-confirmed AD brains. Both hyaline (h-MV) and increased cell number (c-MV) associated MV wall thickening was found in age-matched control (AC) hippocampus without significant change in Aß level (Braak stages 0-III). AC neurons neighboring the h-MV showed lower levels of oxidative DNA/RNA damage and Aß precursor protein (APP), while the neurons around c-MV showed higher oxidative DNA/RNA damage with increased APP expression. Neurons in AC hippocampus without MV wall thickening (thin wall) showed increased DNA/RNA damage and APP levels compared to AC cases with h-MV and c-MV walls. In the AD hippocampus neurons neighboring h-MV walls showed increased levels of Aß and decreased number of dendritic spines (at Braak stages IV-VI). C-MV neighboring neurons in the AD cases showed higher levels of DNA/RNA damage with increased APP at stages II - III, followed by lower levels of oxidative DNA/RNA damage, decreased APP and increased Aß levels with loss of dendritic spines at stages IV-VI. Prolonged treatment of primary human fetal hippocampal neurons with tert-butyl hydroperoxide (TBHP) induced oxidative DNA damage with a sustained increase in APP. Aß increased rapidly and then decreased overtime. Short-term TBHP treated neurons showed lower levels of superoxide (O2• -) without significant DNA damage. Short-term TBHP treatment induced a gradual decrease in APP but an increase in Aß levels over time. In conclusion this study indicates that AD hippocampus at Braak stages II-III are characterized by strong oxidative DNA/RNA damage with increased APP in neurons associated with c-MV, while stages IV-VI are characterized by a slow increase in Aß in neurons neighboring both h-MV and c-MV.


Assuntos
Doença de Alzheimer/patologia , Região CA1 Hipocampal/patologia , Microvasos/patologia , Estresse Oxidativo/fisiologia , Células Piramidais/patologia , Idoso , Idoso de 80 Anos ou mais , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Região CA1 Hipocampal/irrigação sanguínea , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
7.
Eur J Pharmacol ; 807: 71-74, 2017 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-28472666

RESUMO

Despite over a half-century's intensive research worldwide, the currently available antidepressants remain sub-optimal. Therapeutic options for treatment-resistant depression, for instance, are rather limited. Here, we found that rats exhibited a lasting treatment-resistant depressive immobility in response to open space swim test at a high intensity of induction. The induced depressive behavior is associated with a dramatic impairment in spatial learning and memory. Both the depressive immobility and impairment in spatial learning and memory are sensitive to a period of chronic treatment with bryopstatin-1, a relatively selective activator of protein kinase Cε. Bryostatin-1-like analogues therefore might have therapeutic values for the treatment of treatment-resistant depression.


Assuntos
Briostatinas/farmacologia , Transtorno Depressivo Resistente a Tratamento/tratamento farmacológico , Animais , Comportamento Animal/efeitos dos fármacos , Briostatinas/uso terapêutico , Transtorno Depressivo Resistente a Tratamento/fisiopatologia , Masculino , Ratos , Ratos Wistar , Aprendizagem Espacial/efeitos dos fármacos , Fatores de Tempo
8.
J Biol Chem ; 291(32): 16462-76, 2016 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-27330081

RESUMO

Protein kinase Cϵ (PKCϵ) promotes synaptic maturation and synaptogenesis via activation of synaptic growth factors such as BDNF, NGF, and IGF. However, many of the detailed mechanisms by which PKCϵ induces synaptogenesis are not fully understood. Accumulation of PSD-95 to the postsynaptic density (PSD) is known to lead to synaptic maturation and strengthening of excitatory synapses. Here we investigated the relationship between PKCϵ and PSD-95. We show that the PKCϵ activators dicyclopropanated linoleic acid methyl ester and bryostatin 1 induce phosphorylation of PSD-95 at the serine 295 residue, increase the levels of PSD-95, and enhance its membrane localization. Elimination of the serine 295 residue in PSD-95 abolished PKCϵ-induced membrane accumulation. Knockdown of either PKCϵ or JNK1 prevented PKCϵ activator-mediated membrane accumulation of PSD-95. PKCϵ directly phosphorylated PSD-95 and JNK1 in vitro Inhibiting PKCϵ, JNK, or calcium/calmodulin-dependent kinase II activity prevented the effects of PKCϵ activators on PSD-95 phosphorylation. Increase in membrane accumulation of PKCϵ and phosphorylated PSD-95 (p-PSD-95(S295)) coincided with an increased number of synapses and increased amplitudes of excitatory post-synaptic potentials (EPSPs) in adult rat hippocampal slices. Knockdown of PKCϵ also reduced the synthesis of PSD-95 and the presynaptic protein synaptophysin by 30 and 44%, respectively. Prolonged activation of PKCϵ increased synapse number by 2-fold, increased presynaptic vesicle density, and greatly increased PSD-95 clustering. These results indicate that PKCϵ promotes synaptogenesis by activating PSD-95 phosphorylation directly through JNK1 and calcium/calmodulin-dependent kinase II and also by inducing expression of PSD-95 and synaptophysin.


Assuntos
Hipocampo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Proteínas de Membrana/biossíntese , Proteína Quinase C-épsilon/metabolismo , Membranas Sinápticas/metabolismo , Animais , Briostatinas/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteína 4 Homóloga a Disks-Large , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Proteína Quinase C-épsilon/genética , Ratos , Membranas Sinápticas/genética , Sinaptofisina/biossíntese , Sinaptofisina/genética
9.
J Pharmacol Exp Ther ; 357(2): 300-10, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26941170

RESUMO

Fragile X syndrome (FXS) is characterized by synaptic immaturity, cognitive impairment, and behavioral changes. The disorder is caused by transcriptional shutdown in neurons of thefragile X mental retardation 1gene product, fragile X mental retardation protein. Fragile X mental retardation protein is a repressor of dendritic mRNA translation and its silencing leads to dysregulation of synaptically driven protein synthesis and impairments of intellect, cognition, and behavior, and FXS is a disorder that currently has no effective therapeutics. Here, young fragile X mice were treated with chronic bryostatin-1, a relatively selective protein kinase Cεactivator, which induces synaptogenesis and synaptic maturation/repair. Chronic treatment with bryostatin-1 rescues young fragile X mice from the disorder phenotypes, including normalization of most FXS abnormalities in 1) hippocampal brain-derived neurotrophic factor expression, 2) postsynaptic density-95 levels, 3) transformation of immature dendritic spines to mature synapses, 4) densities of the presynaptic and postsynaptic membranes, and 5) spatial learning and memory. The therapeutic effects were achieved without downregulation of metabotropic glutamate receptor (mGluR) 5 in the hippocampus and are more dramatic than those of a late-onset treatment in adult fragile X mice. mGluR5 expression was in fact lower in fragile X mice and its expression was restored with the bryostatin-1 treatment. Our results show that synaptic and cognitive function of young FXS mice can be normalized through pharmacological treatment without downregulation of mGluR5 and that bryostatin-1-like agents may represent a novel class of drugs to treat fragile X mental retardation at a young age and in adults.


Assuntos
Síndrome do Cromossomo X Frágil/tratamento farmacológico , Transtornos da Memória/tratamento farmacológico , Memória Espacial/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Briostatinas/farmacologia , Espinhas Dendríticas/efeitos dos fármacos , Proteína 4 Homóloga a Disks-Large , Ativadores de Enzimas/farmacologia , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/patologia , Síndrome do Cromossomo X Frágil/psicologia , Guanilato Quinases/genética , Guanilato Quinases/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Transtornos da Memória/etiologia , Transtornos da Memória/psicologia , Camundongos , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Proteína Quinase C-épsilon/efeitos dos fármacos , Receptor de Glutamato Metabotrópico 5/metabolismo , Receptores Pré-Sinápticos/efeitos dos fármacos , Sinapses/patologia
10.
J Alzheimers Dis ; 43(2): 491-509, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25125477

RESUMO

In Alzheimer's disease (AD) transgenic mice, activation of synaptogenic protein kinase C ε (PKCε) was found to prevent synaptotoxic amyloid-ß (Aß)-oligomer elevation, PKCε deficits, early synaptic loss, cognitive deficits, and amyloid plaque formation. In humans, to study the role of PKCε in the pathophysiology of AD and to evaluate its possible use as an early AD-biomarker, we examined PKCε and Aß in the brains of autopsy-confirmed AD patients (n = 20) and age-matched controls (AC, n = 19), and in skin fibroblast samples from AD (n = 14), non-AD dementia patients (n = 14), and AC (n = 22). Intraneuronal Aß levels were measured immunohistochemically (using an Aß-specific antibody) in hippocampal pyramidal cells of human autopsy brains. PKCε was significantly lower in the hippocampus and temporal pole areas of AD brains, whereas Aß levels were significantly higher. The ratio of PKCε to Aß in individual CA1 pyramidal cells was markedly lower in the autopsy AD brains versus controls. PKCε was inversely correlated with Aß levels in controls, whereas in AD patients, PKCε showed no significant correlation with Aß. In autopsy brains, PKCε decreased as the Braak score increased. Skin fibroblast samples from AD patients also demonstrated a deficit in PKCε compared to controls and an AD-specific change in the Aß-oligomer effects on PKCε. Together, these data demonstrate that the relationship between Aß levels and PKCε is markedly altered in AD patients' brains and skin fibroblasts, reflecting a loss of protective effect of PKCε against toxic Aß accumulation. These changes of PKCε levels in human skin fibroblasts may provide an accurate, non-invasive peripheral AD biomarker.


Assuntos
Doença de Alzheimer/enzimologia , Doença de Alzheimer/patologia , Encéfalo/enzimologia , Fibroblastos/enzimologia , Proteína Quinase C/deficiência , Pele/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/farmacologia , Análise de Variância , Autopsia , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Feminino , Fibroblastos/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Proteína Quinase C/genética , RNA Mensageiro/metabolismo
11.
J Pharmacol Exp Ther ; 349(3): 393-401, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24659806

RESUMO

Fragile X syndrome (FXS) is caused by transcriptional silencing in neurons of the FMR1 gene product, fragile X mental retardation protein (FMRP), a repressor of dendritic mRNA translation. The lack of FMRP leads to dysregulation of synaptically driven protein synthesis and impairments of intellect, cognition, and behavior, a disorder that currently has no effective therapeutics. Fragile X mice were treated with chronic bryostatin-1, a relatively selective protein kinase ε activator with pharmacological profiles of rapid mGluR desensitization, synaptogenesis, and synaptic maturation/repairing. Differences in the major FXS phenotypes, synapses, and cognitive functions were evaluated and compared among the age-matched groups. Long-term treatment with bryostatin-1 rescues adult fragile X mice from the disorder phenotypes, including normalization of most FXS abnormalities in hippocampal brain-derived neurotrophic factor expression and secretion, postsynaptic density-95 levels, glycogen synthase kinase-3ß phosphorylation, transformation of immature dendritic spines to mature synapses, densities of the presynaptic and postsynaptic membranes, and spatial learning and memory. Our results show that synaptic and cognitive function of adult FXS mice can be normalized through pharmacologic treatment and that bryostatin-1-like agents may represent a novel class of drugs to treat fragile X mental retardation even after postpartum brain development has largely completed.


Assuntos
Briostatinas/uso terapêutico , Síndrome do Cromossomo X Frágil/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Comportamento Espacial/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Animais , Briostatinas/administração & dosagem , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/patologia , Modelos Animais de Doenças , Síndrome do Cromossomo X Frágil/metabolismo , Síndrome do Cromossomo X Frágil/patologia , Síndrome do Cromossomo X Frágil/fisiopatologia , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos , Microscopia Confocal , Microscopia Eletrônica , Proteína Quinase C-épsilon/metabolismo , Sinapses/metabolismo , Sinapses/fisiologia , Sinapses/ultraestrutura
12.
Stroke ; 44(12): 3490-7, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24172582

RESUMO

BACKGROUND AND PURPOSE: Bryostatin, a potent protein kinase C (PKC) activator, has demonstrated therapeutic efficacy in preclinical models of associative memory, Alzheimer disease, global ischemia, and traumatic brain injury. In this study, we tested the hypothesis that administration of bryostatin provides a therapeutic benefit in reducing brain injury and improving stroke outcome using a clinically relevant model of cerebral ischemia with tissue plasminogen activator reperfusion in aged rats. METHODS: Acute cerebral ischemia was produced by reversible occlusion of the right middle cerebral artery (MCAO) in 18- to 20-month-old female Sprague-Dawley rats using an autologous blood clot with tissue plasminogen activator-mediated reperfusion. Bryostatin was administered at 6 hours post-MCAO, then at 3, 6, 9, 12, 15, and 18 days after MCAO. Functional assessment was conducted at 2, 7, 14, and 21 days after MCAO. Lesion volume and hemispheric swelling/atrophy were performed at 2, 7, and 21 days post-MCAO. Histological assessment of PKC isozymes was performed at 24 hours post-MCAO. RESULTS: Bryostatin-treated rats showed improved survival post-MCAO, especially during the first 4 days. Repeated administration of bryostatin post-MCAO resulted in reduced infarct volume, hemispheric swelling/atrophy, and improved neurological function at 21 days post-MCAO. Changes in αPKC expression and εPKC expression in neurons were noted in bryostatin-treated rats at 24 hours post-MCAO. CONCLUSIONS: Repeated bryostatin administration post-MCAO protected the brain from severe neurological injury post-MCAO. Bryostatin treatment improved survival rate, reduced lesion volume, salvaged tissue in infarcted hemisphere by reducing necrosis and peri-infarct astrogliosis, and improved functional outcome after MCAO.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Briostatinas/uso terapêutico , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Encéfalo/patologia , Isquemia Encefálica/patologia , Briostatinas/farmacologia , Modelos Animais de Doenças , Feminino , Gliose/tratamento farmacológico , Gliose/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/patologia , Taxa de Sobrevida
13.
Neurobiol Dis ; 55: 44-62, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23545166

RESUMO

Protein kinase C (PKC) ε and α activation has been implicated in synaptogenesis. We used aged rats to test whether the PKCε/α activator bryostatin and PKCε-specific activator DCP-LA combined with spatial memory training could restore mushroom dendritic spinogenesis and synaptogenesis. Compared with young rats, aged, learning-impaired rats had lower memory retention; lower densities of mushroom spines and synapses in the apical dendrites of CA1 pyramidal neurons; fewer PKCε-containing presynaptic axonal boutons; and lower activation and expression of two PKCε/α substrates, the mRNA-stabilizing protein HuD and brain-derived neurotrophic factor (BDNF). PKC activator treatment combined with spatial memory training restored mushroom spines and mushroom spine synapses; rescued PKCε/α expression and PKC/HuD/BDNF signaling; and normalized memory to the levels seen in young rats. These effects were produced by treatment with either bryostatin or the PKCε-specific activator, DCP-LA. Bryostatin also reversed alterations in GABAergic inhibitory postsynaptic currents (IPSPs) in aged, learning-impaired rats. Thus, our results support the therapeutic potential of PKC activators when added to cognitive rehabilitation for inducing mushroom spine synaptogenesis and reversing memory decline associated with aging.


Assuntos
Envelhecimento , Dendritos/fisiologia , Hipocampo/citologia , Memória/fisiologia , Proteína Quinase C/metabolismo , Sinapses/fisiologia , Anestésicos Locais/farmacologia , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Briostatinas/farmacologia , Caprilatos/farmacologia , Dendritos/efeitos dos fármacos , Dendritos/ultraestrutura , Proteínas ELAV/genética , Proteínas ELAV/metabolismo , Proteína Semelhante a ELAV 4 , Estimulação Elétrica , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Lidocaína/análogos & derivados , Lidocaína/farmacologia , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Memória/efeitos dos fármacos , Técnicas de Patch-Clamp , Células Piramidais/citologia , Células Piramidais/efeitos dos fármacos , Ratos , Sinapses/efeitos dos fármacos , Fatores de Tempo
14.
J Neurosci ; 31(2): 630-43, 2011 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-21228172

RESUMO

Among the pathologic hallmarks of Alzheimer's disease (AD) neurodegeneration, only synaptic loss in the brains of AD patients closely correlates with the degree of dementia in vivo. Here, we describe a molecular basis for this AD loss of synapses: pathological reduction of synaptogenic PKC isozymes and their downstream synaptogenic substrates, such as brain-derived neurotrophic factor. This reduction, particularly of PKC α and ε, occurs in association with elevation of soluble ß amyloid protein (Aß), but before the appearance of the amyloid plaques or neuronal loss in the Tg2576 AD transgenic mouse strain. Conversely, treatment of the Tg2576 mouse brain with the PKC activator, bryostatin-1, restores normal or supranormal levels of PKC α and ε, reduces the level of soluble Aß, prevents and/or reverses the loss of hippocampal synapses, and prevents the memory impairment observed at 5 months postpartum. Similarly, the PKC ε-specific activator, DCP-LA, effectively prevents synaptic loss, amyloid plaques, and cognitive deficits (also prevented by bryostatin-1) in the much more rapidly progressing 5XFAD transgenic strain. These results suggest that synaptic loss and the resulting cognitive deficits depend on the balance between the lowering effects of Aß on PKC α and ε versus the lowering effects of PKC on Aß in AD transgenic mice.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides/metabolismo , Transtornos Cognitivos/psicologia , Proteína Quinase C-épsilon/metabolismo , Sinapses/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Briostatinas/farmacologia , Caprilatos/farmacologia , Ativadores de Enzimas/farmacologia , Hipocampo/patologia , Isoenzimas/metabolismo , Masculino , Aprendizagem em Labirinto , Memória , Camundongos , Camundongos Transgênicos , Placa Amiloide/patologia , Proteína Quinase C-alfa/metabolismo
15.
Neurobiol Dis ; 41(2): 329-37, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20951803

RESUMO

Traumatic brain injury (TBI) is a frequent consequence of vehicle, sport and war related injuries. More than 90% of TBI patients suffer mild injury (mTBI). However, the pathologies underlying the disease are poorly understood and treatment modalities are limited. We report here that in mice, the potent PKC activator bryostatin1 protects against mTBI induced learning and memory deficits and reduction in pre-synaptic synaptophysin and post-synaptic spinophylin immunostaining. An effective treatment has to start within the first 8h after injury, and includes 5 × i.p. injections over a period of 14 days. The treatment is dose dependent. Exploring the effects of the repeated bryostatin1 treatment on the processing of the amyloid precursor protein, we found that the treatment induced an increase in the putative α-secretase ADAM10 and a reduction in ß-secretase activities. Both these effects could contribute towards a reduction in ß-amyloid production. These results suggest that bryostatin1 protects against mTBI cognitive and synaptic sequela by rescuing synapses, which is possibly mediated by an increase in ADAM10 and a decrease in BACE1 activity. Since bryostatin1 has already been extensively used in clinical trials as an anti-cancer drug, its potential as a remedy for the short- and long-term TBI sequelae is quite promising.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/enzimologia , Briostatinas/farmacologia , Fármacos Neuroprotetores/farmacologia , Proteína Quinase C/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Lesões Encefálicas/fisiopatologia , Briostatinas/uso terapêutico , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/uso terapêutico
16.
Proc Natl Acad Sci U S A ; 106(34): 14676-80, 2009 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-19667190

RESUMO

Therapeutics for cerebral ischemia/hypoxia, which often results in ischemic stroke in humans, are a global unmet medical need. Here, we report that bryostatin-1, a highly potent protein kinase C (PKC) activator, interrupts pathophysiological molecular cascades and apoptosis triggered by cerebral ischemia/hypoxia, enhances neurotrophic activity, and induces synaptogenesis in rats. This postischemic therapeutic approach is further shown to preserve learning and memory capacity even 4 months later as well as long-term memory induced before the ischemic event. Our results of electromicroscopic and immunohistochemical analyses of neuronal and synaptic ultra-structure are consistent with a PKC-mediated synaptic remodeling and repair process that confers long-lasting preservation of spatial learning and memory before and after the cerebral ischemic/hypoxic event, suggesting a previously undescribed therapeutic modality for cerebral ischemia/hypoxia and ischemic stroke.


Assuntos
Isquemia Encefálica/prevenção & controle , Ativadores de Enzimas/farmacologia , Memória/efeitos dos fármacos , Proteína Quinase C/metabolismo , Animais , Apoptose/efeitos dos fármacos , Isquemia Encefálica/fisiopatologia , Briostatinas/farmacologia , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/ultraestrutura , Ativação Enzimática/efeitos dos fármacos , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipóxia/fisiopatologia , Marcação In Situ das Extremidades Cortadas , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/fisiologia , Microscopia Eletrônica , Atividade Motora/efeitos dos fármacos , Ratos , Ratos Wistar , Vesículas Sinápticas/efeitos dos fármacos , Vesículas Sinápticas/ultraestrutura , Fatores de Tempo
17.
Proc Natl Acad Sci U S A ; 105(36): 13620-5, 2008 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-18768786

RESUMO

Global cerebral ischemia/hypoxia, as can occur during human stroke, damages brain neural networks and synaptic functions. The recently demonstrated protein kinase C (PKC) activation-induced synaptogenesis in rat hippocampus suggested the potential of PKC-mediated antiapoptosis and synaptogenesis during conditions of neurodegeneration. Consequently, we examined the effects of chronic bryostatin-1, a PKC activator, on the cerebral ischemia/hypoxia-induced impairment of synapses and neurotrophic activity in the hippocampal CA1 area and on hippocampus-dependent spatial learning and memory. Postischemic/hypoxic bryostatin-1 treatment effectively rescued ischemia-induced deficits in synaptogenesis, neurotrophic activity, and spatial learning and memory. These results highlight a neuroprotective signaling pathway, as well as a therapeutic strategy with an extended time window for reducing brain damage due to stroke by activating particular PKC isozymes.


Assuntos
Diferenciação Celular , Neurônios/citologia , Neurônios/enzimologia , Proteína Quinase C/metabolismo , Acidente Vascular Cerebral/enzimologia , Acidente Vascular Cerebral/patologia , Sinapses/enzimologia , Envelhecimento/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Briostatinas/farmacologia , Sobrevivência Celular , Ativação Enzimática/efeitos dos fármacos , Hipóxia/patologia , Isoenzimas/metabolismo , Aprendizagem/efeitos dos fármacos , Masculino , Memória , Microscopia Eletrônica , Ratos , Ratos Wistar , Sinapses/ultraestrutura
18.
Eur J Pharmacol ; 585(1): 76-87, 2008 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-18402935

RESUMO

Protein kinase C (PKC) is involved in synaptic remodeling, induction of protein synthesis, and many other processes important in learning and memory. Activation of neuronal protein kinase C correlates with, and may be essential for, all phases of learning, including acquisition, consolidation, and reconsolidation. Protein kinase C activation is closely tied to hydrolysis of membrane lipids. Phospholipases C and A2 produce 1,2-diacylglycerol and arachidonic acid, which are direct activators of protein kinase C. Phospholipase C also produces inositol triphosphate, which releases calcium from internal stores. Protein kinase C interacts with many of the same pathways as insulin; therefore, it should not be surprising that insulin signaling and protein kinase C activation can both have powerful effects on memory storage and synaptic remodeling. However, investigating the possible roles of insulin in memory storage can be challenging, due to the powerful peripheral effects of insulin on glucose and the low concentration of insulin in the brain. Although peripheral for insulin, synthesized in the beta-cells of the pancreas, is primarily involved in regulating glucose, small amounts of insulin are also present in the brain. The functions of this brain insulin are inadequately understood. Protein kinase C may also contribute to insulin resistance by phosphorylating the insulin receptor substrates required for insulin signaling. Insulin is also responsible insulin-long term depression, a type of synaptic plasticity that is also dependent on protein kinase C. However, insulin can also activate PKC signaling pathways via PLC gamma, Erk 1/2 MAP kinase, and src stimulation. Taken together, the available evidence suggests that the major impact of protein kinase C and its interaction with insulin in the mature, fully differentiated nervous system appears to be to induce synaptogenesis, enhance memory, reduce Alzheimer's pathophysiology, and stimulate neurorepair.


Assuntos
Insulina/fisiologia , Memória/fisiologia , Neurônios/fisiologia , Proteína Quinase C/fisiologia , Sinapses/fisiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Animais , Humanos , Fator de Crescimento Insulin-Like I/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Regeneração Nervosa , Proteína Oncogênica v-cbl/fisiologia , Receptor de Insulina/fisiologia , Transdução de Sinais , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/fisiopatologia
19.
Proc Natl Acad Sci U S A ; 104(49): 19571-6, 2007 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-18073185

RESUMO

Using both scanning confocal and electron microscopic morphometric measurements, we analyzed single dendritic spines of CA1 pyramidal cells in the hippocampi of water maze-trained rats vs. controls. Two days after completion of all training, we observed a memory-specific increase in the number of mushroom spines-all of which make synaptic contacts-but not in the numbers of filopodia or stubby or thin spines, as quantified with double-blind protocols in both scanning confocal and electron microscopic images. This memory-specific increase of mushroom spine number was enhanced by the PKC activator and candidate Alzheimer's disease therapeutic bryostatin, blocked by the PKCalpha-isozyme blocker Ro 31-8220, and accompanied by increases in the number of "perforated" postsynaptic densities, increased numbers of presynaptic vesicles, and the increased occurrence of double-synapse presynaptic boutons associated with the mushroom spines. These and other confocally imaged immunohistochemical results described here involving PKC substrates indicate that individual mushroom spines provide structural storage sites for long-term associative memory and sites for memory-specific synaptogenesis that involve PKC-regulated changes of spine shape, as well as PKC-regulated changes of pre- and postsynaptic ultrastructure.


Assuntos
Espinhas Dendríticas/enzimologia , Memória , Proteína Quinase C-alfa/metabolismo , Células Piramidais/enzimologia , Animais , Axônios/ultraestrutura , Briostatinas/farmacologia , Espinhas Dendríticas/ultraestrutura , Indóis/farmacologia , Aprendizagem em Labirinto , Proteína Quinase C-alfa/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Células Piramidais/ultraestrutura , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ratos , Ratos Endogâmicos , Vesículas Secretórias/enzimologia , Vesículas Secretórias/ultraestrutura
20.
J Neurosci ; 24(48): 10878-87, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15574738

RESUMO

Large increases in cytosolic free Ca2+ ([Ca2+]i) activate several kinases that are important for neuronal plasticity, including Ca2+/calmodulin-dependent kinase II (CaMKII), protein kinase A (PKA), and protein kinase C (PKC). Because it is also known, mainly in non-neuronal systems, that superoxide radicals (O2-) activate these (and other) kinases and because O2- generation by mitochondria is in part [Ca2+]i dependent, we examined in hippocampal neurons the relationship between Ca2+ entry, O2- production, and kinase activity. We found that, after large stimulus-induced [Ca2+]i increases, O2- selectively produced by mitochondria near plasmalemmal sites of Ca2+ entry acts as a modulator to upregulate the two kinases, namely, CaMKII and PKA, whose activities are directly or indirectly phosphorylation dependent. The common mechanism involves O2- inhibition of inactivating protein phosphatases. Conversely, because small [Ca2+]i increases do not promote mitochondrial respiration and O2- generation, weak stimuli favor enhanced phosphatase activity, which therefore leads to suppressed kinase activity. Enhanced O2- production also promoted PKC activity but by a phosphatase-independent pathway. These results suggest that Ca2+-dependent upregulation of mitochondrial O2- production may be a general mechanism for linking Ca2+ entry to enhanced kinase activity and therefore to synaptic plasticity. This mechanism also represents yet another way that mitochondria, acting as calcium sensors, can play a role in neuronal signal transduction.


Assuntos
Sinalização do Cálcio/fisiologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Cálcio/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ácido Egtázico/análogos & derivados , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Proteína Quinase C/metabolismo , Superóxidos/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Ácido Egtázico/farmacologia , Ativação Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Hipocampo/citologia , Hipocampo/metabolismo , Mitocôndrias/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ácido Okadáico/farmacologia , Oligomicinas/farmacologia , Fosfoproteínas Fosfatases/fisiologia , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/fisiologia , Rotenona/farmacologia , Superóxido Dismutase/genética , Superóxido Dismutase/fisiologia , Tetrodotoxina/farmacologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA