Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 15: 1323409, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38352874

RESUMO

Background: Alzheimer's disease (AD) is the most common neurodegenerative disorder affecting memory and cognition. The disease is accompanied by an abnormal deposition of ß-amyloid plaques in the brain that contributes to neurodegeneration and is known to induce glial inflammation. Studies in the APP/PS1 mouse model of ß-amyloid-induced neuropathology have suggested a role for inflammasome activation in ß-amyloid-induced neuroinflammation and neuropathology. Methods: Here, we evaluated the in vivo role of microglia-selective and full body inflammasome signalling in several mouse models of ß-amyloid-induced AD neuropathology. Results: Microglia-specific deletion of the inflammasome regulator A20 and inflammasome effector protease caspase-1 in the AppNL-G-F and APP/PS1 models failed to identify a prominent role for microglial inflammasome signalling in ß-amyloid-induced neuropathology. Moreover, global inflammasome inactivation through respectively full body deletion of caspases 1 and 11 in AppNL-G-F mice and Nlrp3 deletion in APP/PS1 mice also failed to modulate amyloid pathology and disease progression. In agreement, single-cell RNA sequencing did not reveal an important role for Nlrp3 signalling in driving microglial activation and the transition into disease-associated states, both during homeostasis and upon amyloid pathology. Conclusion: Collectively, these results question a generalizable role for inflammasome activation in preclinical amyloid-only models of neuroinflammation.


Assuntos
Doença de Alzheimer , Camundongos , Animais , Doença de Alzheimer/patologia , Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Peptídeos beta-Amiloides , Precursor de Proteína beta-Amiloide/genética , Doenças Neuroinflamatórias , Camundongos Transgênicos , Amiloide , Proteínas Amiloidogênicas
2.
J Neurosci Res ; 100(1): 203-219, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-32253777

RESUMO

A major challenge in medicine is developing potent pain therapies without the adverse effects of opiates. Neuroinflammation and in particular microglial activation have been shown to contribute to these effects. However, the implication of the microglial mu opioid receptor (MOR) is not known. We developed a novel conditional knockout (cKO) mouse line, wherein MOR is deleted in microglia. Morphine analgesic tolerance was delayed in both sexes in cKO mice in the hot plate assay. Opioid-induced hyperalgesia (OIH) as measured in the tail immersion assay was abolished in male cKO mice, and physical dependence to morphine as assessed by naloxone-induced withdrawal was attenuated in female cKO mice. Our results show a sex-dependent contribution of microglial MOR in morphine analgesic tolerance, OIH, and physical dependence. In conclusion, our data suggest that blockade of microglial MOR could represent a therapeutic target for opiate analgesia without the opiate adverse effects.


Assuntos
Morfina , Receptores Opioides mu , Analgésicos , Analgésicos Opioides/efeitos adversos , Animais , Feminino , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Masculino , Camundongos , Microglia , Morfina/efeitos adversos , Receptores Opioides mu/genética
3.
Front Cell Neurosci ; 15: 745178, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34602984

RESUMO

Background: The delta opioid receptor (DOR) contributes to pain control, and a major challenge is the identification of DOR populations that control pain, analgesia, and tolerance. Astrocytes are known as important cells in the pathophysiology of chronic pain, and many studies report an increased prevalence of pain in women. However, the implication of astrocytic DOR in neuropathic pain and analgesia, as well as the influence of sex in this receptor activity, remains unknown. Experimental Approach: We developed a novel conditional knockout (cKO) mouse line wherein DOR is deleted in astrocytes (named GFAP-DOR-KO), and investigated neuropathic mechanical allodynia as well as analgesia and analgesic tolerance in mutant male and female mice. Neuropathic cold allodynia was also characterized in mice of both sexes lacking DOR either in astrocytes or constitutively. Results: Neuropathic mechanical allodynia was similar in GFAP-DOR-KO and floxed DOR control mice, and the DOR agonist SNC80 produced analgesia in mutant mice of both sexes. Interestingly, analgesic tolerance developed in cKO males and was abolished in cKO females. Cold neuropathic allodynia was reduced in mice with decreased DOR in astrocytes. By contrast, cold allodynia was exacerbated in full DOR KO females. Conclusions: These findings show that astrocytic DOR has a prominent role in promoting cold allodynia and analgesic tolerance in females, while overall DOR activity was protective. Altogether this suggests that endogenous- and exogenous-mediated DOR activity in astrocytes worsens neuropathic allodynia while DOR activity in other cells attenuates this form of pain. In conclusion, our results show a sex-specific implication of astrocytic DOR in neuropathic pain and analgesic tolerance. These findings open new avenues for developing tailored DOR-mediated analgesic strategies.

4.
J Alzheimers Dis ; 77(4): 1397-1416, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32894244

RESUMO

BACKGROUND: As a consequence of the discovery of an extracellular component responsible for the progression of tau pathology, tau immunotherapy is being extensively explored in both preclinical and clinical studies as a disease modifying strategy for the treatment of Alzheimer's disease. OBJECTIVE: Describe the characteristics of the anti-phospho (T212/T217) tau selective antibody PT3 and its humanized variant hPT3. METHODS: By performing different immunization campaigns, a large collection of antibodies has been generated and prioritized. In depth, in vitro characterization using surface plasmon resonance, phospho-epitope mapping, and X-ray crystallography experiments were performed. Further characterization involved immunohistochemical staining on mouse- and human postmortem tissue and neutralization of tau seeding by immunodepletion assays. RESULTS AND CONCLUSION: Various in vitro experiments demonstrated a high intrinsic affinity for PT3 and hPT3 for AD brain-derived paired helical filaments but also to non-aggregated phospho (T212/T217) tau. Further functional analyses in cellular and in vivo models of tau seeding demonstrated almost complete depletion of tau seeds in an AD brain homogenate. Ongoing trials will provide the clinical evaluation of the tau spreading hypothesis in Alzheimer's disease.


Assuntos
Anticorpos Monoclonais Humanizados/metabolismo , Anticorpos Monoclonais/metabolismo , Descoberta de Drogas/métodos , Proteínas tau/metabolismo , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais Humanizados/química , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Estrutura Terciária de Proteína , Proteínas tau/química
5.
Neurobiol Dis ; 127: 398-409, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30878534

RESUMO

We have exploited whole brain microscopy to map the progressive deposition of hyperphosphorylated tau in intact, cleared mouse brain. We found that the three-dimensional spreading pattern of hyperphosphorylated tau in the brain of an aging Tau.P301L mouse model did not resemble that observed in AD patients. Injection of synthetic or patient-derived tau fibrils in the CA1 region resulted in a more faithful spreading pattern. Atlas-guided volumetric analysis showed a connectome-dependent spreading from the injection site and also revealed hyperphosphorylated tau deposits beyond the direct anatomical connections. In fibril-injected brains, we also detected a persistent subpopulation of rod-like and swollen microglia. Furthermore, we showed that the hyperphosphorylated tau load could be reduced by intracranial co-administration of, and to a lesser extent, by repeated systemic dosing with an antibody targeting the microtubule-binding domain of tau. Thus, the combination of targeted seeding and in toto staging of tau pathology allowed assessing regional vulnerability in a comprehensive manner, and holds potential as a preclinical drug validation tool.


Assuntos
Encéfalo/metabolismo , Microglia/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo , Envelhecimento/metabolismo , Envelhecimento/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Progressão da Doença , Camundongos , Camundongos Transgênicos , Microglia/patologia , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Neurônios/metabolismo , Neurônios/patologia , Fosforilação , Tauopatias/patologia
6.
Sci Rep ; 7(1): 10406, 2017 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-28871199

RESUMO

Opiates are potent analgesics but their clinical use is limited by side effects including analgesic tolerance and opioid-induced hyperalgesia (OIH). The Opiates produce analgesia and other adverse effects through activation of the mu opioid receptor (MOR) encoded by the Oprm1 gene. However, MOR and morphine metabolism involvement in OIH have been little explored. Hence, we examined MOR contribution to OIH by comparing morphine-induced hyperalgesia in wild type (WT) and MOR knockout (KO) mice. We found that repeated morphine administration led to analgesic tolerance and hyperalgesia in WT mice but not in MOR KO mice. The absence of OIH in MOR KO mice was found in both sexes, in two KO global mutant lines, and for mechanical, heat and cold pain modalities. In addition, the morphine metabolite morphine-3beta-D-glucuronide (M3G) elicited hyperalgesia in WT but not in MOR KO animals, as well as in both MOR flox and MOR-Nav1.8 sensory neuron conditional KO mice. M3G displayed significant binding to MOR and G-protein activation when using membranes from MOR-transfected cells or WT mice but not from MOR KO mice. Collectively our results show that MOR is involved in hyperalgesia induced by chronic morphine and its metabolite M3G.


Assuntos
Hiperalgesia/induzido quimicamente , Derivados da Morfina/efeitos adversos , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Animais , Modelos Animais de Doenças , Tolerância a Medicamentos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Inativação de Genes , Hiperalgesia/genética , Hiperalgesia/metabolismo , Masculino , Camundongos , Morfina/efeitos adversos , Morfina/farmacologia , Derivados da Morfina/farmacologia
7.
Oncotarget ; 7(26): 39118-39135, 2016 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-27259247

RESUMO

Stress has been implicated as a risk factor for the severity and progression of sporadic Alzheimer's disease (AD). Early life experiences determine stress responsivity in later life, and modulate age-dependent cognitive decline. Therefore, we examined whether early life experiences influence AD outcome in a bigenic mouse model which progressively develops combined tau and amyloid pathology (biAT mice).Mice were subjected to either early life stress (ELS) or to 'positive' early handling (EH) postnatally (from day 2 to 9). In biAT mice, ELS significantly compromised long term survival, in contrast to EH which increased life expectancy. In 4 month old mice, ELS-reared biAT mice displayed increased hippocampal Aß levels, while these levels were reduced in EH-reared biAT mice. No effects of ELS or EH were observed on the brain levels of APP, protein tau, or PSD-95. Dendritic morphology was moderately affected after ELS and EH in the amygdala and medial prefrontal cortex, while object recognition memory and open field performance were not affected. We conclude that despite the strong transgenic background, early life experiences significantly modulate the life expectancy of biAT mice. Parallel changes in hippocampal Aß levels were evident, without affecting cognition of young adult biAT mice.


Assuntos
Doença de Alzheimer/patologia , Amiloide/genética , Amiloide/metabolismo , Estresse Psicológico , Tonsila do Cerebelo/metabolismo , Animais , Comportamento Animal , Peso Corporal , Mapeamento Encefálico , Transtornos Cognitivos/genética , Transtornos Cognitivos/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Proteína 4 Homóloga a Disks-Large/genética , Feminino , Heterozigoto , Hipocampo/metabolismo , Homozigoto , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Fosforilação , Córtex Pré-Frontal/metabolismo , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento
8.
Brain Res ; 1606: 125-32, 2015 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-25708149

RESUMO

Myelin is essential for efficient signal transduction in the nervous system comprising of multiple proteins. The intricacies of the regulation of the formation of myelin, and its components, are not fully understood. Here, we describe the characterization of a novel myelin basic protein (Mbp) mutant mouse, mbp(jive), which spontaneously occurred in our mouse colony. These mice displayed the onset of a shaking gait before 3 weeks of age and seizure onset before 2 months of age. Due to a progressive increase of seizure intensity, mbp(jive) mice experienced premature lethality at around 3 months of age. Mbp mRNA transcript or protein was undetectable and, accordingly, genetic analysis demonstrated a homozygous loss of exons 3 to 6 of Mbp. Peripheral nerve conductance was mostly unimpaired. Additionally, we observed grave structural changes in white matter predominant structures were detected by T1, T2 and diffusion weighted magnetic resonance imaging. We additionally observed that Mbp-deficiency results in an upregulation of Qkl, Mag and Cnp, suggestive of a regulatory feedback mechanism whereby compensatory increases in Qkl have downstream effects on Mag and Cnp. Further research will clarify the role and specifications of this myelin feedback loop, as well as determine its potential role in therapeutic strategies for demyelinating disorders.


Assuntos
Camundongos Mutantes/genética , Camundongos Mutantes/fisiologia , Proteína Básica da Mielina/genética , Bainha de Mielina/genética , Ativação Transcricional , Animais , Encéfalo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/genética , Mutação , Fenótipo , Transdução de Sinais/genética , Regulação para Cima , Substância Branca/patologia
9.
Neurobiol Dis ; 67: 119-32, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24704314

RESUMO

Tau.P301L transgenic mice suffer precocious mortality between ages 8 and 11 months, resulting from upper airway defects caused by tauopathy in autonomic brainstem circuits that control breathing (Dutschmann et al., 2010). In individual mice, the clinical phenotype evolves progressively and rapidly (3-6weeks) from clasping, over general motor impairment to severe reduction in body-weight into the terminal phase that announces imminent death (<3days). Surprisingly, co-expression of GSK3ß with Tau.P301L significantly prolonged survival of bigenic biGT mice (Terwel et al., 2008), which we here assign to delayed development of brainstem tauopathy. Eventually, brainstem tauopathy became as prominent in old biGT mice in the specified brainstem nuclei as in the parental Tau.P301L mice, resulting in similar clinical deterioration and terminal phase preceding death, although at later age. Biochemically, in both genotypes the pathway to neurofibrillary tangles and neuropil threads was similar: phosphorylation of protein Tau and formation of soluble oligomers and insoluble aggregates, ending in the typical tangles and threads of tauopathy. The extra GSK3ß activity led to expected increased phosphorylation of protein Tau, particularly at residues S262 and S396, which we must conclude to delay the aggregation of protein Tau in the brainstem of aging biGT mice. The unexpected, paradoxical alleviation of the brainstem problems in biGT mice allowed them to grow older and thereby develop more severe tauopathy in forebrain than Tau.P301L mice, which succumb at younger age.


Assuntos
Tronco Encefálico/enzimologia , Quinase 3 da Glicogênio Sintase/metabolismo , Tauopatias/enzimologia , Proteínas tau/química , Proteínas tau/metabolismo , Animais , Encéfalo/enzimologia , Encéfalo/metabolismo , Tronco Encefálico/metabolismo , Feminino , Glicogênio Sintase Quinase 3 beta , Masculino , Camundongos , Camundongos Transgênicos , Fosforilação , Análise de Sobrevida , Tauopatias/metabolismo
10.
Eur J Neurosci ; 40(2): 2442-53, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24754737

RESUMO

The microtubule-associated protein Tau is responsible for a large group of neurodegenerative disorders, known as tauopathies, including Alzheimer's disease. Tauopathy result from augmented and/or aberrant phosphorylation of Tau. Besides aging and various genetic and epigenetic defects that remain largely unknown, an important non-genetic agent that contributes is hypothermia, eventually caused by anesthesia. Remarkably, tauopathy in brains of hibernating mammals is not pathogenic, and, because it is fully reversible, is even considered to be neuroprotective. Here, we assessed the terminal phase of Tau.P301L mice and bigenic crosses with mice lacking glycogen synthase kinase 3 (GSK3)α completely, or GSK3ß specifically in neurons. We also analysed biGT bigenic mice that co-express Tau.P301L with GSK3ß.S9A and develop severe forebrain tauopathy with age. We found that the precocious mortality of Tau.P301L mice was typified by hypothermia that aggravated Tau phosphorylation, but, surprisingly, independently of GSK3α/ß. The important contribution of hypothermia at the time of death of mice with tauopathy suggests that body temperature should be included as a parameter in the analysis of pre-clinical models, and, by extension, in patients suffering from tauopathy.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Processamento de Proteína Pós-Traducional , Tauopatias/metabolismo , Proteínas tau/metabolismo , Animais , Quinase 3 da Glicogênio Sintase/genética , Humanos , Hipotermia/metabolismo , Hipotermia/fisiopatologia , Camundongos , Neurônios/metabolismo , Fosforilação , Prosencéfalo/citologia , Prosencéfalo/metabolismo , Prosencéfalo/patologia , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Tauopatias/fisiopatologia , Proteínas tau/genética , Proteínas tau/toxicidade
11.
PLoS One ; 9(2): e87605, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24498342

RESUMO

The stratum lacunosum moleculare (SLM) is the connection hub between entorhinal cortex and hippocampus, two brain regions that are most vulnerable in Alzheimer's disease. We recently identified a specific synaptic deficit of Nectin-3 in transgenic models for tauopathy. Here we defined cognitive impairment and electrophysiological problems in the SLM of Tau.P301L mice, which corroborated the structural defects in synapses and dendritic spines. Reduced diffusion of DiI from the ERC to the hippocampus indicated defective myelinated axonal pathways. Ultrastructurally, myelinated axons in the temporoammonic pathway (TA) that connects ERC to CA1 were damaged in Tau.P301L mice at young age. Unexpectedly, the myelin defects were even more severe in bigenic biGT mice that co-express GSK3ß with Tau.P301L in neurons. Combined, our data demonstrate that neuronal expression of protein Tau profoundly affected the functional and structural organization of the entorhinal-hippocampal complex, in particular synapses and myelinated axons in the SLM. White matter pathology deserves further attention in patients suffering from tauopathy and Alzheimer's disease.


Assuntos
Axônios/metabolismo , Encéfalo/metabolismo , Fibras Nervosas Mielinizadas/metabolismo , Sinapses/metabolismo , Tauopatias/genética , Proteínas tau/genética , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Animais , Axônios/patologia , Axônios/ultraestrutura , Encéfalo/patologia , Encéfalo/fisiopatologia , Transtornos Cognitivos/genética , Transtornos Cognitivos/fisiopatologia , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Modelos Animais de Doenças , Córtex Entorrinal/metabolismo , Córtex Entorrinal/patologia , Córtex Entorrinal/fisiopatologia , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Atividade Motora/fisiologia , Mutação , Fibras Nervosas Mielinizadas/patologia , Fibras Nervosas Mielinizadas/ultraestrutura , Sinapses/patologia , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia , Tauopatias/metabolismo , Tauopatias/fisiopatologia , Proteínas tau/metabolismo
12.
PLoS One ; 8(12): e84442, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24376810

RESUMO

The microtubule associated protein tau causes primary and secondary tauopathies by unknown molecular mechanisms. Post-translational O-GlcNAc-ylation of brain proteins was demonstrated here to be beneficial for Tau.P301L mice by pharmacological inhibition of O-GlcNAc-ase. Chronic treatment of ageing Tau.P301L mice mitigated their loss in body-weight and improved their motor deficits, while the survival was 3-fold higher at the pre-fixed study endpoint at age 9.5 months. Moreover, O-GlcNAc-ase inhibition significantly improved the breathing parameters of Tau.P301L mice, which underpinned pharmacologically the close correlation of mortality and upper-airway defects. O-GlcNAc-ylation of brain proteins increased rapidly and stably by systemic inhibition of O-GlcNAc-ase. Conversely, biochemical evidence for protein Tau.P301L to become O-GlcNAc-ylated was not obtained, nor was its phosphorylation consistently or markedly affected. We conclude that increasing O-GlcNAc-ylation of brain proteins improved the clinical condition and prolonged the survival of ageing Tau.P301L mice, but not by direct biochemical action on protein tau. The pharmacological effect is proposed to be located downstream in the pathological cascade initiated by protein Tau.P301L, opening novel venues for our understanding, and eventually treating the neurodegeneration mediated by protein tau.


Assuntos
Encéfalo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Piranos/farmacologia , Mecânica Respiratória/fisiologia , Tauopatias/tratamento farmacológico , Tauopatias/fisiopatologia , Tiazóis/farmacologia , beta-N-Acetil-Hexosaminidases/antagonistas & inibidores , Análise de Variância , Animais , Western Blotting , Feminino , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Camundongos Transgênicos , Pletismografia , Piranos/síntese química , Mecânica Respiratória/efeitos dos fármacos , Tiazóis/síntese química , Proteínas tau/genética
13.
PLoS One ; 8(5): e63589, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23704923

RESUMO

Cell adhesion molecules are important structural substrates, required for synaptic plasticity and synaptogenesis. CAMs differ widely in their expression throughout different brain regions and their specific structural and functional roles in the brain remain to be elucidated. Here, we investigated selected cell adhesion molecules for alterations in expression levels and neuronal localization in validated mouse models for Alzheimer's disease that mimic the age-related progression of amyloid accumulation and tauopathy. Among the cell adhesion molecules analyzed, Nectin-3 expression was affected most and specifically in all mouse models with tauopathy. In particular was Nectin-3 depleted from the specific region of the hippocampus, known as the stratum lacunosum and moleculare, in mice that express wild-type or mutant human protein Tau, either chronically or sub-acutely. Tauopathy progresses from the entorhinal cortex to the hippocampus by unknown mechanisms that could involve transport by the myelinated axons of the temporoammonic and perforant pathways. The decreased expression of Nectin-3 in the stratum lacunosum moleculare is an early marker of impaired transport, and eventual synaptic problems, caused by beginning tauopathy.


Assuntos
Encéfalo/metabolismo , Encéfalo/patologia , Moléculas de Adesão Celular/metabolismo , Regulação para Baixo/genética , Tauopatias/metabolismo , Animais , Moléculas de Adesão Celular/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Nectinas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sinaptossomos/metabolismo , Tauopatias/patologia
14.
Mol Brain ; 6: 27, 2013 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-23705847

RESUMO

BACKGROUND: GSK3ß is involved in a wide range of physiological functions, and is presumed to act in the pathogenesis of neurological diseases, from bipolar disorder to Alzheimer's disease (AD). In contrast, the GSK3α isozyme remained largely ignored with respect to both aspects. RESULTS: We generated and characterized two mouse strains with neuron-specific or with total GSK3α deficiency. Behavioral and electrophysiological analysis demonstrated the physiological importance of neuronal GSK3α, with GSK3ß not compensating for impaired cognition and reduced LTP. Interestingly, the passive inhibitory avoidance task proved to modulate the phosphorylation status of both GSK3 isozymes in wild-type mice, further implying both to function in cognition. Moreover, GSK3α contributed to the neuronal architecture of the hippocampal CA1 sub-region that is most vulnerable in AD. Consequently, practically all parameters and characteristics indicated that both GSK3 isoforms were regulated independently, but that they acted on the same physiological functions in learning and memory, in mobility and in behavior. CONCLUSIONS: GSK3α proved to be regulated independently from GSK3ß, and to exert non-redundant physiological neurological functions in general behavior and in cognition. Moreover, GSK3α contributes to the pathological phosphorylation of protein Tau.


Assuntos
Cognição/fisiologia , Pleiotropia Genética , Quinase 3 da Glicogênio Sintase/deficiência , Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas tau/metabolismo , Animais , Comportamento Animal , Peso Corporal , Encéfalo/enzimologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Região CA1 Hipocampal/enzimologia , Região CA1 Hipocampal/patologia , Cruzamentos Genéticos , Feminino , Genótipo , Glicogênio Sintase Quinase 3 beta , Humanos , Infertilidade Masculina/enzimologia , Infertilidade Masculina/patologia , Integrases/metabolismo , Isoenzimas/metabolismo , Estimativa de Kaplan-Meier , Potenciação de Longa Duração , Masculino , Camundongos , Camundongos Knockout , Atividade Motora , Proteínas Mutantes/metabolismo , Neurônios/enzimologia , Neurônios/patologia , Tamanho do Órgão , Especificidade de Órgãos , Fenótipo , Fosforilação
15.
J Neurosci ; 31(49): 18036-47, 2011 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-22159117

RESUMO

Cognitive demise correlates with progressive brain tauopathy in dementing patients. Improved cognition of young Tau.P301L mice contrasts with dysfunction later in life and remains unexplained (Boekhoorn et al., 2006). To unravel early mechanisms, we composed a correlative time line of clinical symptoms, cognitive defects, and biochemical and pathological traits, including comprehensive analysis of dendritic spines in specified regions of the cortex and hippocampus of young and adult Tau.P301L mice. Remarkably, young Tau.P301L mice have not more, but more mature spines than wild-type mice, revealing the anatomical substrate for their improved cognition and LTP. Spine maturation remained high in the hippocampus of adult Tau.P301L mice. However, spines regressed in length paralleling impaired cognition and increased Tau phosphorylation (Terwel et al., 2005). Conversely, spine maturation was unaffected in adult Tau.4R mice, while spine density was increased and length reduced similar to Tau.P301L mice. To explain how protein Tau promoted spinogenesis, we analyzed hippocampal synaptosomes and dendritic spines for mouse and human Tau. While synaptosomes were positive for both mouse and human Tau, weak variable reaction in spines was observed only after fixation according to Bouin. Mouse Tau was absent from spines in wild-type mice, dissociating the pathological actions of Tau in transgenic mice by relocalization into dendrites and spines from the physiological actions of protein Tau in axons only. We conclude that mutant protein Tau modulates cognition and morphology of spines similarly and in both directions, with pathology later in life coinciding with increased phosphorylation and relocalization of Tau from axons to soma and processes.


Assuntos
Encéfalo/patologia , Transtornos Cognitivos , Espinhas Dendríticas/patologia , Leucina/genética , Neurônios/patologia , Prolina/genética , Proteínas tau/genética , Fatores Etários , Animais , Proteínas de Bactérias/genética , Encéfalo/citologia , Células Cultivadas , Transtornos Cognitivos/genética , Transtornos Cognitivos/patologia , Transtornos Cognitivos/fisiopatologia , Modelos Animais de Doenças , Feminino , Humanos , Proteínas Luminescentes/genética , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Fosforilação/genética , Sinaptossomos/metabolismo , Proteínas tau/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...